Skip to main content

Cangrelor ameliorates CLP-induced pulmonary injury in sepsis by inhibiting GPR17

Abstract

Background

Sepsis is a common complication of severe wound injury and infection, with a very high mortality rate. The P2Y12 receptor inhibitor, cangrelor, is an antagonist anti-platelet drug.

Methods

In our study, we investigated the protective mechanisms of cangrelor in CLP-induced pulmonary injury in sepsis, using C57BL/6 mouse models.

Results

TdT-mediated dUTP Nick-End Labeling (TUNEL) and Masson staining showed that apoptosis and fibrosis in lungs were alleviated by cangrelor treatment. Cangrelor significantly promoted surface expression of CD40L on platelets and inhibited CLP-induced neutrophils in Bronchoalveolar lavage fluid (BALF) (pā€‰<ā€‰0.001). We also found that cangrelor decreased the inflammatory response in the CLP mouse model and inhibited the expression of inflammatory cytokines, IL-1Ī² (pā€‰ā€‰<ā€‰0.01), IL-6 (pā€‰<ā€‰0.05), and TNF-Ī± (pā€‰<ā€‰0.001). Western blotting and RT-PCR showed that cangrelor inhibited the increased levels of G-protein-coupled receptor 17 (GPR17) induced by CLP (pā€‰<ā€‰0.001).

Conclusion

Our study indicated that cangrelor repressed the levels of GPR17, followed by a decrease in the inflammatory response and a rise of neutrophils in BALF, potentially reversing CLP-mediated pulmonary injury during sepsis.

Background

Sepsis is a serious disease and will lead a high mortality rate of approximately 22% in all over the world [1]. It has several features like out of controlled inflammatory response, coagulation disorder, and immune dysfunctions [2]. The cause of sepsis is complex, and its main cause is a response caused by the hostā€™s immune response to the pathogen [2, 3]. Overall, the physiological and pathological mechanisms of sepsis are well researched. A lot of mediators such as cytokines, chemokines, and free radicals participate in the development of sepsis [4]. The lung is one of the initial target organ of the systemic inflammatory response caused by sepsis, leading to alveolar or capillary epithelial cell injury, diffuse pulmonary interstitial edema, and inflammatory cell exudation (i.e., acute lung injury/respiratory distress syndrome, progressive respiratory distress, and hypoxemia) and then acute respiratory failure and non-cardiogenic pulmonary edema [5,6,7,8]. Despite numerous treatment strategies, drugs that effectively improve the treatment of sepsis remain to be developed. Antibiotics are the key method for clinical treatment, but the death rate of sepsis still reaches an astonishing number of 30ā€“40% [9].

In the past few years, the treatment of sepsis-related coagulopathy has been widely investigated. Recent studies have found that there is a coagulationā€“inflammation cross-reaction system with plateletā€“leukocyte aggregates as the core pathogenesis of sepsis [10]. With inhibition of platelet activation by anti-platelet drugs being used to treat sepsis/acute lung injury, the high risk of patients could be reduced organ damage and alleviated conditions [11, 12]. Former studies have found that P2Y12 receptor antagonists such as clopidogrel and ticagrelor treat sepsis and lung damage through anti-platelet aggregation effects [13,14,15]. The P2Y12 receptor belongs to a protein-coupled receptor and is one of two receptors on platelets. The P2Y12 receptor is also the initiation channel for platelet activation, and the receptor strongly amplifies signal activation. This amplification is displayed not only in the magnification of the platelet aggregation signal, as well as in subsequent multi-step signaling, including the release of platelet particles and the enhancement of platelet-promoting activity [16, 17]. P2Y12 receptor antagonists prevent the formation of neutrophilā€“platelet aggregates by inhibiting the recruitment of neutrophils, significantly reducing thrombocytopenia, and also reducing lung damage caused by sepsis [18,19,20]. However, the therapeutic mechanism of cangrelor on sepsis has not been reported. Our research aims were to characterize the potential effect of cangrelor on sepsis and to identify its mechanism of action.

Materials and methods

Drugs and reagents

Cangrelor (98% pure) was obtained from Vcare PharmaTech Co., Ltd (Shanghai, China). Myeloperoxidase (MPO) kit was obtained from Nanjing Jiancheng (Jiangsu, China). BCA Protein Assay was obtained from Thermo Fisher (MA, USA). Anti-CD40L (#15094, 1:1000) and Ī²-actin (#4970, 1:1000) were obtained from Cell Signalling Technology (MA, USA). Anti-GPR17 (ab75553, 1:500) was obtained from Abcam (Shanghai, China). Enzyme-linked immunosorbent assay (ELISA) kits were obtained from RapidBio (CA, USA). TRIzol reagent was obtained from Life Technology (CA, USA). PrimeScriptĀ® RT Master Mix and SYBRĀ® Premix Ex Taqā„¢ were purchased from Takara (Dalian, China). All other reagents were obtained from Sigma-Aldrich (Shanghai, China).

Animal treatment

C57BL/6 mice (20ā€‰Ā±ā€‰2Ā g) were purchased from Shanghai Laboratory Animal Center (Shanghai, China) and fed according to guidelines approved by the Experimental Animal Ethical Committee of Pudong New Area Gongli Hospital. They were raised in a constant temperature and humidity room (22ā€‰Ā±ā€‰1Ā Ā°C, 65ā€‰Ā±ā€‰5% humidity) with standard diet and water in Pudong New Area Gongli Hospital. For the survival study, 62 mice were randomly separated into 4 groups: (1) sham (nā€‰=ā€‰8), (2) cecal ligation and double puncture (CLP) (nā€‰=ā€‰18), (3) CLPā€‰+ā€‰cangrelor (5Ā mg/kg) (nā€‰=ā€‰18), and (4) CLPā€‰+ā€‰cangrelor (20Ā mg/kg) (nā€‰=ā€‰18). Another 32 mice were randomly separated into 4 groups: (1) sham (nā€‰=ā€‰8), (2) cecal ligation and double puncture (CLP) (nā€‰=ā€‰8), (3) CLPā€‰+ā€‰cangrelor (5Ā mg/kg) (nā€‰=ā€‰8), and (4) CLPā€‰+ā€‰cangrelor (20Ā mg/kg) (nā€‰=ā€‰8). The CLP was performed on isoflurane-anesthetized mice as previously described [21, 22]. Sham control animals underwent a laparotomy without ligation or double puncture. Mice were orally administrated with cangrelor (5 or 20Ā mg/kg) 2Ā h before surgery. After surgery but prior to emergence, mice were fluid-resuscitated (1Ā mL/mouse of sterile saline, subcutaneously). For the mechanism study, 32 mice were randomly separated into four groups: (1) sham (nā€‰=ā€‰8), (2) cecal ligation and double puncture (CLP) (nā€‰=ā€‰8), (3) CLPā€‰+ā€‰cangrelor (5Ā mg/kg) (nā€‰=ā€‰8), and (4) CLPā€‰+ā€‰cangrelor (20Ā mg/kg) (nā€‰=ā€‰8). Mice were killed 24Ā h after induction of CLP, plasma and lung were saved.

Masson staining of the lung

Paraffin sections of lung tissues were analyzed after several methods containing dewaxing rehydration, Ponceau S Fuchs in acid solution staining for 300Ā s, by washing in 1% phosphomolybdic acid differentiation solution for 5Ā min, aniline blue solution counterstaining for 300Ā s, with 1% glacial acetic acid for 1Ā min, alcohol gradient dehydration, transparency production using dimethylbenzene and xylene, and mounting.

Flow cytometry

For detection of surface CD40L expression on platelets, plasma was placed for 10Ā min at 36.5Ā Ā°C that blocked FcĪ³III/II receptors to decrease nonspecific labeling, next incubated with fluorescein isothiocyanate-conjugated anti-CD41. Immobilization of cells with 1% formaldehyde; erythrocytes were lysed with fluorescence-activated cell sorting lysing solution (BD Biosciences, Shanghai, China), and neutrophils and platelets were then recovered by centrifugation at 300Ɨg for 10Ā min. Platelet activation was analyzed using PEā€conjugated JON/A antibody (Emfret Analytics, WĆ¼rzburg, Germany), which bound to the high affinity conformation of mouse Ī±ffini.

RNA isolation and quantitative real-time PCR (qRT-PCR)

mRNA was obtained using TRIzol, respectively. cDNA was synthesized using M-MLV reverse transcriptase and Sangon reverse transcription primers. Relative transcript abundance was determined by the 2āˆ’Ī”Ī”Ct method and mRNA expression was normalized against Ī²-actin. All amplification analyses were completed on QuantStudioā„¢ 6 Flex Real-Time PCR System.

Protein extraction and western blot analysis

Lung tissues were lysed by RIPA buffer and protein concentrations were assessed by the BCA protein assay. About 20Ā Ī¼g of protein from each example was divided by a 10% SDSā€“polyacrylamide gel and transferred to polyvinylidene fluoride membranes. Membranes were blocked with 10% skim milk in Tris-Buffered Saline with Tween-20 (TBST) and incubated with primary antibodies overnight at 4Ā Ā°C. Membranes were incubated with the corresponding secondary antibody for 1Ā h at room temperature and washed in TBST. Protein signals were detected using the Super ECL Plus Detection Reagent.

Enzyme-linked immunosorbent assay (ELISA)

Concentrations of the inflammatory cytokines in mice serum were measured with an ELISA kit following to manufacture protocols.

Statistical analysis

The effects of Cangrelor on the survival of mice were analyzed using the Kaplanā€“Meier curve and log-rank test. All other results were analyzed using one-way ANOVA analysis by SPSS statistical software for Windows, version 22.0 (SPSS, Chicago, IL, USA). The measurement data are expressed as the meanā€‰Ā±ā€‰SD. A value of pā€‰<ā€‰0.05 was deemed statistically significant.

Results

Cangrelor alleviates CLP-induced pulmonary injury

First, as shown in Fig.Ā 1, the survival rate was 72.22% on the first 24Ā h, decreased to 55.56% on the next 24Ā h, and decreased a stable level of 38.89% on 72Ā h after CLP (pā€‰<ā€‰0.001). Treating with 5Ā mg/kg cangrelor did not enhance the survival rate compared with CLP group, while treating with 20Ā mg/kg cangrelor significantly improved the survival rate compared with CLP group (pā€‰<ā€‰0.001). Next, HE staining showed noticeable histopathologic changes including congestion, inflammatory cell infiltration, necrosis and degeneration while treating with 20Ā mg/kg cangrelor significantly ameliorate the histopathologic changes induced by CLP (Fig.Ā 2A). In addition, we assessed the levels of apoptosis in mice lung tissues, using a TUNEL assay. The results showed that in the CLP group, lung tissues had a higher level of apoptotic cells, but cangrelor (20Ā mg/kg) reversed this result (Fig.Ā 2B). Next, we assessed the anti-inflammatory effect of cangrelor. Masson staining showed that CLP mice had fibrotic changes, while there was significantly less damage in the CLPā€‰+ā€‰cangrelor (20Ā mg/kg) group, as showed by blue staining (Fig.Ā 2C).

Fig. 1
figure 1

Cangrelor improved the survival rate of sepsis mice. nā€‰=ā€‰8 for control group and nā€‰=ā€‰18 for all other groups

Fig. 2
figure 2

Cangrelor alleviates CLP-induced pulmonary injury. A Representative images of HE of mouse lung sections. B Representative images of TUNEL staining of mouse lung sections. C Representative images of Masson staining of mouse lung sections

Cangrelor regulates platelet aggregation and neutrophil recruitment.

The platelets surface level of CD40L was measured (Fig.Ā 3A). The mean fluorescence intensity (MFI) was 72 in the sham group and 41 in the CLP group (pā€‰<ā€‰0.01). After the administration of cangrelor, the MFI was 46 when treated with 5Ā mg/kg cangrelor and 91 when treated with 20Ā mg/kg cangrelor which was significantly increased compared with CLP group (pā€‰<ā€‰0.001). Western blotting also presented that the level of platelet CD40L was meaningfully decreased in the CLP group in contrast to the sham group, while cangrelor (20Ā mg/kg) increased this reduction (Fig.Ā 3B). A cell assay of the broncho-alveolar lavage fluid (BALF) indicated the neutrophils in the broncho-alveolar was 0.26ā€‰Ć—ā€‰106 in the sham mice while it was 1.76ā€‰Ć—ā€‰106 when induced by CLP. After cangrelor treatment, the number of neutrophils decreased to 0.87ā€‰Ć—ā€‰106 (pā€‰<ā€‰0.001) (Fig.Ā 3C). In addition, the pulmonary wet/dry ratio in the CLP group increased, which indicated the formation of pulmonary edema (pā€‰<ā€‰0.001). However, treatment with 20Ā mg/kg cangrelor significantly reversed this edema compared with CLP group (pā€‰<ā€‰0.01) (Fig.Ā 3D).

Fig. 3
figure 3

Cangrelor regulates platelet aggregation and neutrophil recruitment. A Platelet surface expression CD40L. B Western blots of platelet lysates CD40L. C The number of neutrophils in BALF. D The levels of edema formation in the lung. Results are presented as meansā€‰Ā±ā€‰SD (nā€‰=ā€‰8). **pā€‰<ā€‰0.01, *** pā€‰<ā€‰0.001 compared to the sham control; ##Pā€‰<ā€‰0.01, ###Pā€‰<ā€‰0.001 compared to CLP

Cangrelor ameliorates the inflammatory response through GPR17

CLP-induced levels of MPO in mice lung tissues were significantly increased (pā€‰<ā€‰0.01), whereas they decreased significantly in the presence of cangrelor (pā€‰<ā€‰0.01) (Fig.Ā 4A). Using an ELISA, the concentrations of TNF-Ī±, IL-1Ī², and IL-6 in mice serum were meaningfully raised in the presence of CLP (all pā€‰<ā€‰0.001), however, these levels were reduced by cangrelor (20Ā mg/kg) (IL-1Ī²: pā€‰<ā€‰0.01, TNF-Ī±: pā€‰<ā€‰0.05, IL-6: pā€‰<ā€‰0.001) (Fig.Ā 4B). Former research has reported that cangrelor is the antagonist of GPR17 [23]. As shown in Fig.Ā 4C, D western blotting and RT-PCR results indicated CLP upregulated GPR17 level in lung tissues(pā€‰<ā€‰0.001), whereas the administration of cangrelor (20Ā mg/kg) decreased its expression(pā€‰<ā€‰0.001).

Fig. 4
figure 4

Cangrelor ameliorates the inflammatory response through GPR17. A MPO concentration in lung tissue. B Serum concentrations of TNF-Ī±, IL-1Ī², and IL-6 (C) mRNA level of GPR17. D Western blot of GPR17 in lung tissue. Results are presented as meansā€‰Ā±ā€‰SD (nā€‰=ā€‰8). **pā€‰<ā€‰0.01, ***pā€‰<ā€‰0.001 compared to the sham control; ##pā€‰<ā€‰0.01, ###pā€‰<ā€‰0.001 compared to CLP

Discussion

In our research, we demonstrated that cangrelor alleviated CLP-induced lung injury in abdominal sepsis by regulating platelet aggregation and the inflammatory response.

Previous studies reported that P2Y12 receptor antagonists such as clopidogrel and ticagrelor alleviate sepsis and lung damage through anti-platelet aggregation [13,14,15]. In addition, increasing evidence has shown that P2Y12 receptor antagonists decrease the inflammatory response in cardiovascular events, thrombosis, and atherosclerosis [18, 24], which indicates that P2Y12 regulation can affect inflammation through mechanisms that have not yet been fully clarified. Cangrelor is a second-generation P2Y12 antagonist and past studies have found that cangrelor alleviates the inflammatory response in lung fibrosis [25], chronic inflammatory pain [26], and neuropathic pain [27]. In the present study, we found that pre-treatment of cangrelor decreased CLP-induced pulmonary injury by modulating the inflammatory response and significantly increased survival in sepsis mice. However, the diagnosis and treatment of sepsis usually occur after the onset of the disease. Hence, we will further investigate the time responses of cangrelor and evaluated the therapeutic effect of cangrelor at different time after CLP. Besides, lipopolysaccharide (LPS)-stimulated lung epithelial/endothelial cells are commonly used to mimic in vitro sepsis-induced lung injury model [28, 29]. What is more, Amison et al. [30] found that pulmonary neutrophil recruitment induced by intranasal LPS administration was inhibited in mice administered either with P2Y receptor antagonists. Hence, further studies are required to establish the effectiveness of treatment with the P2Y12 receptor antagonists, cangrelor in LPS-induced in vitro model.

Previous studies have also reported the release of soluble CD40L from platelets in sepsis-regulated platelet-dependent neutrophil lung accumulation [31, 32]. However, the mechanism for modulating CD40L platelet shedding in sepsis is not clear. In our study, CLP induced a decrease of surface CD40L, which was significantly increased after treatment with cangrelor. In addition, the increase of neutrophils in BALF was attenuated by cangrelor treatment. The increased of lymphocytes, macrophages and eosinophils in BALF also indicates lung damage. The immune regulation of macrophages is also considered to be an important target for sepsis [33]. Further research should be undertaken to investigate the effect of cangrelor on macrophage-specific therapeutic targeting their immunometabolism during sepsis and regulation of macrophage.

Cangrelor has been confirmed to effectively inhibit the expression of GPR17 [23, 34, 35]. There is increasing evidence that the modulation of GPR17 could ameliorate inflammatory responses and injury, which suggested that GPR17 plays an important role in injury reparation [36]. For example, inhibiting GPR17 repressed the activation of microglia and alleviated inflammation induced by ischemic in rats [34, 37]. Inhibition of GPR17 also protects against myocardial fibrosis caused by cardiac ischemia [38], and we further confirmed that the administration of cangrelor reversed the rise of GPR17 induced by CLP, as well as ameliorating the inflammatory response. However, the relationship between P2Y12 receptor and GPR17 is still unknown. Besides, whether cangrelor interacts with GPR17 directly or through other unknown pathways remains unclear. In future research, we will investigate drug binding site of cangrelor on GPR17 via molecular docking or surface plasmon resonance analysis.

Conclusion

Our study suggested that cangrelor inhibited the expression of GPR17 in lung tissue, thus attuned the inflammatory response and the recruitment of neutrophils in BALF, and ameliorated CLP-mediated pulmonary injury during sepsis. In summary, our findings revealed that cangrelor could ameliorate lung injury and increased survival rate of mice with CLP-induced sepsis. These results suggest that cangrelor could be a potential target treatment for sepsis-induced lung injury.

Availability of data and materials

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

Abbreviations

BALF:

Bronchoalveolar lavage fluid

CLP:

Cecal ligation and puncture

GPR17:

G-protein-coupled receptor 17

MPO:

Myeloperoxidase

MFI:

Mean fluorescence intensity

TUNEL:

TdT-mediated dUTP nick-end labeling

HE:

Staining hematoxylinā€“eosin staining

BCA:

Butyleyanoacrylate

RIPA:

Radio immunoprecipitation assay

qRT-PCR:

Quantitative reverse transcription polymerase chain reaction

TBST:

Tris-buffered saline with tween-20

ELISA:

Enzyme-linked immunosorbent assay

LPS:

Lipopolysaccharide

References

  1. Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al. Global, regional, and national sepsis incidence and mortality, 1990ā€“2017: analysis for the global burden of disease study. Lancet. 2020;395:200ā€“11.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  2. Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, Angus DC, Rubenfeld GD, Singer M, Sepsis Definitions Task F. Developing a new definition and assessing new clinical criteria for septic shock: for the third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315:775ā€“87.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  3. Zhou H, Deng M, Liu Y, Yang C, Hoffman R, Zhou J, Loughran PA, Scott MJ, Neal MD, Billiar TR. Platelet HMGB1 is required for efficient bacterial clearance in intra-abdominal bacterial sepsis in mice. Blood Adv. 2018;2:638ā€“48.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  4. Cinel I, Opal SM. Molecular biology of inflammation and sepsis: a primer. Crit Care Med. 2009;37:291ā€“304.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  5. Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ, Huffnagle GB. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1:16113.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  6. National Heart L, Blood Institute ACTN, Truwit JD, Bernard GR, Steingrub J, Matthay MA, Liu KD, Albertson TE, Brower RG, Shanholtz C, et al. Rosuvastatin for sepsis-associated acute respiratory distress syndrome. N Engl J Med. 2014;370:2191ā€“200.

    ArticleĀ  Google ScholarĀ 

  7. Force ADT, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307:2526ā€“33.

    Google ScholarĀ 

  8. Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, Herridge M, Randolph AG, Calfee CS. Acute respiratory distress syndrome. Nat Rev Dis Primers. 2019;5:18.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  9. Zhou J, Qian C, Zhao M, Yu X, Kang Y, Ma X, Ai Y, Xu Y, Liu D, An Y, et al. Epidemiology and outcome of severe sepsis and septic shock in intensive care units in mainland China. PLoS ONE. 2014;9: e107181.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  10. Cerletti C, Tamburrelli C, Izzi B, Gianfagna F, de Gaetano G. Platelet-leukocyte interactions in thrombosis. Thromb Res. 2012;129:263ā€“6.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  11. Losche W, Boettel J, Kabisch B, Winning J, Claus RA, Bauer M. Do aspirin and other antiplatelet drugs reduce the mortality in critically ill patients? Thrombosis. 2012;2012:720254.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  12. Winning J, Reichel J, Eisenhut Y, Hamacher J, Kohl M, Deigner HP, Claus RA, Bauer M, Losche W. Anti-platelet drugs and outcome in severe infection: clinical impact and underlying mechanisms. Platelets. 2009;20:50ā€“7.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  13. Rahman M, Gustafsson D, Wang Y, Thorlacius H, Braun OO. Ticagrelor reduces neutrophil recruitment and lung damage in abdominal sepsis. Platelets. 2014;25:257ā€“63.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  14. Liverani E, Rico MC, Tsygankov AY, Kilpatrick LE, Kunapuli SP. P2Y12 receptor modulates sepsis-induced inflammation. Arterioscler Thromb Vasc Biol. 2016;36:961ā€“71.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  15. Seidel M, Winning J, Claus RA, Bauer M, Losche W. Beneficial effect of clopidogrel in a mouse model of polymicrobial sepsis. J Thromb Haemost. 2009;7:1030ā€“2.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  16. Gachet C. ADP receptors of platelets and their inhibition. Thromb Haemost. 2001;86:222ā€“32.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  17. Kunapuli SP, Dorsam RT, Kim S, Quinton TM. Platelet purinergic receptors. Curr Opin Pharmacol. 2003;3:175ā€“80.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  18. Nylander S, Schulz R. Effects of P2Y12 receptor antagonists beyond platelet inhibitionā€“comparison of ticagrelor with thienopyridines. Br J Pharmacol. 2016;173:1163ā€“78.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  19. Wu L, Zhao F, Dai M, Li H, Chen C, Nie J, Wang P, Wang DW. P2y12 receptor promotes pressure overload-induced cardiac remodeling via platelet-driven inflammation in mice. Hypertension. 2017;70:759ā€“69.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  20. Mansour A, Bachelot-Loza C, Nesseler N, Gaussem P, Gouin-Thibault I. P2Y12 inhibition beyond thrombosis: effects on inflammation. Int J Mol Sci. 2020;21:1391.

    ArticleĀ  CASĀ  PubMed CentralĀ  Google ScholarĀ 

  21. Kahner BN, Shankar H, Murugappan S, Prasad GL, Kunapuli SP. Nucleotide receptor signaling in platelets. J Thromb Haemost. 2006;4:2317ā€“26.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  22. Quinton TM, Murugappan S, Kim S, Jin J, Kunapuli SP. Different G protein-coupled signaling pathways are involved in alpha granule release from human platelets. J Thromb Haemost. 2004;2:978ā€“84.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  23. Marucci G, Dal Ben D, Lambertucci C, Santinelli C, Spinaci A, Thomas A, Volpini R, Buccioni M. The G protein-coupled receptor GPR17: overview and update. Chem Med Chem. 2016;11:2567ā€“74.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  24. Liverani E, Kilpatrick LE, Tsygankov AY, Kunapuli SP. The role of P2Y (1)(2) receptor and activated platelets during inflammation. Curr Drug Targ. 2014;15:720ā€“8.

    ArticleĀ  CASĀ  Google ScholarĀ 

  25. Zhan TW, Tian YX, Wang Q, Wu ZX, Zhang WP, Lu YB, Wu M. Cangrelor alleviates pulmonary fibrosis by inhibiting GPR17-mediated inflammation in mice. Int Immunopharmacol. 2018;62:261ā€“9.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  26. Beko K, Kovanyi B, Goloncser F, Horvath G, Denes A, Kornyei Z, Botz B, Helyes Z, Muller CE, Sperlagh B. Contribution of platelet P2Y12 receptors to chronic complete Freundā€™s adjuvant-induced inflammatory pain. J Thromb Haemost. 2017;15:1223ā€“35.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  27. Horvath G, Goloncser F, Csolle C, Kiraly K, Ando RD, Baranyi M, Kovanyi B, Mate Z, Hoffmann K, Algaier I, et al. Central P2Y12 receptor blockade alleviates inflammatory and neuropathic pain and cytokine production in rodents. Neurobiol Dis. 2014;70:162ā€“78.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  28. Xia W, Zhang H, Pan Z, Li G, Zhou Q, Hu D, Liu Y. Inhibition of MRP4 alleviates sepsis-induced acute lung injury in rats. Int Immunopharmacol. 2019;72:211ā€“7.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  29. Chen Q, Liu J, Wang W, Liu S, Yang X, Chen M, Cheng L, Lu J, Guo T, Huang F. Sini decoction ameliorates sepsis-induced acute lung injury via regulating ACE2-Ang (1ā€“7)-mas axis and inhibiting the MAPK signaling pathway. Biomed Pharmacother. 2019;115:108971.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  30. Amison RT, Arnold S, Oā€™Shaughnessy BG, Cleary SJ, Ofoedu J, Idzko M, Page CP, Pitchford SC. Lipopolysaccharide (LPS) induced pulmonary neutrophil recruitment and platelet activation is mediated via the P2Y1 and P2Y14 receptors in mice. Pulm Pharmacol Ther. 2017;45:62ā€“8.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  31. Rahman M, Zhang S, Chew M, Ersson A, Jeppsson B, Thorlacius H. Platelet-derived CD40L (CD154) mediates neutrophil upregulation of Mac-1 and recruitment in septic lung injury. Ann Surg. 2009;250:783ā€“90.

    ArticleĀ  PubMedĀ  Google ScholarĀ 

  32. Rahman M, Zhang S, Chew M, Syk I, Jeppsson B, Thorlacius H. Platelet shedding of CD40L is regulated by matrix metalloproteinase-9 in abdominal sepsis. J Thromb Haemost. 2013;11:1385ā€“98.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  33. Kumar V. Targeting macrophage immunometabolism: Dawn in the darkness of sepsis. Int Immunopharmacol. 2018;58:173ā€“85.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  34. Ciana P, Fumagalli M, Trincavelli ML, Verderio C, Rosa P, Lecca D, Ferrario S, Parravicini C, Capra V, Gelosa P, et al. The orphan receptor GPR17 identified as a new dual uracil nucleotides/cysteinyl-leukotrienes receptor. EMBO J. 2006;25:4615ā€“27.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  35. Lecca D, Trincavelli ML, Gelosa P, Sironi L, Ciana P, Fumagalli M, Villa G, Verderio C, Grumelli C, Guerrini U, et al. The recently identified P2Y-like receptor GPR17 is a sensor of brain damage and a new target for brain repair. PLoS ONE. 2008;3:e3579.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  36. Matsui H, Sopko NA, Hannan JL, Reinhardt AA, Kates M, Yoshida T, Liu X, Castiglione F, Hedlund P, Weyne E, et al. M1 macrophages are predominantly recruited to the major pelvic ganglion of the rat following cavernous nerve injury. J Sex Med. 2017;14:187ā€“95.

    ArticleĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

  37. Zhao B, Zhao CZ, Zhang XY, Huang XQ, Shi WZ, Fang SH, Lu YB, Zhang WP, Xia Q, Wei EQ. The new P2Y-like receptor G protein-coupled receptor 17 mediates acute neuronal injury and late microgliosis after focal cerebral ischemia in rats. Neuroscience. 2012;202:42ā€“57.

    ArticleĀ  CASĀ  PubMedĀ  Google ScholarĀ 

  38. Cosentino S, Castiglioni L, Colazzo F, Nobili E, Tremoli E, Rosa P, Abbracchio MP, Sironi L, Pesce M. Expression of dual nucleotides/cysteinyl-leukotrienes receptor GPR17 in early trafficking of cardiac stromal cells after myocardial infarction. J Cell Mol Med. 2014;18:1785ā€“96.

    ArticleĀ  CASĀ  PubMedĀ  PubMed CentralĀ  Google ScholarĀ 

Download references

Acknowledgements

Not applicable.

Funding

The present study was funded by Shanghai Pudong New Area Health System Excellent Youth Medical Personnel Training Program (No. PWRq2017-09), Shanghai Pudong New Area Health System Key Specialty construction Project (No. PWZzk2017-05) and Shanghai Municipal Health and Family Planning Commission Research Project (No. 201640405).

Author information

Authors and Affiliations

Authors

Contributions

QK, QJ and DG designed study, edited and corrected the manuscript. QL and RL performed the experiments and drafted the article. GL, MH, GC and LX assisted perform the study and analyzed data. All the authors read and approved the final manuscript.

Corresponding authors

Correspondence to Dongfeng Guo or Qi Kang.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Experimental Animal Ethical Committee of Pudong New Area Gongli Hospital.

Consent for publication

Written informed consent for publication was collected from each participant.

Competing interests

All the authors declare they have no competing interests.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luo, Q., Liu, R., Qu, K. et al. Cangrelor ameliorates CLP-induced pulmonary injury in sepsis by inhibiting GPR17. Eur J Med Res 26, 70 (2021). https://doi.org/10.1186/s40001-021-00536-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-021-00536-4

Keywords