P2X7 purinergic receptor: A potential target in heart diseases (Review)

  • Authors:
    • Anfal F. Bin Dayel
    • Asma S. Alonazi
    • Tahani K. Alshammari
    • Nouf M. Alrasheed
  • View Affiliations

  • Published online on: February 15, 2023     https://doi.org/10.3892/mmr.2023.12961
  • Article Number: 74
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The P2X7 purinergic receptor (P2X7R) is a non‑selective cation channel activated by high levels of adenosine triphosphate that are commonly present in serious conditions. Activation of this purinergic receptor is closely related to the development of various disease states including inflammatory and neurodegenerative disorders, orthopedic diseases and types of cancer. Accumulating evidence has shown that the P2X7R plays a crucial role in the development of various heart diseases. For example, activation of P2X7Rs may alleviate myocardial ischemia‑reperfusion injury by releasing endogenous cardiac protective substances. In contrast to these findings, activation of P2X7Rs can promote the development of acute myocardial infarction and myocarditis by inducing inflammatory responses. Activation of these receptors can also contribute to the development of different types of cardiomyopathies including diabetic cardiomyopathy, dilated cardiomyopathy and hypertrophic cardiomyopathy by inducing cardiac hypertrophy, fibrosis and apoptosis. Notably, inhibition of P2X7Rs can improve cardiac structure and function abnormalities following acute myocardial infarction, reduction of inflammatory responses following myocarditis and attenuation of the cardiomyopathy process. Furthermore, recent evidence has demonstrated that P2X7Rs are highly active in patients infected with coronavirus disease‑2019 (COVID‑19). Hyperactivation of P2X7Rs in COVID‑19 may induce severe myocardial injury through the activation of several signaling pathways. The present study reviewed the important role of the P2X7R in cardiac dysfunctions and discusses its use as a possible new therapeutic approach for the prevention and treatment of several myocardial diseases.

Introduction

Cardiovascular diseases are the main cause of mortality globally, accounting for ~30% of annual worldwide mortalities (1). This number is expected to rise to ~40% by the year 2030 (2). The high mortality rate of cardiovascular diseases indicates that further assessments are required to understand the mechanisms underlying the development of these diseases and identify other pharmacological agents targeting protection against heart injuries. The P2X7 purinergic receptor (P2X7R) has been implicated in several signaling pathways and in the development of a variety of pathological conditions including chronic neuropathic pain, neurodegenerative diseases, such as multiple sclerosis, inflammatory disorders, orthopedic diseases, such as osteoporosis and cancerous diseases such as lung cancer (310). Studies have highlighted the role of the P2X7R in the development of heart diseases, such as acute myocardial infarction, myocardial ischemia-reperfusion injury and myocarditis (1113). It is interesting to note that the importance of the P2X7R in heart injury mediated by the coronavirus disease-2019 (COVID-19) has been highlighted by various studies (1416). These studies have revealed a potential new approach that positions the P2X7R as a prognostic cardiac biomarker and pharmacological target for the prevention and treatment of heart injuries, which may increase survival and improve the quality of life in patients with heart diseases. The present review article provided a brief overview of the properties of the P2X7R, its distribution in the heart and its pathological role in heart diseases, with a particular focus on acute myocardial infarction, myocardial ischemia-reperfusion injury, autoimmune myocarditis and various types of cardiomyopathies as well as myocardial injury induced by COVID-19.

P2X7R

P2X7R belongs to a family of purinergic receptors. This family is categorized into two main groups, namely the P1 and P2 receptors (17). P2 receptors are subdivided into P2X receptors (P2XRs), which are ligand-gated ion channels and P2Y receptors (P2YRs), which are G-protein coupled receptors (18,19). A total of seven P2XR subtypes (P2X1R-P2X7R) and eight P2YR subtypes (P2Y1R, P2Y2R, P2Y4R, P2YR6 and P2Y11R-P2Y14R) have been identified to date (20,21). All subtypes of P2XR are non-selective cation channels activated by exogenous adenosine triphosphate (ATP), which is triggered by the efflux of K+ and the influx of Na+ and Ca2+ (22). ATP is stored intracellularly in synaptic vesicles and is released from neuronal and non-neuronal cells in response to various stimuli such as hypoxia, pain, infection and inflammation (23,24). It is released into the extracellular space by vesicular exocytosis or pore-forming channels by pannexin 1 and connexin 43 (2326). The release of ATP by vesicular exocytosis is a calcium-dependent release that is dependent on an increase in intracellular calcium concentration, whereas the ATP release through pore-forming channels is a calcium-independent release (27).

P2X7R has distinctive features that differentiate it from the other P2XRs. One of these features is that it is activated by ATP with a half maximal effective concentration (EC50) value in the range of 0.1–1 mM. This range is higher than that of the other P2XRs (EC50, 1–10 µM) (28). Its activation requires a massive amount of extracellular ATP, which does not usually exist in normal cells (29). It seems likely that P2X7Rs have low activity levels under normal conditions (Fig. 1A). By contrast, during certain pathological conditions, including hypoxia, inflammation, pain, cellular damage and other stress conditions, high levels of extracellular ATP can activate P2X7Rs and subsequent multiple signaling pathways (30). Activation of P2X7Rs opens cationic channels that facilitate the flux of small cations, such as K+, Na+ and Ca2+, resulting in the activation of several intracellular signaling pathways (31). These pathways include the activation of nucleotide-like receptor family pyrin domain member 3 (NLRP3) inflammasome, NF-κB, nuclear factor of activated T cells (NFAT), hypoxia-inducible factor 1-alpha (HIF-1α) and ERK1/2 as well as the formation of reactive oxygen species (ROS; Fig. 1B) (32,33). Furthermore, sustained activation of P2X7Rs opens large pores that facilitate the passage of large cations and organic dyes, such as choline and ethidium, respectively, which result in apoptotic cell death (3436).

The NLRP3 inflammasome is the most notable response following P2X7R activation. The binding of ATP to P2X7Rs allows K+ efflux and Na+ and Ca2+ influx. Low levels of intracellular K+ induce the configuration of the NLRP3 inflammasome with apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). This stimulates caspase-1 that cleaves the pro-inflammatory cytokines pro-IL-1β and pro-IL-18 to form IL-1β and IL-18, thus contributing to a series of inflammatory responses (37). These cytokines may induce profibrotic TGF-β1, resulting in fibrosis (38). Furthermore, the NLRP3 inflammasome with ASC activates caspase-1 that cleaves Gasdermin-D, which forms membrane pores and promotes the inflammatory cell death program (39). P2X7R induces inflammation by activation of NF-κB which increases several inflammatory cytokine genes, such as TNF-α and IL-1β (40). Similarly, P2X7R activation promotes NFAT function, which in turn leads to IL-2 inflammatory cytokine secretion, downregulation of glycogen synthase kinase activity and lymphocyte proliferation (32,41,42). HIF-1α is also a marked response following P2X7R activation. Ca2+ influx induced by P2X7R activation upregulates HIF-1α through phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway (43,44). This association between P2X7R and HIF-1α may promote angiogenesis by activating the vascular endothelial growth factor (VEGF) secretion (45,46). Furthermore, Ca2+ influx after P2X7R activation may induce cell proliferation and migration by enhancing ERK1/2 phosphorylation and activating NF-κB transcription, which in turn leads to the secretion of matrix metalloproteinases (MMPs) (47,48). In addition, the P2X7R activation may induce cell death by the formation of ROS (49). Taken together, the data indicate that P2X7R is possibly a starting point for the activation of several intracellular signaling pathways, which in turn cause inflammation, fibrosis, proliferation, angiogenesis and cell death. These multiple intracellular signaling pathways induced by the P2X7R activity indicate that P2X7Rs may represent an attractive target for the prevention and treatment of numerous pathological conditions.

P2X7R in the heart

P2X7Rs are distributed widely throughout the body. In the heart, P2X7Rs are found in embryonic stem cell-derived cardiomyocytes and their activation can increase the expression levels of several cardiac-specific genes, such as α-myosin heavy chain (α-MHC) and α-actinin (50). They are also expressed in epicardium-derived cells, indicating that they play roles in embryonic cardiac growth and development (51). P2X7Rs are mainly present in the sinoatrial node, right atrium and left ventricular. In the rat heart, P2X7R is highly expressed in the right atrium and left ventricular, while in the human heart it is highly expressed in the right atrium (52). Furthermore, these receptors are present in cardiac muscle cells notably atrial cardiomyocytes and other cardiac cells, such as cardiac endothelial cells and cardiac fibroblasts (53,54). The wide distribution of the P2X7R in cardiac cells suggests that this receptor has a crucial role in several cardiac pathological conditions, which are discussed in detail below.

P2X7R in acute myocardial infarction

Acute myocardial infarction (AMI), commonly called a heart attack, is a serious condition characterized by sustained ischemia and reduced blood flow to the heart muscle, resulting in an accelerated death of heart muscle cells (55). Several cellular and molecular changes occur following myocardial infarction, which can be categorized into the inflammatory, proliferative and maturation phases. Firstly, the inflammatory phase (the early phase within 0–4 days) is characterized by the release of ROS and pro-inflammatory mediators as well as fibrin deposition and necrosis of cardiomyocytes. Secondly, the proliferative phase (within ~1–2 weeks) is characterized by extracellular matrix deposition, myofibroblast differentiation, angiogenesis and the formation of tissue granules. Thirdly, the maturation phase (from weeks to months), is characterized by apoptosis of myofibroblasts and the formation of a mature scar (5658).

A strong relationship has been noted between P2X7R expression and AMI. P2X7Rs are upregulated in an experimental in vivo AMI model (13,59,60). A clinical study confirmed that P2X7R mRNA expression was upregulated in patients with AMI (61), suggesting that P2X7Rs may be used as predictive biomarkers in AMI. Previous studies have shown that the upregulation of P2X7R expression in AMI is commonly accompanied by an enhanced inflammatory response. For example, overexpression of P2X7R following myocardial infarction increases the NLRP3 inflammasome and release the proinflammatory cytokine IL-1β (59). Furthermore, activation of P2X7Rs aggravated AMI injury in an animal model resulting in increased ROS levels and vasopressin activity (60). Taken together, these studies indicate that the P2X7R plays a key role in the pathogenesis of myocardial infarction.

The inhibition of P2X7Rs may enhance cardiac function and improve survival following AMI. Gao et al (13) indicated that inhibition of P2X7Rs with short hairpin RNA attenuated sympathetic hyperinnervation by suppressing nerve growth factor levels. This study further indicated that inhibition of P2X7Rs ameliorated inflammatory infiltration by suppressing NF-κB activation and improved cardiac dysfunction by inhibiting the AKT/ERK1/2 signaling pathways (13). Furthermore, Mezzaroma et al (62) demonstrated that inhibition of P2X7Rs with small interfering RNA prevents caspase-1 activity and ameliorates cardiac remodeling. The role of P2X7R in AMI was confirmed by using specific P2X7R antagonists. It was shown that the P2X7R antagonist pyridoxalphosphate-6-azophenyl-2′,4-disulfonic acid hindered the formation of the ASC/cryopyrin inflammasome and reduced infarct size as well as cell death following AMI (62). In addition, the application of the P2X7R antagonist Brilliant Blue G (BBG) attenuated sympathetic hyperactivity and cardiac dysfunction by reducing oxidative stress as well as vasopressinergic cell activation in AMI rats (60). These studies indicate that P2X7R is a possible candidate for the treatment of myocardial infarction, particularly during the early inflammatory phase. The effect of P2X7Rs in the late phases of myocardial infarction remains to be elucidated. Therefore, further studies are required to verify whether the P2X7R plays a key role during the proliferation and maturation phases following myocardial infarction.

P2X7R in myocardial ischemia-reperfusion injury

Myocardial ischemia/reperfusion (I/R) injury is a complex condition characterized by the restoration of blood flow to the ischemic heart muscle (63). P2X7R expression is upregulated in the experimental model of myocardial I/R injury (64,65). There is controversy regarding the ability of P2X7R to exacerbate or reduce myocardial I/R injury. A previous study indicated that the P2X7R promoted cardiac damage by inducing inflammatory responses in myocardial I/R injury. In an animal model of myocardial I/R injury, overexpression of the P2X7R was shown to increase the activity of NF-κB and release several inflammatory cytokines, such as IL-6, IL-8, IL-10 and TNF-α (64). However, it is still unknown whether P2X7R inhibitors can preserve heart function in response to myocardial I/R injury. Therefore, further assessment studies on the effect of the P2X7R-induced inflammatory response in myocardial I/R injury are recommended.

In contrast to these findings, other studies have shown that the activation of the P2X7R can alleviate myocardial I/R injury by stimulating the release of endogenous cardioprotectants. During myocardial I/R injury, it was demonstrated that pannexin-1 interacts with P2X7Rs to form a channel. The activation of this channel is responsible for the release of the cardioprotectants adenosine and sphingosine 1-phosphate (11). These cardioprotectants can attenuate mitochondrial damage, prevent myocardial apoptosis and improve myocardial I/R injury via the activation of the PI3K/AKT signaling pathway (66). This was confirmed by using pannexin-1 and P2X7R antagonists in an animal model. Vessey et al (66) indicated that the pannexin-1 antagonist carbenoxolone and the P2X7R antagonist BBG increases infarct size and blocks cardioprotection in a rat experimental myocardial I/R injury model. Furthermore, overexpression of the P2X7R following myocardial ischemia increases ERK1/2 phosphorylation, suggesting that the P2X7R may prevent myocardial apoptosis and attenuate cardiomyocyte injury in response to I/R (65,67). Considering all this evidence, the activation of P2X7R seems to be beneficial in myocardial I/R injury, suggesting that the P2X7R agonist is a possible target for the prevention of myocardial I/R injury.

P2X7R in autoimmune myocarditis

Autoimmune myocarditis is an inflammatory disease of the myocardium characterized by the infiltration of inflammatory monocytes, macrophages and CD4+ helper T cells into the myocardium and consequently fibrosis and necrosis (6870). P2X7R expression is upregulated in ~50% of the experimental models of autoimmune myocarditis, which leads to CD4+ helper T cell and macrophage infiltration (12). A previous study using a mouse model of autoimmune cardiomyopathy indicated that the wild-type mice demonstrated increases IL-1β and IL-17 cytokine production. However, the levels of these cytokines are decreased in P2X7R−/−mice (71). In addition, treatment of mice with autoimmune myocarditis mice with the P2X7R antagonist A740003 improves their cardiac function by inhibiting CD4+ helper T cell and macrophage infiltration (12). These studies indicated that the P2X7R is a possible target for the treatment of autoimmune myocarditis.

P2X7R in cardiomyopathies

Cardiomyopathy is a disorder of the cardiac muscle characterized by structural and functional changes of cardiomyocytes (72). A total of two types of cardiomyopathies have been identified, primary or secondary. Primary cardiomyopathies can be divided into three main classes as follows: Genetic (hypertrophic cardiomyopathy), mixed-genetic and non-genetic (dilated cardiomyopathy) and acquired (inflammatory cardiomyopathy). Secondary cardiomyopathies are usually associated with a variety of systemic disorders (diabetes) and toxicity of specific drugs (chemotherapy) (72). Cardiac fibrosis is a key feature of various cardiomyopathies and is defined as an excess deposition of the extracellular matrix including type I collagen by cardiac fibroblasts (73). It is notable that activation of P2X7Rs by the P2X7R agonist BzATP elevates the protein expression of profibrotic markers, including TGF-β1, connective tissue growth factor (CTGF) and α-smooth muscle actin (α-SMA) in neonatal rat cardiac fibroblasts (67). This indicates that that P2X7Rs participate in the development of cardiac fibrosis (74).

An association between P2X7 expression and cardiomyopathy has been shown since overexpression of this receptor has been observed in various forms of cardiomyopathy. It has been demonstrated that P2X7Rs are upregulated in mouse models of experimental diabetes (68) and experimental dilated cardiomyopathies (75). Overexpression of P2X7Rs in animal cardiomyopathy models is commonly accompanied by cardiac hypertrophy, fibrosis and apoptosis. For example, overexpression of P2X7R in a mouse model of diabetic cardiomyopathy increases cardiac hypertrophy markers, such as atrial natriuretic peptide and β-myosin heavy chain, fibrosis markers, such as collagen I and TGF-β1 and apoptosis markers, such as caspase 3 and Bax (75). A genetic study demonstrated an association between P2X7R expression and cardiomyopathy. In humans, the single nucleotide polymorphism of P2X7R (E186K) that results in loss of function is associated with hypertrophic cardiomyopathy (76).

Inhibition of P2X7R can attenuate cardiac fibrosis in cardiomyopathies. Cardiac fibrosis markers including collagen I, CTGF, α-SMA and TGF-β1 are reduced in a P2X7R knockdown model. This was supported by using P2X7R antagonists in an animal model of cardiomyopathy. The application of the P2X7R antagonist BBG attenuated cardiac fibrosis by inhibiting the NLRP3/IL-1β signaling pathway (74). The application of the P2X7R antagonist A438079 ameliorates cardiac hypertrophy, fibrosis and apoptosis by inhibiting the PKCβ/ERK signaling pathway (75). These data demonstrate that P2X7Rs have a potential role in the prevention and/or treatment of various forms of cardiomyopathy.

Involvement of the P2X7R in heart injury mediated by COVID-19

COVID-19 is a respiratory viral infection characterized by the excessive and sustained production of inflammatory cytokines, the so-called cytokine storm (15,77). This disease can cause serious injuries in various organ systems, including the cardiovascular system. Viral myocarditis is the most common form of heart injury mediated by COVID-19 (78). Arrhythmia and myocardial infarction have also been reported as heart injuries in patients with COVID-19 (79,80). There is growing evidence that demonstrates the importance of purinergic receptors, particularly the P2X7R, in COVID-19 (81). It is important to note that the P2X7R is hyperactivated in patients infected with the COVID-19 viral strain. This hyperactivity can cause myocardial injury by the activation of several intracellular signaling pathways. The first possible pathway is the cytokine storm. During this viral infection, the P2X7R is stimulated by high levels of ATP resulting in NLRP3 inflammasome activation and considerable inflammatory cytokine production. These cytokines contribute to the cardiac inflammatory responses that may cause AMI, viral myocarditis and arrhythmia (14,81). The second possible pathway involves the angiotensin-converting enzyme (ACE) II. During COVID-19 infection, the P2X7R triggers pathways associated with the action of the renin-angiotensin-aldosterone system (RAAS) (81). The main effector molecule in the RAAS is angiotensin II, which causes vasoconstriction, cardiac hypertrophy and apoptosis. This molecule is upregulated in several pathological conditions including cardiovascular diseases. In fact, ACE generates angiotensin II from angiotensin I, while ACE2 inhibits the activity of angiotensin II by transferring it to angiotensin 1–7. Therefore, ACE2 has a cardioprotective effect against myocardial injury (82). It has been demonstrated that during the COVID-19 viral infection, the virus enters human cells by binding to ACE2 resulting in the downregulation of the ACE2 signaling pathway, which can potentially cause myocardial injury (8385). Another possible pathway is that of VEGF. Hyperactivity of P2X7Rs following COVID-19 has been shown to increase VEGF production (15). This may stimulate angiogenesis in cardiac cells which in turn leads to cardiac hypertrophy. Collectively, these studies indicate that the P2X7R plays a major role in myocardial injury caused by COVID-19. It can be hypothesized that inhibition of P2X7R is a promising therapeutic target for the prevention or treatment of cardiac injuries in patients with COVID-19.

Conclusion

In conclusion, the aforementioned studies have shown that the P2X7R plays an essential role in the development of several heart diseases. The majority of the studies have demonstrated that the activation of the P2X7R promotes the development of AMI, autoimmune myocarditis and various types of cardiomyopathies including diabetic cardiomyopathy, dilated cardiomyopathy and hypertrophic cardiomyopathy. Activation of the P2X7R during COVID-19 infection may also enhance the process of myocardial injury via the activation of several intracellular signaling pathways. Based on this evidence, it is likely that the P2X7R can be used as a prognostic indicator for the detection of various heart diseases. Overall, P2X7R inhibitors appear to be a promising therapeutic target for the prevention or treatment of heart diseases, as these inhibitors have been shown to have primarily anti-inflammatory effects against AMI and myocarditis, as well as antifibrotic and antiapoptotic effects in the case of cardiomyopathies. This view is supported by a recent review that draws attention to purinergic receptors as therapeutic targets for the treatment of cardiovascular disease (86). However, it is important to note the efficacy of P2X7R inhibitors during the progression of heart disease when these inhibitors are used in clinical cardiology, as they may not have the same beneficial effects in different types of heart disease. This has been observed in experimental models of myocardial infarction, the use of P2X7R inhibitors may enhance cardiac function and significantly improve survival in the early inflammatory phase following myocardial infarction (13,60,62). On the other hand, the use of P2X7R inhibitors may increase infarct size and abrogate the cardioprotective effects of adenosine and sphingosine 1-phosphate in myocardial I/R injury (66). To develop a complete profile of P2X7R in the setting of heart diseases, additional experimental studies are required to elucidate the effects of P2X7R inhibitors on other common cellular and molecular features associated with heart diseases. For example, the effects of P2X7R inhibitors can be examined on cardiac fibroblast proliferation, myofibroblast differentiation, angiogenesis and scar formation following myocardial infarction. In addition, further studies are required to demonstrate the effect of P2X7R inhibitors on the COVID-19 signaling pathways associated with myocardial injury.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Data sharing is not applicable to this article, as no data sets were generated or analyzed during the current study.

Authors' contributions

AD conducted the literature research, wrote the manuscript and designed the figure. AA, TA and NA contributed to the writing and revisions of the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

WHO, . Cardiovascular Diseases (CVDs). Fact sheet. WHO; Geneva: 2021

2 

Mathers CD and Loncar D: Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 3:e4422006. View Article : Google Scholar : PubMed/NCBI

3 

Takenouchi T, Sekiyama K, Sekigawa A, Fujita M, Waragai M, Sugama S, Iwamaru Y, Kitani H and Hashimoto M: P2X7 receptor signaling pathway as a therapeutic target for neurodegenerative diseases. Arch Immunol Ther Exp (Warsz). 58:91–96. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Zhang WJ, Zhu ZM and Liu ZX: The role and pharmacological properties of the P2X7 receptor in neuropathic pain. Brain Res Bull. 155:19–28. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Alves LA, Bezerra RJS, Faria RX, Ferreira LG and da Silva Frutuoso V: Physiological roles and potential therapeutic applications of the P2X7 receptor in inflammation and pain. Molecules. 18:10953–10972. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Li Q, Zhu X, Song W, Peng X and Zhao R: The P2X7 purinergic receptor: A potential therapeutic target for lung cancer. J Cancer Res Clin Oncol. 146:2731–2741. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Yang C, Shi S, Su Y, Tong JS and Li L: P2X7R promotes angiogenesis and tumour-associated macrophage recruitment by regulating the NF-κB signalling pathway in colorectal cancer cells. J Cell Mol Med. 24:10830–10841. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Giannuzzo A, Saccomano M, Napp J, Ellegaard M, Alves F and Novak I: Targeting of the P2X7 receptor in pancreatic cancer and stellate cells. Int J Cancer Res. 139:2540–2552. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Huang H, He YM, Lin MM, Wang Y, Zhang X, Liang L and He X: P2X7Rs: New therapeutic targets for osteoporosis. Purinergic Signal. Feb 2–2022.(Epub ahead of print). View Article : Google Scholar

10 

Grygorowicz T, Strużyńska L, Sulkowski G, Chalimoniuk M and Sulejczak D: Temporal expression of P2X7 purinergic receptor during the course of experimental autoimmune encephalomyelitis. Neurochem Int. 57:823–829. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Vessey DA, Li L and Kelley M: Pannexin-I/P2X 7 purinergic receptor channels mediate the release of cardioprotectants induced by ischemic pre-and postconditioning. J Cardiovasc Pharmacol Ther. 15:190–195. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Zempo H, Sugita Y, Ogawa M, Watanabe R, Suzuki J and Isobe M: A P2X7 receptor antagonist attenuates experimental autoimmune myocarditis via suppressed myocardial CD4+ T and macrophage infiltration and NADPH oxidase 2/4 expression in mice. Heart Vessels. 30:527–533. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Gao H, Yin J, Shi Y, Hu H, Li X, Xue M, Cheng W, Wang Y, Li X, Li Y, et al: Targeted P2X7R shRNA delivery attenuates sympathetic nerve sprouting and ameliorates cardiac dysfunction in rats with myocardial infarction. Cardiovasc Ther. 35:2017. View Article : Google Scholar

14 

Dos Anjos F, Simões JLB, Assmann CE, Carvalho FB and Bagatini MD: Potential therapeutic role of purinergic receptors in cardiovascular disease mediated by SARS-CoV-2. J Immunol Res. 2020:86320482020. View Article : Google Scholar : PubMed/NCBI

15 

Di Virgilio F, Tang Y, Sarti AC and Rossato M: A rationale for targeting the P2X7 receptor in Coronavirus disease 19. Br J Pharmacol. 177:4990–4994. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Batista Simões JL, Sobierai LD, Pereira SM, Rodrigues dos Santos MV and Bagatini MD: Therapeutic potential of P2X7 purinergic receptor modulation in the main organs affected by the COVID-19 Cytokine Storm. Curr Pharm Des. 28:1798–1814. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Burnstock G: A basis for distinguishing two types of purinergic receptor. Cell Membrane Receptors for Drugs and Hormone: A Multidisciplinary Approach. 107–118. 1978.

18 

Burnstock G and Kennedy C: Is there a basis for distinguishing two types of P2-purinoceptor? Gen Pharmacol. 16:433–440. 1985. View Article : Google Scholar : PubMed/NCBI

19 

Abbracchio MP and Burnstock G: Purinoceptors: Are there families of P2X and P2Y purinoceptors? Pharmacol Ther. 64:445–475. 1994. View Article : Google Scholar : PubMed/NCBI

20 

North RA: Molecular physiology of P2X receptors. Physiol Rev. 82:1013–1067. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Abbracchio MP, Burnstock G, Boeynaems JM, Barnard EA, Boyer JL, Kennedy C, Knight GE, Fumagalli M, Gachet C, Jacobson KA and Weisman GA: International Union of Pharmacology LVIII: Update on the P2Y G protein-coupled nucleotide receptors: From molecular mechanisms and pathophysiology to therapy. Pharmacol Rev. 58:281–341. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Burnstock G and Verkhratsky A: Receptors for purines and pyrimidines. Springer Berlin; Heidelberg: 2012, View Article : Google Scholar

23 

Bodin P and Burnstock G: Purinergic signalling: ATP release. Neurochem Res. 26:959–969. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Dosch M, Gerber J, Jebbawi F and Beldi G: Mechanisms of ATP release by inflammatory cells. Int J Mol Sci. 19:12222018. View Article : Google Scholar : PubMed/NCBI

25 

Sawada K, Echigo N, Juge N, Miyaji T, Otsuka M, Omote H, Yamamoto A and Moriyama Y: Identification of a vesicular nucleotide transporter. Proc Natl Acad Sci USA. 105:5683–5686. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Junger WG: Immune cell regulation by autocrine purinergic signalling. Nat Rev Immunol. 11:201–212. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Xiong Y, Sun S, Teng S, Jin M and Zhou Z: Ca2+-dependent and Ca2+-independent ATP release in astrocytes. Front Mol Neurosci. 11:2242018. View Article : Google Scholar : PubMed/NCBI

28 

Abbracchio MP, Burnstock G, Verkhratsky A and Zimmermann H: Purinergic signalling in the nervous system: An overview. Trends Neurosci. 32:19–29. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Kuzmin AI, Lakomkin VL, Kapelko VI and Vassort G: Interstitial ATP level and degradation in control and postmyocardial infarcted rats. Am J Physiol. 275:C766–C771. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Jiang LH, Baldwin JM, Roger S and Baldwin SA: Insights into the molecular mechanisms underlying mammalian P2X7 receptor functions and contributions in diseases, revealed by structural modeling and single nucleotide polymorphisms. Front Pharmacol. 4:552013. View Article : Google Scholar : PubMed/NCBI

31 

Burnstock G and Kennedy C: P2X receptors in health and disease. Adv Pharmacol. 61:333–372. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Adinolfi E, Giuliani AL, De Marchi E, Pegoraro A, Orioli E and Di Virgilio F: The P2X7 receptor: A main player in inflammation. Biochem Pharmacol. 151:234–244. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Andrejew R, Oliveira-Giacomelli Á, Ribeiro DE, Glaser T, Arnaud-Sampaio VF, Lameu C and Ulrich H: The P2X7 receptor: Central hub of brain diseases. Front Mol Neurosci. 13:1242020. View Article : Google Scholar : PubMed/NCBI

34 

Virginio C, MacKenzie A, Rassendren FA, North RA and Surprenant A: Pore dilation of neuronal P2X receptor channels. Nat Neurosci. 2:315–321. 1999. View Article : Google Scholar : PubMed/NCBI

35 

Wiley JS, Sluyter R, Gu BJ, Stokes L and Fuller SJ: The human P2X7 receptor and its role in innate immunity. Tissue Antigens. 78:321–332. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Alves LA, de Melo Reis RA, de Souza CA, de Freitas MS, Teixeira PC, Neto Moreira Ferreira D and Xavier RF: The P2X7 receptor: Shifting from a low-to a high-conductance channel-an enigmatic phenomenon? Biochim Biophys Acta. 1838:2578–2587. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Bartlett R, Stokes L and Sluyter R: The P2X7 receptor channel: Recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev. 66:638–675. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Artlett CM: The role of the NLRP3 inflammasome in fibrosis. Open Rheumatol J. 6:80–86. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H and Lieberman J: Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 535:153–158. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Ferrari D, Wesselborg S, Bauer MK and Schulze-Osthoff K: Extracellular ATP activates transcription factor NF-kappaB through the P2Z purinoreceptor by selectively targeting NF-kappaB p65 (RelA). J Cell Biol. 139:1635–1643. 1997. View Article : Google Scholar : PubMed/NCBI

41 

Ferrari D, Stroh C and Schulze-Osthoff K: P2X7/P2Z purinoreceptor-mediated activation of transcription factor NFAT in microglial cells. J Biol Chem. 274:13205–13210. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Yip L, Woehrle T, Corriden R, Hirsh M, Chen Y, Inoue Y, Ferrari V, Insel PA and Junger WG: Autocrine regulation of T-cell activation by ATP release and P2X7 receptors. FASEB J. 23:1685–1693. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Zhang Y, Cheng H, Li W, Wu H and Yang Y: Highly-expressed P2X7 receptor promotes growth and metastasis of human HOS/MNNG osteosarcoma cells via PI3K/Akt/GSK3β/β-catenin and mTOR/HIF1α/VEGF signaling. Int J Cancer. 145:1068–1082. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Langhner E, Taghavi P, Chiles K, Mahon PC and Semenza GL: HEER2 (neu) signaling increase the rate of hypoxia inducible factor 1-alpha (HIF-1-alpha) synthesis: Novel mechanism for HIF-mediated vascular endothelial growth factor expression. Mol Cell Bioi. 21:3995–4004. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Amoroso F, Capece M, Rotondo A, Cangelosi D, Ferracin M, Franceschini A, Raffaghello L, Pistoia V, Varesio L and Adinolfi E: The P2X7 receptor is a key modulator of the PI3K/GSK3β/VEGF signaling network: Evidence in experimental neuroblastoma. Oncogene. 34:5240–5251. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Hill LM, Gavala ML, Lenertz LY and Bertics PJ: Extracellular ATP may contribute to tissue repair by rapidly stimulating purinergic receptor X7-dependent vascular endothelial growth factor release from primary human monocytes. J Immunol. 185:3028–3034. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Tafani M, Schito L, Pellegrini L, Villanova L, Marfe G, Anwar T, Rosa R, Indelicato M, Fini M, Pucci B and Russo MA: Hypoxia-increased RAGE and P2X7R expression regulates tumor cell invasion through phosphorylation of Erk1/2 and Akt and nuclear translocation of NF-{kappa}B. Carcinogenesis. 32:1167–1175. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Ji Z, Xie Y, Guan Y, Zhang Y, Cho KS, Ji M and You Y: Involvement of P2X7 receptor in proliferation and migration of human glioma cells. Biomed Res Int. 2018:85913972018. View Article : Google Scholar : PubMed/NCBI

49 

Bartlett R, Yerbury JJ and Sluyter R: P2X7 receptor activation induces reactive oxygen species formation and cell death in murine EOC13 microglia. Mediators Inflamm. 2013:2718132013. View Article : Google Scholar : PubMed/NCBI

50 

Mazrouei S, Sharifpanah F, Bekhite MM, Figulla HR, Sauer H and Wartenberg M: Cardiomyogenesis of embryonic stem cells upon purinergic receptor activation by ADP and ATP. Purinergic Signal. 11:491–506. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Hesse J, Leberling S, Boden E, Friebe D, Schmidt T, Ding Z, Dieterich P, Deussen A, Roderigo C, Rose CR, et al: CD73-derived adenosine and tenascin-C control cytokine production by epicardium-derived cells formed after myocardial infarction. FASEB J. 31:3040–3053. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Musa H, Tellez JO, Chandler NJ, Greener ID, Mączewski M, Mackiewicz U, Beresewicz A, Molenaar P, Boyett MR and Dobrzynski H: P2 purinergic receptor mRNA in rat and human sinoatrial node and other heart regions. Naunyn Schmiedebergs Arch Pharmacol. 379:541–549. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Barth K, Pfleger C, Linge A, Sim JA, Surprenant A, Steinbronn N, Strasser RH and Kasper M: Increased P2X7R expression in atrial cardiomyocytes of caveolin-1 deficient mice. Histochem. Cell Biol. 134:31–38. 2010.PubMed/NCBI

54 

Gentile D, Natale M, Lazzerini PE, Capecchi PL and Laghi-Pasini F: The role of P2X7 receptors in tissue fibrosis: A brief review. Purinergic Signal. 11:435–440. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD; Joint ESC/ACCF/AHA/WHF Task Force for Universal Definition of Myocardial Infarction; Authors/Task Force Members Chairpersons, ; Thygesen K, Alpert JS, et al: Third universal definition of myocardial infarction. J Am Coll Cardiol. 60:1581–1598. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Liehn EA, Postea O, Curaj A and Marx N: Repair after myocardial infarction, between fantasy and reality: The role of chemokines. J Am Coll Cardiol. 58:2357–2362. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Forte E, Furtado MB and Rosenthal N: The interstitium in cardiac repair: Role of the immune-stromal cell interplay. Nat Rev Cardiol. 15:601–616. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Ferrini A, Stevens MM, Sattler S and Rosenthal N: Toward regeneration of the heart: Bioengineering strategies for immunomodulation. Front Cardiovasc Med. 6:262019. View Article : Google Scholar : PubMed/NCBI

59 

Yin J, Wang Y, Hu H, Li X, Xue M, Cheng W, Wang Y, Li X, Yang N, Shi Y and Yan S: P2X7 receptor inhibition attenuated sympathetic nerve sprouting after myocardial infarction via the NLRP3/IL-1β pathway. J Cell Mol Med. 21:2695–2710. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Cheng W, Sun Y, Wu Q, Ooi K, Feng Y, Xia C and Zhu D: Paraventricular nucleus P2X7 receptors aggravate acute myocardial infarction injury via ROS-induced vasopressin-V1b activation in rats. Neurosci Bull. 37:641–656. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Shi XX, Zheng KC, Shan PR, Zhang L, Wu SJ and Huang ZQ: Elevated circulating level of P2X7 receptor is related to severity of coronary artery stenosis and prognosis of acute myocardial infarction. Cardiol J. 28:453–459. 2021. View Article : Google Scholar : PubMed/NCBI

62 

Mezzaroma E, Toldo S, Farkas D, Seropian IM, Van Tassell BW, Salloum FN, Kannan HR, Menna AC, Voelkel NF and Abbate A: The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc Natl Acad Sci USA. 108:19725–19730. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Frank A, Bonney M, Bonney S, Weitzel L, Koeppen M and Eckle T: Myocardial ischemia reperfusion injury: From basic science to clinical bedside. Semin Cardiothorac Vasc Anesth. 16:123–132. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Gu M, Zheng AB, Jin J, Cui Y, Zhang N, Che ZP, Wang Y, Zhan J and Tu WJ: Cardioprotective effects of genistin in rat myocardial ischemia-reperfusion injury studies by regulation of P2X7/NF-κB pathway. Evid Based Complement Alternat Med. 2016:53812902016. View Article : Google Scholar : PubMed/NCBI

65 

Tu G, Zou L, Liu S, Wu B, Lv Q, Wang S, Xue Y, Zhang C, Yi Z, Zhang X, et al: Long noncoding NONRATT021972 siRNA normalized abnormal sympathetic activity mediated by the upregulation of P2X7 receptor in superior cervical ganglia after myocardial ischemia. Purinergic Signal. 12:521–535. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Vessey DA, Li L and Kelley M: Ischemic preconditioning requires opening of pannexin-1/P2X7 channels not only during preconditioning but again after index ischemia at full reperfusion. Mol Cell Biochem. 351:77–84. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Wang Y: Mitogen-activated protein kinases in heart development and diseases. Circulation. 116:1413–1423. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Magnani JW and Dec GW: Myocarditis: Current trends in diagnosis and treatment. Circulation. 113:876–890. 2006. View Article : Google Scholar : PubMed/NCBI

69 

Fung G, Luo H, Qiu Y, Yang D and McManus B: Myocarditis. Circ Res. 118:496–514. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Amoah BP, Yang H, Zhang P, Su Z and Xu H: Immunopathogenesis of myocarditis: The interplay between cardiac fibroblast cells, dendritic cells, macrophages and CD 4+ T cells. Scand J Immunol. 82:1–9. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Martinez CG, Zamith-Miranda D, Da Silva MG, Ribeiro KC, Brandão IT, Silva CL, Diaz BL, Bellio M, Persechini PM and Kurtenbach E: P2×7 purinergic signaling in dilated cardiomyopathy induced by auto-immunity against muscarinic M2 receptors: Autoantibody levels, heart functionality and cytokine expression. Sci Rep. 5:169402015. View Article : Google Scholar : PubMed/NCBI

72 

Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, Moss AJ, Seidman CE, Young JB; American Heart Association, ; et al: Contemporary definitions and classification of the cardiomyopathies: An American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention. Circulation. 113:1807–1816. 2006. View Article : Google Scholar : PubMed/NCBI

73 

Eijgenraam TR, Silljé HHW and de Boer RA: Current understanding of fibrosis in genetic cardiomyopathies. Trends Cardiovasc Med. 30:353–361. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Zhou J, Tian G, Quan Y, Li J, Wang X, Wu W, Li M and Liu X: Inhibition of P2X7 purinergic receptor ameliorates cardiac fibrosis by suppressing NLRP3/IL-1β pathway. Oxid Med Cell Longev. 2020:79562742020. View Article : Google Scholar : PubMed/NCBI

75 

Huang S, Wang W, Li L, Wang T, Zhao Y, Lin Y, Huang W, Wang Y and Huang Z: P2X7 receptor deficiency ameliorates STZ-induced cardiac damage and remodeling through PKCβ and ERK. Front Cell Dev Biol. 9:6920282021. View Article : Google Scholar : PubMed/NCBI

76 

Biswas A, Raza A, Das S, Kapoor M, Jayarajan R, Verma A, Shamsudheen KV, Murry B, Seth S, Bhargava B, et al: Loss of function mutation in the P2X7, a ligand-gated ion channel gene associated with hypertrophic cardiomyopathy. Purinergic Signal. 15:205–210. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Whitworth J: COVID-19: A fast evolving pandemic. Trans R Soc Trop Med Hyg. 114:241–248. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Siripanthong B, Nazarian S, Muser D, Deo R, Santangeli P, Khanji MY, Cooper LT Jr and Chahal CAA: Recognizing COVID-19-related myocarditis: The possible pathophysiology and proposed guideline for diagnosis and management. Heart Rhythm. 17:1463–1471. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Kang Y, Chen T, Mui D, Ferrari V, Jagasia D, Scherrer-Crosbie M, Chen Y and Han Y: Cardiovascular manifestations and treatment considerations in COVID-19. Heart. 106:1132–1141. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Guzik TJ, Mohiddin SA, Dimarco A, Patel V, Savvatis K, Marelli-Berg FM, Madhur MS, Tomaszewski M, Maffia P, D'Acquisto F, et al: COVID-19 and the cardiovascular system: Implications for risk assessment, diagnosis, and treatment options. Cardiovasc Res. 116:1666–1687. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Ribeiro DE, Oliveira-Giacomelli Á, Glaser T, Arnaud-Sampaio VF, Andrejew R, Dieckmann L, Baranova J, Lameu C, Ratajczak MZ and Ulrich H: Hyperactivation of P2X7 receptors as a culprit of COVID-19 neuropathology. Mol Psychiatry. 26:1044–1059. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K and Zhang C: Angiotensin-converting enzyme 2 and angiotensin 1–7: Novel therapeutic targets. Nat Rev Cardiol. 11:413–426. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, Guo L, Guo R, Chen T, Hu J, et al: Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun. 11:16202020. View Article : Google Scholar : PubMed/NCBI

84 

Nishiga M, Wang DW, Han Y, Lewis DB and Wu JC: COVID-19 and cardiovascular disease: From basic mechanisms to clinical perspectives. Nat Rev Cardiol. 17:543–558. 2020. View Article : Google Scholar : PubMed/NCBI

85 

De Mello WC and Danser AH: Angiotensin II and the heart: On the intracrine renin-angiotensin system. Hypertension. 35:1183–1188. 2000. View Article : Google Scholar : PubMed/NCBI

86 

Wernly B and Zhou Z: More purinergic receptors deserve attention as therapeutic targets for the treatment of cardiovascular disease. Am J Physiol Heart Circ Physiol. 319:H723–H729. 2020. View Article : Google Scholar : PubMed/NCBI

87 

Wagner JA and Kelly RB: Topological organization of proteins in an intracellular secretory organelle: The synaptic vesicle. Proc Natl Acad Sci USA. 76:4126–4130. 1979. View Article : Google Scholar : PubMed/NCBI

88 

Bours MJ, Swennen EL, Di Virgilio F, Cronstein BN and Dagnelie PC: Adenosine 5′-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation. Pharmacol Ther. 112:358–404. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 27 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bin Dayel AF, Alonazi AS, Alshammari TK and Alrasheed NM: P2X7 purinergic receptor: A potential target in heart diseases (Review). Mol Med Rep 27: 74, 2023
APA
Bin Dayel, A.F., Alonazi, A.S., Alshammari, T.K., & Alrasheed, N.M. (2023). P2X7 purinergic receptor: A potential target in heart diseases (Review). Molecular Medicine Reports, 27, 74. https://doi.org/10.3892/mmr.2023.12961
MLA
Bin Dayel, A. F., Alonazi, A. S., Alshammari, T. K., Alrasheed, N. M."P2X7 purinergic receptor: A potential target in heart diseases (Review)". Molecular Medicine Reports 27.3 (2023): 74.
Chicago
Bin Dayel, A. F., Alonazi, A. S., Alshammari, T. K., Alrasheed, N. M."P2X7 purinergic receptor: A potential target in heart diseases (Review)". Molecular Medicine Reports 27, no. 3 (2023): 74. https://doi.org/10.3892/mmr.2023.12961