Skip to main content
Purinergic Signalling logoLink to Purinergic Signalling
. 2021 Mar 22;17(2):229–240. doi: 10.1007/s11302-021-09774-x

Targeting P2 receptors in purinergic signaling: a new strategy of active ingredients in traditional Chinese herbals for diseases treatment

Xiaopeng Ai 1,3,#, Xing Dong 1,#, Ying Guo 1, Peng Yang 2, Ya Hou 1, Jinrong Bai 1, Sanyin Zhang 1,3,, Xiaobo Wang 1,
PMCID: PMC8155138  PMID: 33751327

Abstract

Adenosine triphosphate (ATP) and its metabolites adenosine diphosphate, adenosine monophosphate, and adenosine in purinergic signaling pathway play important roles in many diseases. Activation of P2 receptors (P2R) channels and subsequent membrane depolarization can induce accumulation of extracellular ATP, and furtherly cause kinds of diseases, such as pain- and immune-related diseases, cardiac dysfunction, and tumorigenesis. Active ingredients of traditional Chinese herbals which exhibit superior pharmacological activities on diversified P2R channels have been considered as an alternative strategy of disease treatment. Experimental evidence of potential ingredients in Chinese herbs targeting P2R and their pharmacological activities were outlined in the study.

Keywords: P2 receptors, Purinergic signaling pathway, Active ingredients of Chinese herbs

Introduction

Purine substances, being responsible for information transmission, define metabolic adenosine triphosphate (ATP), adenosine diphosphate (ADP), adenosine monophosphate (AMP), and adenosine by nucleotides [1]. Purinergic signaling pathway refers to the system of purinergic signal transduction consisting of endogenous nucleosides, nucleotides, and their receptors [2]. Sensitized purinergic receptors may perform different functions in the manner of neurotransmitter, autocrine or paracrine, and cause pain, inflammation, ischemic stroke, diabetes mellitus, and cancer [3, 4]. In the family of purinergic receptors, adenosine-activated P1 receptors (P1R) are comprised of four subtypes, A1R, A2AR, A2BR, and A3R [5]. ATP- and ADP-activated P2R are subdivided into two subtypes, P2XR and P2YR [6, 7]. The hydrolyzed ATP starts working after binding to P2R and then ecto-ATPases on the surface of most cells extract one and two phosphorus atoms of ATP, turning them into ADP and AMP, respectively, or even adenosine alone [8, 9]. In addition, ectonuleoside triphosphate phoshohydrolases (ENTPDases), eto-5’-nucleotidase (5’-NT), eto-nucleotide pyrophosphatase/phosphodoesterases (ENPPs), and alkaline phosphatases (APs) are involved in the degradation of ATP [1012]. ATP signaling can be classified into short term and long term [13]. In the purinergic signaling pathway, extracellular ATP is hydrolyzed to different ATP analogs via several enzymes [14]. ATP has been shown to function on purinergic receptors and then activate or inhibit the effector enzymes coupled with G proteins, thereby activating the amplification of the downstream signal cascade [15, 16]. ATP may also directly target the gated ion channels by ligands, allowing signal transduction to be accomplished by ion flow across the membrane [17]. Meanwhile, in both neurons and non-neurons, extracellular ATP plays a very vital biological role [18]. In the central and peripheral nervous systems, short-term ATP signaling could serve as a co-transmitter and neuromodulator to participate in platelet aggregation, endocrine and exocrine secretion, and blood vessel regulation [19]. However, cell proliferation, differentiation, and apoptosis are affected by long-term ATP signaling. It is also involved in the healing of wounds, atherosclerosis, and renovation of epithelial cells, inflammation, and also tumorigenesis [2022]. The development of distinct effects depends on the concentration of extracellular ATP, state of the target cell, and subtype of purinergic receptor [23] (Table 1).

Table 1.

Chinese herbals derived-compounds targeting purinergic P2 receptors and involved mechanisms.

graphic file with name 11302_2021_9774_Tab1a_HTML.jpg

graphic file with name 11302_2021_9774_Tab1b_HTML.jpg

MF, molecular formula; MW, molecular weight

Numerous studies have shown that a significant number of diseases are involved in purinergic signaling pathways [24, 25]. Nevertheless, most of the medications used for clinical treatment of the above-mentioned diseases have weak therapeutic effects and some obvious side effects, compromising patients’ quality of life and further raising the burden of society’s health care [26, 27]. Therefore, it is urgently needed to develop alternatives with better therapeutic effect and little impact on other normal physiological functions. In the clinical prevention and treatment of purine disorders with less side effects, conventional Chinese medicines (TCMs) such as acupuncture medicinal plants and their herbal extracts prospectively have specific advantages [2830]. In addition, several studies have shown that novel unique molecular entities derived from TCMs are capable of upgrading these diseases via P2R modulation [31, 32]. Thus, the purpose of this review was to summarize the active ingredients of TCMs that have potential cure effects by regulating P2 purinergic signaling pathways.

Active ingredients of TCMs for painful diseases management via P2X3R and P2X7R

As an uncomfortable feeling caused by tissue injury, depending on the time or period, pain can be divided into acute pain (physiological pain) and chronic pain (pathological pain) [3335]. Although acute pain evoked by trauma or surgery has self-healing properties, if left unchecked, it can be converted into chronic pain [36]. It can be further categorized into neuropathic pain, cancer pain, and inflammatory pain [37]. However, a growing number of animal experiments have shown that intradermal injection of ATP and its analogs can induce in vivo pain-defending behaviors [3840]. Neurons may be stimulated by activated nociceptors with ATP to generate extreme inward currents leading to neuropathic pain and abnormal behavioral manifestations [41, 42]. In detail, in acute isolation and culture of dorsal root ganglia (DRG) neurons, ATP or its agonists released by post-synaptic or impaired nerves can induce inward depolarizing currents and elicit pain mediated by P2XR [43]. Therefore, there is a close connection, particularly in neuropathic pain, between pain and purinergic signaling pathways. Researchers have recently found that some of the established TCM compounds had an effect on pain through purinergic P2R [44].

Tetramethylpyrazine (TMP) pain relief was closely linked to its robust anti-inflammatory effects as an active component of the amide alkaloid derived from Ligusticum walliichi [4547]. Nociceptive reactions caused by ATP or β-meATP have been shown to be inhibited by intrathecal application of TMP [48]. Meanwhile, intracellular records have shown that TMP can reduce the depolarization potential of ATP or β-meATP induced by the DRG nerve cell membrane [49]. By blocking the expression of P2X3R in trigeminal neuralgia, TMP may also inhibit P2X3R-mediated pain transmission in neuropathic primary sensory neurons in burned rats [50, 51]. Moreover, in HEK293 cells transfected with a P2X3R plasmid, TMP retained ATP-activated currents [52]. Ligusticum walliichi sodium ferulate (SF) could restrain hyperalgesia in DRG neurons and initiate pain and primary afferent sensitization in chronic constriction injury (CCI) rats by reducing pain transmission mediated by P2X3R [53, 54]. Similarly, decreased P2X3R expression and sensitization by lappaconitin reduced the analgesic effect of CCI rat DRG neurons [55, 56]. Hesperidin inhibited P2X3R mediated nociceptive transmission in the DRG of CCI rats [57]. Sinomenine’s suppressed expression of P2X3R can explain its anti-inflammatory pain effects [5860], while 1.8-cineole anti-inflammatory effects have contributed to the relief of pathological pain triggered by P2X3R stimulation [61, 62]. Robust anti-inflammatory, anti-cell proliferation, and anti-liver and kidney fibrosis is exerted by Emodin primarily isolated from rhubarb, polygonum, and buckthorn [6367]. More specifically, emodin blocked the thermal and mechanical allodynia transmission of neuropathic pain by reducing P2X2R and P2X3R expression [68]. Amusingly, curcumin nanoparticles with improved bioavailability can inhibit activation of P2X3R and P2Y12R, resulting in mechanical hyperalgesia and thermal hyperalgesia in gp120-treated rats by desensitizing the primary afferents of DRG [6971]. The natural Gardenia jasminoides Ellis fruit glycoside ligand gardenoside can be a possible medication for the treatment of sciatica by decreasing P2X3R and P2X7R [72]. Puerarin has been predominantly used clinically for pain-related cardiovascular disorders such as angina pectoris, myocardial infarction, and coronary heart disease as a flavonoid glycoside identified from radix puerariae [7375]. The mechanisms involved may be linked to the reduction of P2X3R and P2X7R mRNA and protein levels in peripheral mononuclear cells [7679]. In Polygonum cuspidatum, resveratrol (RES) relieves pain in peripheral and central inflammatory diseases by decreasing arachidonic acid metabolism [80]. In addition, RES suppressed neuropathic pain transmission in DRG satellite glial cells and relieved mechanical hyperalgesia in rats treated with gp120 by inhibiting P2X3R and P2X7R [8184]. Astragalin therapy has been reported to be able to reduce neuropathic pain by blocking P2X4-mediated signaling in rat DRG [85]. Artemisinin could inhibit the upregulation of P2X4R in satellite glial cells as a form of sesquiterpene lactone in Artemisia annua leaves, thereby alleviating abnormal mechanical and thermal pain in rats with neuropathic pain [8688]. Similarly, the anti-inflammatory and anti-oxidant properties of osthole may be impaired by downregulation of P2X4R and relief of mechanical and thermal hyperalgesia in DM rats [89, 90], as well as by neuropathic pain and nociceptive reactions [91]. Accumulated evidence indicates that purines contribute to the initiation and development of pain as a key component. To relieve pain-related diseases, the majority of active ingredients in TCMs will mainly target P2X3R and P2X7R.

Active ingredients of TCMs for inflammation-related diseases management via P2X4R and P2X7R

As a normal and essential reaction to infection, cell stress, and tissue damage, immoderate inflammation involves a complex process that helps the body to recognize and battle foreign antigens, causing rheumatoid arthritis, atherosclerosis, and cancer [9294]. The ATP purinergic signaling mediator has been shown to contribute to the fine tuning of inflammation and immune responses, effectively eliminating minimal damage to healthy tissues [95, 96]. Evidence has shown that the upregulated P2XR of immunocytes can trigger an inflammatory cascade [97]. Predominantly expressed P2X7R in various types of macrophages participated in immune responses and inflammatory diseases [98]. Predominantly expressed P2X7R in various types of macrophages participated in immune responses and inflammatory diseases [99].

By decreasing ATP hydrolysis, curcuminoids from ginger and turmeric exerted anti-inflammatory properties in hypertensive rats [100102]. The anti-inflammatory properties of mangiferin were correlated with the activation of adenosine-CD73 signaling pathway, attenuating renal ischemia reperfusion injury [103]. The P2X7R-related inflammation responses induced by ATP could be repressed by emodin as a potential P2X7R antagonist [104106]. Rhein is a natural bioactive derivative of anthraquinone extensively found widely in TCMs such as rhubarb, polygonum multiflorum, and aloe. They have anti-inflammation, anti-tumor, and anti-angiogenesis activities [107109]. Rhein has been reported to inhibit ATP-induced inflammatory responses through inhibition of P2X4R and P2X7R, as evidenced by depressed development of Ca2+ and ROS, as well as by suppressed expression of MMP-9, COX-2, IL-6, and IL-1β [110, 111]. Naringin inflammatory inhibition has also been confirmed in relation to the reduction of P2X7 expression in gp120-induced inflammation [112, 113]. Gardenia is a widely used TCMs in clinic in the treatment of inflammation and fever [114, 115]. Its bioactive compound geniposide is related to interfering with P2Y14R expression and inhibition of ERK1/2-mediated inflammatory events [116]. Interestingly, it was announced that low adenosine expression may intensify the inflammatory injury of renal ischemia reperfusion insults in rats, which was reversed by administration of ferulic acid [117, 118]. In extremely glucose-challenged human umbilical vein endothelial cells, evodiamine-suppressed P2X4R and P2X7R lead to inflammation recurrence [119121]. Bullatin A selectively antagonizing P2X7R was closely linked to ATP-induced BV-2 cell inflammatory injury inhibition [122, 123]. Reduced P2X7R expression was correlated with the anti-inflammatory effect of cis-RES, leading to suppression of ROS production and activation of caspase-1/4 [124, 125]. Existing inflammatory suppression of curcumin stopped post-stroke depression development by inhibiting the activity of P2X7R [126, 127]. According to the above literature, the active ingredient of TCMs can explicitly inhibit P2X4R and P2X7R activity, thereby restricting the expression of inflammatory factors such as IL-6 and IL-1β.

Active ingredients of TCMs for myocardial ischemia diseases management via P2X3R

Ischemic cardiomyopathy mainly manifested as metabolic disorders of cardiomyocytes, heart enlargement, arrhythmia, and heart failure, becoming the leading cause of death in the world [128130]. Adenine nucleotides and nucleosides can function on purinergic P2XR of cardiovascular system and improve the performance of ischemic myocardium by enhancing contractility and relaxation [131, 132]. Puerarin therapy reduced ATP-activated currents in DRG neurons and upregulated P2X3R in the superior cervical ganglia (SCG) and cardiac sympathetic nerves after myocardial ischemia [55, 133]. Oxymatrine withstood the exacerbated sympathoexcitatory reflex by decreasing the activation effect of P2X3R in SCG and DRG neurons, which blocked nociceptive transmission triggered by myocardial ischemia in rats [134, 135]. In diabetic cardiac autonomic neuropathy, treatment with baicalin reduced elevated expression and activation of P2Y12R and repressed phosphorylation of p38-induced inflammatory injury [136, 137]. In conclusion, TCMs active ingredients are potential drugs to confine myocardial ischemia diseases by ameliorating ATP metabolic disorders and reducing P2X3R in purinergic signaling.

Active ingredients of TCMs for cancer management via P2X7R

Cancer has become one of the major diseases affecting the health and longevity of people, given the increasing morbidity and mortality [138, 139]. It has been established that there are relatively high levels of ATP in the tumor microenvironment, raising the amount of purinoreceptors [140142]. Natural products actually play an important role in the treatment of cancer, with a large number of anti-cancer agents used in clinical therapies coming from a range of natural sources, including plants, animals, and microorganisms [143]. Zhang et al. showed that emodin exhibited strong cytotoxic activity against leukemia cells of K562 by inhibiting adenosine deaminase (ADA) activity [144]. Simultaneously, emodin could reduce the invasiveness of human breast and lung cancer cells as a P2X7R antagonist [145]. Uncaria tomentosa (Willd.) DC. (Rubiaceae). Rubiaceae have excellent anti-oxidant efficiency and are used for a number of therapeutic purposes, including cancer, arthritis and inflammatory diseases [146]. Its hydroalcoholic extract can decrease P2X7R expression in MDA-MB-231 cells; tumor invasion and metastasis may be restrained [147]. The above studies indicated that the active ingredients of TCMs could be a possible tool for cancer treatment by reducing ATP concentration and P2X7R activity in the microenvironment of the tumor.

Active ingredients of TCMs for platelet-related diseases management via inhibiting adenine nucleotides and nucleosides

Platelets activation can result in the expression and/or activation of surface receptors, secretion of vasoactive substances, adhesion, aggregation, and finally thrombus formation [148, 149]. In addition, thromboxane A2 and ADP secreted agonists in turn generate subsequent transmembrane signaling [150]. Adenine nucleotides and nucleosides such as ATP, ADP, and adenosine modulate platelet aggregation [151]. Isorhapontigenin (ISO) isolated from Gnetum cleistostachyum has been shown to exhibit effects of anti-cancer, anti-oxidant, and anti-inflammatory [152154]. It has also been stated that ISO has selective inhibitory effects on platelet activation induced by ADP, likely by binding to P2Y12R [155]. The activity of platelets with ectonucleotidase was shown to exert a hypotensive effect by increasing circulating ATP concentrations [156]. Similarly, by suppressing the hydrolysis of ATP, ADP, and AMP, ginger and turmeric rhizomes may effectively decrease platelet ectonucleotidase and adenosine deaminase activity in hypertensive rats [157]. Scutia buxifolia Reissek extracts led to the depression of NTPDase and CD73 function, defending against platelet aggregation caused by ADP in rats [158, 159]. Similarly, the therapeutic effects of activation of cAMP and cGMP on thrombosis were also observed in SF [160]. The above evidence suggests that by inhibiting the concentration of adenine nucleotides and nucleosides, the active ingredients of TCMs could minimize platelet aggregation. By systematically and deliberately reviewing the literature of active ingredients for purinergic signaling.

Conclusion and future prospects

Growing studies have reported that P2R is closely linked to pain-, inflammation-, cardiovascular disease-, cancer-, and platelet-related diseases in the purinergic signaling pathway [43, 97, 132, 142, 151]. According to the review of the above literature, we found that these above diseases could be notably alleviated by inhibiting the behaviors of P2X3, P2X4, and P2X7. TCMs exert curative effects and lower prices with their own special mechanism, as well as more plentiful natural resources [161163]. However, there are still many concerns about using of TCMs to treat diseases that are related with purinergic signaling. First, we found that toxic reactions caused by purine receptors targeting were rarely investigated. For example, evidence has shown that long-term usage of rhein can cause liver and kidney toxicity [164]. In addition, testing the in vivo metabolic process and tissue distribution of active ingredients is crucial. A strong forum for systematic elucidation of drug absorption, delivery, metabolism, excretion, and toxicity may be the combined metabonomics, serum pharmacochemistry, and tissue-targeted and microfluidic-dependent mass spectrometry imaging [165167]. Meanwhile, studies have shown that some of the active compounds, such as palmatine, are not readily absorbed in the body and have lower bioavailability [168]. To improve the solubility and concentration of drugs in the tissue, biocompatible molecular nanotechnology, and targeted drug technology can be executed. It is surprising that the CRISPR-edited P2XR approach can be used to investigate the in-depth mechanisms of purine disorders in diseases and drug efficacy [169].

Biographies

Xiaopeng Ai

is a doctoral student at Chengdu University of Traditional Chinese Medicine. He received his master’s degree from Traditional Chinese Medicine. His research focused on pharmacodynamics and material basis of traditional Chinese medicine. graphic file with name 11302_2021_9774_Figa_HTML.jpg

Sanyin Zhang

is a Professor in the Innovative Institute of Chinese Medicine and Pharmacy at the Chengdu University of Traditional Chinese Medicine, and a doctor at Chengdu Integrated TCM & Western Medicine Hospital. He received his PhD from Chengdu University of Traditional Chinese Medicine. His research mainly focuses on the mechanisms of Qi and blood generation in TCM involving purine signaling. His discoveries have greatly promoted the prevention and treatment of purine disorders related cardiac-cerebral vascular diseases with traditional Chinese medicine.

Xiaobo Wang

received his doctor’s degree from Chengdu University of TraditionalChinese Medicine. Dr. Wang mainly focused on the relationship between mitochondrialenergy synthesis, apoptosis and cellular mechanisms of hypoxic brain injury.

Authors’ contribution

Sanyin Zhang and Xiaobo Wang conceived the study; Xing Dong, Xiaopeng Ai, Ying Guo, Peng Yang, Ya Hou, and Jinrong Bai collected, analyzed, and interpreted the relevant literatures; Xing Dong and Ya Hou drew all the figures; Xiaopeng Ai, Xing Dong, and Xiaobo Wang wrote the manuscript; Sanyin Zhang and Xiaobo Wang supervised the study and revised the manuscript. The final version of the manuscript was read and approved by all authors.

Funding

This work is supported by the Key R&D project of Provincial Department of Education (18ZA0180), the Science & Technology Department of Sichuan Province (2021YJ0175) and the National Natural Science Foundation of China (82004058).

Data availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Declarations

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Informed consent

Not applicable

Conflict of interest

All authors declare no competing interests.

Footnotes

Publisher’s note

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

Xiaopeng Ai and Xing Dong contributed equally to this work.

Contributor Information

Sanyin Zhang, Email: tcmzsy@cdutcm.edu.cn.

Xiaobo Wang, Email: VitaDrwang@cdutcm.edu.cn.

References

  • 1.Zhang WJ, Hu CG, Zhu ZM, Luo HL. Effect of P2X7 receptor on tumorigenesis and its pharmacological properties. Biomed Pharmacother. 2020;125:109844. doi: 10.1016/j.biopha.2020.109844. [DOI] [PubMed] [Google Scholar]
  • 2.Di Virgilio F, Sarti AC, Coutinho-Silva R. Purinergic signaling, DAMPs, and inflammation. Am J Physiol Cell Physiol. 2020;318(5):C832–C835. doi: 10.1152/ajpcell.00053.2020. [DOI] [PubMed] [Google Scholar]
  • 3.Li Q, Zhu X, Song W, Peng X, Zhao R. The P2X7 purinergic receptor: a potential therapeutic target for lung cancer. J Cancer Res Clin Oncol. 2020;146(11):2731–2741. doi: 10.1007/s00432-020-03379-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Shen JB, Toti KS, Chakraborty S, Kumar TS, Cronin C, Liang BT, Jacobson KA. Prevention and rescue of cardiac dysfunction by methanocarba adenosine monophosphonate derivatives. Purinergic Signal. 2020;16(1):61–72. doi: 10.1007/s11302-020-09688-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Boison D, Jarvis MF (2020) Adenosine kinase: a key regulator of purinergic physiology. Biochem Pharmacol 114321 [DOI] [PMC free article] [PubMed]
  • 6.Bragança B, Correia-de-Sá P. Resolving the ionotropic P2X4 receptor mystery points towards a new therapeutic target for cardiovascular diseases. Int J Mol Sci. 2020;21(14):5005. doi: 10.3390/ijms21145005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.He JR, Yu SG, Tang Y, Illes P. Purinergic signaling as a basis of acupuncture-induced analgesia. Purinergic Signal. 2020;16(3):297–304. doi: 10.1007/s11302-020-09708-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Dwyer KM, Kishore BK, Robson SC. Conversion of extracellular ATP into adenosine: a master switch in renal health and disease. Nat Rev Nephrol. 2020;16(9):509–524. doi: 10.1038/s41581-020-0304-7. [DOI] [PubMed] [Google Scholar]
  • 9.Sinha P, Paswan RK, Kumari A, Kumar S, Bimal S, Das P, Lal CS. Magnesium-dependent ecto-ATP diphosphohydrolase activity in Leishmania donovani. Curr Microbiol. 2016;73(6):811–819. doi: 10.1007/s00284-016-1130-9. [DOI] [PubMed] [Google Scholar]
  • 10.da Silva W, da Rocha TN, de Melo AJ, da Silva VHF, de Souza ACA, Ribeiro IC, de Oliveira TA, de Souza LA, Andrade LKR, de Moraes JVB, Diogo MA, de Castro RB, Polêto MD, Afonso LCC, Fietto JLR. ENTPDases from pathogenic trypanosomatids and purinergic signaling: shedding light towards biotechnological applications. Curr Top Med Chem. 2020;21:213–226. doi: 10.2174/1568026620666201005125146. [DOI] [PubMed] [Google Scholar]
  • 11.Yegutkin GG (2020) Adenosine metabolism in the vascular system. Biochem Pharmacol 114373 [DOI] [PubMed]
  • 12.Zhang CJ, Zhu N, Long J, Wu HT, Wang YX, Liu BY, Liao DF, Qin L (2020) Celastrol induces lipophagy via the LXRα/ABCA1 pathway in clear cell renal cell carcinoma. Acta Pharmacol Sin. 10.1038/s41401-020-00572-6 [DOI] [PMC free article] [PubMed]
  • 13.Aksentijević D, Zervou S, Eykyn TR, McAndrew DJ, Wallis J, Schneider JE, Neubauer S, Lygate CA. Age-dependent decline in cardiac function in guanidinoacetate-n-methyltransferase knockout mice. Front Physiol. 2020;10:1535. doi: 10.3389/fphys.2019.01535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Le TT, Berg NK, Harting MT, Li X, Eltzschig HK, Yuan X. Purinergic signaling in pulmonary inflammation. Front Immunol. 2019;10:1633. doi: 10.3389/fimmu.2019.01633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Horioka M, Ceraudo E, Lorenzen E, Sakmar TP, Huber T (2020) Purinergic receptors crosstalk with CCR5 to amplify Ca2+ signaling. Cell Mol Neurobiol. 10.1007/s10571-020-01002-1 [DOI] [PMC free article] [PubMed]
  • 16.Szrejder M, Rachubik P, Rogacka D, Audzeyenka I, Rychłowski M, Angielski S, Piwkowska A. Extracellular ATP modulates podocyte function through P2Y purinergic receptors and pleiotropic effects on AMPK and cAMP/PKA signaling pathways. Arch Biochem Biophys. 2020;695:108649. doi: 10.1016/j.abb.2020.108649. [DOI] [PubMed] [Google Scholar]
  • 17.Sattler C, Eick T, Hummert S, Schulz E, Schmauder R, Schweinitz A, Unzeitig C, Schwede F, Benndorf K. Unravelling the intricate cooperativity of subunit gating in P2X2 ion channels. Sci Rep. 2020;10(1):21751. doi: 10.1038/s41598-020-78672-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hørlyck S, Cai C, Helms HC, Lauritzen M, Brodin B. ATP induces contraction of cultured brain capillary pericytes, via activation of P2Y type purinergic receptors. Am J Physiol Heart Circ Physiol. 2020;320:H699–H712. doi: 10.1152/ajpheart.00560.2020. [DOI] [PubMed] [Google Scholar]
  • 19.Mahaut Smith MP, Evans RJ, Vial C. Development of a P2X1-eYFP receptor knock-in mouse to track receptors in real time. Purinergic Signal. 2019;15(3):397–402. doi: 10.1007/s11302-019-09666-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Garcez ML, Damiani AP, Pacheco R, Rodrigues L, de Abreu LL, Alves MC, de Andrade VM, Boeck CR. Caffeine neuroprotection decreases a2a adenosine receptor content in aged mice. Neurochem Res. 2019;44(4):787–795. doi: 10.1007/s11064-018-02710-3. [DOI] [PubMed] [Google Scholar]
  • 21.Dănilă MD, Piollet M, Aburel OM, Angoulvant D, Lefort C, Chadet S, Roger S, Muntean MD, Ivanes F. Modulation of P2Y11-related purinergic signaling in inflammation and cardio-metabolic diseases. Eur J Pharmacol. 2020;876:173060. doi: 10.1016/j.ejphar.2020.173060. [DOI] [PubMed] [Google Scholar]
  • 22.Andrejew R, Oliveira-Giacomelli Á, Ribeiro DE, Glaser T, Arnaud-Sampaio VF, Lameu C, Ulrich H. The P2X7 receptor: Central hub of brain diseases. Front Mol Neurosci. 2020;13:124. doi: 10.3389/fnmol.2020.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.den Ouden JE, Zaman GJR, Dylus J, van Doornmalen AM, Mulder WR, Grobben Y, van Riel WE, de Hullu JA, Buijsman RC, van Altena AM. Chemotherapy sensitivity testing on ovarian cancer cells isolated from malignant ascites. Oncotarget. 2020;11(49):4570–4581. doi: 10.18632/oncotarget.27827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schneider R, Leven P, Glowka T, Kuzmanov I, Lysson M, Schneiker B, Miesen A, Baqi Y, Spanier C, Grants I, Mazzotta E, Villalobos-Hernandez E, Kalff JC, Müller CE, Christofi FL, Wehner S (2020) A novel P2X2-dependent purinergic mechanism of enteric gliosis in intestinal inflammation. EMBO Mol Med:e12724 [DOI] [PMC free article] [PubMed]
  • 25.Krajewski JL. P2X3-containing receptors as targets for the treatment of chronic pain. Neurotherapeutics. 2020;17(3):826–838. doi: 10.1007/s13311-020-00934-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Albulushi A, Olson J, Xie F, Qian L, Mathers D, Aboeata A, Porter T. Effect of intermittent high mechanical index impulses on left ventricular strain. J Am Soc Echocardiogr. 2020;S0894-7317(20):30768–30769. doi: 10.1016/j.echo.2020.11.013. [DOI] [PubMed] [Google Scholar]
  • 27.Li JJ, Liu ZX, Zhang YL, Xue GY. P2X receptors and trigeminal neuralgia. Neuroreport. 2019;30(10):725–729. doi: 10.1097/WNR.0000000000001266. [DOI] [PubMed] [Google Scholar]
  • 28.Liu YH, Chang YC, Chen LK, Su PA, Ko WC, Tsai YS, Chen YH, Lai HC, Wu CY, Hung YP, Tsai PJ. The ATP-P2X7 signaling axis is an essential sentinel for intracellular clostridium difficile pathogen-induced inflammasome activation. Front Cell Infect Microbiol. 2018;8:84. doi: 10.3389/fcimb.2018.00084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Liu X, Gao ZG, Wu Y, Stevens RC, Jacobson KA, Zhao S. Salvianolic acids from antithrombotic traditional Chinese medicine Danshen are antagonists of human P2Y1 and P2Y12 receptors. Sci Rep. 2018;8(1):8084. doi: 10.1038/s41598-018-26577-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wu H, Gao S, Fu M, Sakurai T, Terakawa S. Fucoidan inhibits Ca2+ responses induced by a wide spectrum of agonists for G protein coupled receptors. Mol Med Rep. 2018;17(1):1428–1436. doi: 10.3892/mmr.2017.8035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Li SH, Li L, Yang RN, Liang SD. Compounds of traditional Chinese medicine and neuropathic pain. Chin J Nat Med. 2020;18(1):28–35. doi: 10.1016/S1875-5364(20)30002-9. [DOI] [PubMed] [Google Scholar]
  • 32.Zou L, Gong Y, Liu S, Liang S. Natural compounds acting at P2 receptors alleviate peripheral neuropathy. Brain Res Bull. 2019;151:125–131. doi: 10.1016/j.brainresbull.2018.12.017. [DOI] [PubMed] [Google Scholar]
  • 33.Dinakar P, Stillman AM. Pathogenesis of pain. Semin Pediatr Neurol. 2016;23(3):201–208. doi: 10.1016/j.spen.2016.10.003. [DOI] [PubMed] [Google Scholar]
  • 34.Wang VC, Mullally WJ. Pain neurology. Am J Med. 2020;133(3):273–280. doi: 10.1016/j.amjmed.2019.07.029. [DOI] [PubMed] [Google Scholar]
  • 35.Qaseem A, Wilt TJ, McLean RM, Forciea MA. Clinical guidelines committee of the American college of physicians. Noninvasive treatments for acute, subacute, and chronic low back pain: a clinical practice guideline from the American college of physicians. Ann Intern Med. 2017;166(7):514–530. doi: 10.7326/M16-2367. [DOI] [PubMed] [Google Scholar]
  • 36.Celentano V, Pellino G, Rottoli M, Colombo F, Sampietro G, Spinelli A, Selvaggi F (2020) SICCR Current status of Crohn’s disease surgery collaborative. Single-incision laparoscopic surgery (SILS) for the treatment of ileocolonic Crohn’s disease: a propensity score-matched analysis. Int J Colorectal Dis. 10.1007/s00384-020-03821-6 [DOI] [PMC free article] [PubMed]
  • 37.Longo R, Oudshoorn A, Befus D. Cannabis for chronic pain: a rapid systematic review of randomized control trials. Pain Manag Nurs. 2020;S1524-9042(20):30225–30223. doi: 10.1016/j.pmn.2020.11.006. [DOI] [PubMed] [Google Scholar]
  • 38.Kuan YH, Shyu BC. Nociceptive transmission and modulation via P2X receptors in central pain syndrome. Mol Brain. 2016;9(1):58. doi: 10.1186/s13041-016-0240-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pui Ping C, Akhtar MN, Israf DA, Perimal EK, Sulaiman MR. Possible participation of ionotropic glutamate receptors and l-arginine-nitric oxide-cyclic guanosine monophosphate-ATP-sensitive K+ channel pathway in the antinociceptive activity of cardamonin in acute pain animal models. Molecules. 2020;25(22):5385. doi: 10.3390/molecules25225385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.He Y, Makarczyk MJ, Lin H. Role of mitochondria in mediating chondrocyte response to mechanical stimuli. Life Sci. 2020;263:118602. doi: 10.1016/j.lfs.2020.118602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ding S, Yu Q, Wang J, Zhu L, Li T, Guo X, Zhang X. Activation of ATF3/AP-1 signaling pathway is required for P2X3-induced endometriosis pain. Hum Reprod. 2020;35(5):1130–1144. doi: 10.1093/humrep/deaa061. [DOI] [PubMed] [Google Scholar]
  • 42.Arribas-Blázquez M, Olivos-Oré LA, Barahona MV, Sánchez de la Muela M, Solar V, Jiménez E, Gualix J, McIntosh JM, Ferrer-Montiel A, Miras-Portugal MT, Artalejo AR. Overexpression of P2X3 and P2X7 receptors and TRPV1 channels in adrenomedullary chromaffin cells in a rat model of neuropathic pain. Int J Mol Sci. 2019;20(1):155. doi: 10.3390/ijms20010155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Khoja S, Shah V, Garcia D, Asatryan L, Jakowec MW, Davies DL. Role of purinergic P2X4 receptors in regulating striatal dopamine homeostasis and dependent behaviors. J Neurochem. 2016;139(1):134–148. doi: 10.1111/jnc.13734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Fu J, Zhu X, Wang W, Lu H, Zhang Z, Liu T, Xu H, Fu H, Ma S, Luo Y. 1, 6-di-O-caffeoyl-β-D-glucopyranoside, a natural compound from Callicarpa nudiflora Hook impairs P2Y12 and thromboxane A2 receptor-mediated amplification of platelet activation and aggregation. Phytomedicine. 2017;36:273–282. doi: 10.1016/j.phymed.2017.10.012. [DOI] [PubMed] [Google Scholar]
  • 45.Zhang M, Gao F, Teng F, Zhang C. Tetramethylpyrazine promotes the proliferation and migration of brain endothelial cells. Mol Med Rep. 2014;10(1):29–32. doi: 10.3892/mmr.2014.2169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yan S, Chen L, Wei X, Cheng L, Kong L, Liu X, Zhang X, Liu H. Tetramethylpyrazine analogue CXC195 ameliorates cerebral ischemia-reperfusion injury by regulating endothelial nitric oxide synthase phosphorylation via PI3K/Akt signaling. Neurochem Res. 2015;40(3):446–454. doi: 10.1007/s11064-014-1485-x. [DOI] [PubMed] [Google Scholar]
  • 47.Chen HY, Xu DP, Tan GL, Cai W, Zhang GX, Cui W, Wang JZ, Long C, Sun YW, Yu P, Tsim KW, Zhang ZJ, Han YF, Wang YQ. A potent multi-functional neuroprotective derivative of tetramethylpyrazine. J Mol Neurosci. 2015;56(4):977–987. doi: 10.1007/s12031-015-0566-x. [DOI] [PubMed] [Google Scholar]
  • 48.Liang SD, Gao Y, Xu CS, Xu BH, Mu SN. Effect of tetramethylpyrazine on acute nociception mediated by signaling of P2X receptor activation in rat. Brain Res. 2004;995(2):247–252. doi: 10.1016/j.brainres.2003.09.070. [DOI] [PubMed] [Google Scholar]
  • 49.Gao Y, Xu C, Liang S, Zhang A, Mu S, Wang Y, Wan F. Effect of tetramethylpyrazine on primary afferent transmission mediated by P2X3 receptor in neuropathic pain states. Brain Res Bull. 2008;77(1):27–32. doi: 10.1016/j.brainresbull.2008.02.026. [DOI] [PubMed] [Google Scholar]
  • 50.Leng YF, Gao XM, Wang SX, Xing YH. Effects of tetramethylpyrazine on neuronal apoptosis in the superficial dorsal horn in a rat model of neuropathic pain. Am J Chin Med. 2012;40(6):1229–1239. doi: 10.1142/S0192415X12500917. [DOI] [PubMed] [Google Scholar]
  • 51.Gao Y, Xu C, Yu K, Li G, Wan F, Liu S, Lin J, Liu H, Zhang J, Li X, Liang S. Effect of tetramethylpyrazine on DRG neuron P2X3 receptor involved in transmitting pain after burn. Burns. 2010;36(1):127–134. doi: 10.1016/j.burns.2009.04.032. [DOI] [PubMed] [Google Scholar]
  • 52.Xiong W, Tan M, He L, Ou X, Jin Y, Yang G, Huang L, Shen Y, Guan S, Xu C, Li G, Liu S, Xu H, Liang S, Gao Y. Inhibitory effects of tetramethylpyrazine on pain transmission of trigeminal neuralgia in CCI-ION rats. Brain Res Bull. 2017;134:72–78. doi: 10.1016/j.brainresbull.2017.07.005. [DOI] [PubMed] [Google Scholar]
  • 53.Zhang A, Xu C, Liang S, Gao Y, Li G, Wei J, Wan F, Liu S, Lin J. Role of sodium ferulate in the nociceptive sensory facilitation of neuropathic pain injury mediated by P2X(3) receptor. Neurochem Int. 2008;53(6-8):278–282. doi: 10.1016/j.neuint.2008.08.008. [DOI] [PubMed] [Google Scholar]
  • 54.Zhang A, Gao Y, Zhong X, Xu C, Li G, Liu S, Lin J, Li X, Zhang Y, Liu H, Linag S. Effect of sodium ferulate on the hyperalgesia mediated by P2X3 receptor in the neuropathic pain rats. Brain Res. 2010;1313:215–221. doi: 10.1016/j.brainres.2009.11.067. [DOI] [PubMed] [Google Scholar]
  • 55.Tu WZ, Cheng RD, Cheng B, Lu J, Cao F, Lin HY, Jiang YX, Wang JZ, Chen H, Jiang SH. Analgesic effect of electroacupuncture on chronic neuropathic pain mediated by P2X3 receptors in rat dorsal root ganglion neurons. Neurochem Int. 2012;60(4):379–386. doi: 10.1016/j.neuint.2012.01.006. [DOI] [PubMed] [Google Scholar]
  • 56.Ou S, Zhao YD, Xiao Z, Wen HZ, Cui J, Ruan HZ. Effect of lappaconitine on neuropathic pain mediated by P2X3 receptor in rat dorsal root ganglion. Neurochem Int. 2011;58(5):564–573. doi: 10.1016/j.neuint.2011.01.016. [DOI] [PubMed] [Google Scholar]
  • 57.Tao J, Liu L, Fan Y, Wang M, Li L, Zou L, Yuan H, Shi L, Yang R, Liang S, Liu S. Role of hesperidin in P2X3 receptor-mediated neuropathic pain in the dorsal root ganglia. Int J Neurosci. 2019;129(8):784–793. doi: 10.1080/00207454.2019.1567512. [DOI] [PubMed] [Google Scholar]
  • 58.Gao T, Hao J, Wiesenfeld-Hallin Z, Wang DQ, Xu XJ. Analgesic effect of sinomenine in rodents after inflammation and nerve injury. Eur J Pharmacol. 2013;721(1-3):5–11. doi: 10.1016/j.ejphar.2013.09.062. [DOI] [PubMed] [Google Scholar]
  • 59.Gao T, Shi T, Wang DQ, Wiesenfeld-Hallin Z, Xu XJ. Repeated sinomenine administration alleviates chronic neuropathic pain-like behaviours in rodents without producing tolerance. Scand J Pain. 2014;5(4):249–255. doi: 10.1016/j.sjpain.2014.05.006. [DOI] [PubMed] [Google Scholar]
  • 60.Rao S, Liu S, Zou L, Jia T, Zhao S, Wu B, Yi Z, Wang S, Xue Y, Gao Y, Xu C, Li G, Xu H, Zhang C, Liang S. The effect of sinomenine in diabetic neuropathic pain mediated by the P2X3 receptor in dorsal root ganglia. Purinergic Signal. 2017;13(2):227–235. doi: 10.1007/s11302-016-9554-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cai ZM, Peng JQ, Chen Y, Tao L, Zhang YY, Fu LY, Long QD, Shen XC. 1,8-Cineole: a review of source, biological activities, and application. J Asian Nat Prod Res. 2020;28:1–17. doi: 10.1080/10286020.2020.1839432. [DOI] [PubMed] [Google Scholar]
  • 62.Zhang YL, Liu YG, Li Q, Wang XD, Zheng XB, Yang BL, Wan B, Ma JM, Liu ZX. 1,8-cineole decreases neuropathic pain probably via a mechanism mediating P2X3 receptor in the dorsal root ganglion. Neurochem Int. 2018;121:69–74. doi: 10.1016/j.neuint.2018.09.007. [DOI] [PubMed] [Google Scholar]
  • 63.Chan TC, Chang CJ, Koonchanok NM, Geahlen RL. Selective inhibition of the growth of ras-transformed human bronchial epithelial cells by emodin, a protein-tyrosine kinase inhibitor. Biochem Biophys Res Commun. 1993;193(3):1152–1158. doi: 10.1006/bbrc.1993.1746. [DOI] [PubMed] [Google Scholar]
  • 64.Chen HC, Hsieh WT, Chang WC, Chung JG. Aloe-emodin induced in vitro G2/M arrest of cell cycle in human promyelocytic leukemia HL-60 cells. Food Chem Toxicol. 2004;42(8):1251–1257. doi: 10.1016/j.fct.2004.03.002. [DOI] [PubMed] [Google Scholar]
  • 65.Li HL, Chen HL, Li H, Zhang KL, Chen XY, Wang XW, Kong QY, Liu J. Regulatory effects of emodin on NF-kappaB activation and inflammatory cytokine expression in RAW 264.7 macrophages. Int J Mol Med. 2005;16(1):41–47. [PubMed] [Google Scholar]
  • 66.Wang C, Zhang D, Ma H, Liu J. Neuroprotective effects of emodin-8-O-beta-D-glucoside in vivo and in vitro. Eur J Pharmacol. 2007;577(1-3):58–63. doi: 10.1016/j.ejphar.2007.08.033. [DOI] [PubMed] [Google Scholar]
  • 67.Wu Y, Tu X, Lin G, Xia H, Huang H, Wan J, Cheng Z, Liu M, Chen G, Zhang H, Fu J, Liu Q, Liu DX. Emodin-mediated protection from acute myocardial infarction via inhibition of inflammation and apoptosis in local ischemic myocardium. Life Sci. 2007;1(17-18):1332–1338. doi: 10.1016/j.lfs.2007.08.040. [DOI] [PubMed] [Google Scholar]
  • 68.Gao Y, Liu H, Deng L, Zhu G, Xu C, Li G, Liu S, Xie J, Liu J, Kong F, Wu R, Li G, Liang S. Effect of emodin on neuropathic pain transmission mediated by P2X2/3 receptor of primary sensory neurons. Brain Res Bull. 2011;84(6):406–413. doi: 10.1016/j.brainresbull.2011.01.017. [DOI] [PubMed] [Google Scholar]
  • 69.Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol. 2009;41(1):40–59. doi: 10.1016/j.biocel.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhao S, Yang J, Han X, Gong Y, Rao S, Wu B, Yi Z, Zou L, Jia T, Li L, Yuan H, Shi L, Zhang C, Gao Y, Li G, Liu S, Xu H, Liu H, Liang S. Effects of nanoparticle-encapsulated curcumin on HIV-gp120-associated neuropathic pain induced by the P2X3 receptor in dorsal root ganglia. Brain Res Bull. 2017;135:53–61. doi: 10.1016/j.brainresbull.2017.09.011. [DOI] [PubMed] [Google Scholar]
  • 71.Jia T, Rao J, Zou L, Zhao S, Yi Z, Wu B, Li L, Yuan H, Shi L, Zhang C, Gao Y, Liu S, Xu H, Liu H, Liang S, Li G. Nanoparticle-encapsulated curcumin inhibits diabetic neuropathic pain involving the P2Y12 receptor in the dorsal root ganglia. Front Neurosci. 2018;11:755. doi: 10.3389/fnins.2017.00755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yu M, Su B, Zhang X. Gardenoside suppresses the pain in rats model of chronic constriction injury by regulating the P2X3 and P2X7 receptors. J Recept Signal Transduct Res. 2018;38(3):198–203. doi: 10.1080/10799893.2018.1468782. [DOI] [PubMed] [Google Scholar]
  • 73.Pan B, Fang S, Zhang J, Pan Y, Liu H, Wang Y, Li M, Liu L. Chinese herbal compounds against SARS-CoV-2: puerarin and quercetin impair the binding of viral S-protein to ACE2 receptor. Comput Struct Biotechnol J. 2020;18:3518–3527. doi: 10.1016/j.csbj.2020.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yeung DK, Leung SW, Xu YC, Vanhoutte PM, Man RY. Puerarin, an isoflavonoid derived from Radix puerariae, potentiates endothelium-independent relaxation via the cyclic AMP pathway in porcine coronary artery. Eur J Pharmacol. 2006;552(1-3):105–111. doi: 10.1016/j.ejphar.2006.08.078. [DOI] [PubMed] [Google Scholar]
  • 75.Gao Q, Yang B, Ye ZG, Wang J, Bruce IC, Xia Q. Opening the calcium-activated potassium channel participates in the cardioprotective effect of puerarin. Eur J Pharmacol. 2007;574(2-3):179–184. doi: 10.1016/j.ejphar.2007.07.018. [DOI] [PubMed] [Google Scholar]
  • 76.Liu S, Zhang C, Shi Q, Li G, Song M, Gao Y, Xu C, Xu H, Fan B, Yu S, Zheng C, Zhu Q, Wu B, Peng L, Xiong H, Wu Q, Liang S. Puerarin blocks the signaling transmission mediated by P2X3 in SG and DRG to relieve myocardial ischemic damage. Brain Res Bull. 2014;101:57–63. doi: 10.1016/j.brainresbull.2014.01.001. [DOI] [PubMed] [Google Scholar]
  • 77.Liu S, Wang M, Wang N, Li S, Sun R, Xing J, Wang Y, Yu S, Li L, Li G, Liang S. Exploring the molecular mechanism of the effect of puerarin on P2X3. Int J Biol Macromol. 2020;142:484–491. doi: 10.1016/j.ijbiomac.2019.09.120. [DOI] [PubMed] [Google Scholar]
  • 78.Li X, Zhang J, Gao Y, Yang Y, Xu C, Li G, Guo G, Liu S, Xie J, Liang S. Puerarin alleviates burn-related procedural pain mediated by P2X(3) receptors. Purinergic Signal. 2011;7(4):489–497. doi: 10.1007/s11302-011-9248-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhang J, Li X, Gao Y, Guo G, Xu C, Li G, Liu S, Huang A, Tu G, Peng H, Qiu S, Fan B, Zhu Q, Yu S, Zheng C, Liang S. Effects of puerarin on the inflammatory role of burn-related procedural pain mediated by P2X(7) receptors. Burns. 2013;39(4):610–618. doi: 10.1016/j.burns.2012.08.013. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang Y, Zheng L, Zheng Y, Zhou C, Huang P, Xiao X, Zhao Y, Hao X, Hu Z, Chen Q, Li H, Wang X, Fukushima K, Wang G, Li C. Assembly and annotation of a draft genome of the medicinal plant polygonum cuspidatum. Front Plant Sci. 2019;10:1274. doi: 10.3389/fpls.2019.01274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cui Y, Li Y, Ning J, Mi Y, Wang X, Qiu Z, Li L, Gou X. Resveratrol alleviates diabetic mechanical allodynia in rats by downregulating P2X3R. Mol Med Rep. 2020;22(2):957–963. doi: 10.3892/mmr.2020.11157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Guo J, Wang C, Niu X, Zhou F, Li H, Gao W (2019) Effects of resveratrol in the signaling of neuropathic pain involving P2X3 in the dorsal root ganglion of rats. Acta Neurol Belg. 10.1007/s13760-019-01126-2 [DOI] [PubMed]
  • 83.Xie J, Liu S, Wu B, Li G, Rao S, Zou L, Yi Z, Zhang C, Jia T, Zhao S, Schmalzing G, Hausmann R, Nie H, Li G, Liang S. The protective effect of resveratrol in the transmission of neuropathic pain mediated by the P2X7 receptor in the dorsal root ganglia. Neurochem Int. 2017;103:24–35. doi: 10.1016/j.neuint.2016.12.006. [DOI] [PubMed] [Google Scholar]
  • 84.Wu B, Ma Y, Yi Z, Liu S, Rao S, Zou L, Wang S, Xue Y, Jia T, Zhao S, Shi L, Li L, Yuan H, Liang S. Resveratrol-decreased hyperalgesia mediated by the P2X7 receptor in gp120-treated rats. Mol Pain. 2017;13:1744806917707667. doi: 10.1177/1744806917707667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang M, Cai X, Wang Y, Li S, Wang N, Sun R, Xing J, Liang S, Liu S. Astragalin alleviates neuropathic pain by suppressing P2X4-mediated signaling in the dorsal root ganglia of rats. Front Neurosci. 2021;14:570831. doi: 10.3389/fnins.2020.570831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Yu Y, Zu C, He D, Li Y, Chen Q, Chen Q, Wang H, Wang R, Chaurasiya B, Zaro JL, Wang Y, Tu J, Sun C. pH-dependent reversibly activatable cell-penetrating peptides improve the antitumor effect of artemisinin-loaded liposomes. J Colloid Interface Sci. 2020;S0021-9797(20):31443. doi: 10.1016/j.jcis.2020.10.103. [DOI] [PubMed] [Google Scholar]
  • 87.Huang TE, Deng YN, Hsu JL, Leu WJ, Marchesi E, Capobianco ML, Marchetti P, Navacchia ML, Guh JH, Perrone D, Hsu LC. Evaluation of the anticancer activity of a bile acid-dihydroartemisinin hybrid ursodeoxycholic-dihydroartemisinin in hepatocellular carcinoma cells. Front Pharmacol. 2020;11:599067. doi: 10.3389/fphar.2020.599067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ying M, Liu H, Zhang T, Jiang C, Gong Y, Wu B, Zou L, Yi Z, Rao S, Li G, Zhang C, Jia T, Zhao S, Yuan H, Shi L, Li L, Liang S, Liu S. Effect of artemisinin on neuropathic pain mediated by P2X4 receptor in dorsal root ganglia. Neurochem Int. 2017;108:27–33. doi: 10.1016/j.neuint.2017.02.004. [DOI] [PubMed] [Google Scholar]
  • 89.Zhang L, Wu Y, Yang G, Gan H, Sang D, Zhou J, Su L, Wang R, Ma L. Design, synthesis and biological evaluation of novel osthole-based derivatives as potential neuroprotective agents. Bioorg Med Chem Lett. 2020;30(24):127633. doi: 10.1016/j.bmcl.2020.127633. [DOI] [PubMed] [Google Scholar]
  • 90.Yuan H, Ouyang S, Yang R, Li S, Gong Y, Zou L, Jia T, Zhao S, Wu B, Yi Z, Liu H, Shi L, Li L, Gao Y, Li G, Xu H, Liu S, Zhang C, Liang S. Osthole alleviated diabetic neuropathic pain mediated by the P2X4 receptor in dorsal root ganglia. Brain Res Bull. 2018;142:289–296. doi: 10.1016/j.brainresbull.2018.08.008. [DOI] [PubMed] [Google Scholar]
  • 91.Su X, Wu B, Zhang W, Ji YH, Wang Q, Tan ZY. Inhibitory effects of columbianadin on nociceptive behaviors in a neuropathic pain model, and on voltage-gated calcium currents in dorsal root ganglion neurons in mice. Front Pharmacol. 2020;10:1522. doi: 10.3389/fphar.2019.01522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yang B, Maddison LA, Zaborska KE, Dai C, Yin L, Tang Z, Zang L, Jacobson DA, Powers AC, Chen W. RIPK3-mediated inflammation is a conserved β cell response to ER stress. Sci Adv. 2020;6(51):eabd7272. doi: 10.1126/sciadv.abd7272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kuprash DV, Nedospasov SA. Molecular and cellular mechanisms of inflammation. Biochemistry (Mosc) 2016;81(11):1237–1239. doi: 10.1134/S0006297916110018. [DOI] [PubMed] [Google Scholar]
  • 94.Foley JF. Focus issue: Inflammatory mechanisms. Sci Signal. 2015;8(366):eg2. doi: 10.1126/scisignal.aaa9599. [DOI] [PubMed] [Google Scholar]
  • 95.Mazzotta E, Villalobos-Hernandez E, Kalff JC, Müller CE, Christofi FL, Wehner S (2020) A novel P2X2-dependent purinergic mechanism of enteric gliosis in intestinal inflammation. EMBO Mol Med:e12724 [DOI] [PMC free article] [PubMed]
  • 96.Burnstock G, Knight GE. The potential of P2X7 receptors as a therapeutic target, including inflammation and tumour progression. Purinergic Signal. 2018;14(1):1–18. doi: 10.1007/s11302-017-9593-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Demeules M, Scarpitta A, Abad C, Gondé H, Hardet R, Pinto-Espinoza C, Eichhoff AM, Schäfer W, Haag F, Koch-Nolte F, Adriouch S. Evaluation of P2X7 receptor function in tumor contexts using raav vector and nanobodies (AAVnano) Front Oncol. 2020;10:1699. doi: 10.3389/fonc.2020.01699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Coutinho-Silva R, Ojcius DM, Górecki DC, Persechini PM, Bisaggio RC, Mendes AN, Marks J, Burnstock G, Dunn PM. Multiple P2X and P2Y receptor subtypes in mouse J774, spleen and peritoneal macrophages. Biochem Pharmacol. 2005;69(4):641–655. doi: 10.1016/j.bcp.2004.11.012. [DOI] [PubMed] [Google Scholar]
  • 99.Lee HW, Kim J, Yim JH, Lee HK, Pyo S. Anti-inflammatory activity of lobaric acid via suppressing NF-κB/MAPK pathways or NLRP3 inflammasome activation. Planta Med. 2019;85(4):302–311. doi: 10.1055/a-0777-2420. [DOI] [PubMed] [Google Scholar]
  • 100.Hamza AA, Heeba GH, Hamza S, Abdalla A, Amin A. Standardized extract of ginger ameliorates liver cancer by reducing proliferation and inducing apoptosis through inhibition oxidative stress/ inflammation pathway. Biomed Pharmacother. 2020;134:111102. doi: 10.1016/j.biopha.2020.111102. [DOI] [PubMed] [Google Scholar]
  • 101.Akinyemi AJ, Thomé GR, Morsch VM, Bottari NB, Baldissarelli J, de Oliveira LS, Goularte JF, Belló-Klein A, Duarte T, Duarte M, Boligon AA, Athayde ML, Akindahunsi AA, Oboh G, Schetinger MR. Effect of ginger and turmeric rhizomes on inflammatory cytokines levels and enzyme activities of cholinergic and purinergic systems in hypertensive rats. Planta Med. 2016;82(7):612–620. doi: 10.1055/s-0042-102062. [DOI] [PubMed] [Google Scholar]
  • 102.Rasool ST, Alavala RR, Kulandaivelu U, Sreeharsha N. Non-invasive delivery of nano-emulsified sesame oil-extract of turmeric attenuates lung inflammation. Pharmaceutics. 2020;12(12):1206. doi: 10.3390/pharmaceutics12121206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang B, Wan J, Gong X, Kuang G, Cheng X, Min S. Mangiferin attenuates renal ischemia-reperfusion injury by inhibiting inflammation and inducing adenosine production. Int Immunopharmacol. 2015;25(1):148–154. doi: 10.1016/j.intimp.2014.11.011. [DOI] [PubMed] [Google Scholar]
  • 104.Zhang Q, Hu F, Guo F, Zhou Q, Xiang H, Shang D (2019) Emodin attenuates adenosine triphosphate-induced pancreatic ductal cell injury in vitro via the inhibition of the P2X7/NLRP3 signaling pathway. Oncol Rep. 10.3892/or.2019.7270 [DOI] [PubMed]
  • 105.Zhu S, Wang Y, Wang X, Li J, Hu F. Emodin inhibits ATP-induced IL-1β secretion, ROS production and phagocytosis attenuation in rat peritoneal macrophages via antagonizing P2X7 receptor. Pharm Biol. 2014;52(1):51–57. doi: 10.3109/13880209.2013.810648. [DOI] [PubMed] [Google Scholar]
  • 106.Liu L, Zou J, Liu X, Jiang LH, Li J. Inhibition of ATP-induced macrophage death by emodin via antagonizing P2X7 receptor. Eur J Pharmacol. 2010;640(1-3):15–19. doi: 10.1016/j.ejphar.2010.04.036. [DOI] [PubMed] [Google Scholar]
  • 107.Zhou YX, Xia W, Yue W, Peng C, Rahman K, Zhang H (2015) Rhein: A review of pharmacological activities. Evid Based Complement Alternat Med:578107 [DOI] [PMC free article] [PubMed]
  • 108.Wu C, Cao H, Zhou H, Sun L, Xue J, Li J, Bian Y, Sun R, Dong S, Liu P, Sun M. Research progress on the antitumor effects of rhein: literature review. Anticancer Agents Med Chem. 2017;17(12):1624–1632. doi: 10.2174/1871520615666150930112631. [DOI] [PubMed] [Google Scholar]
  • 109.He ZH, Zhou R, He MF, Lau CB, Yue GG, Ge W, But PP. Anti-angiogenic effect and mechanism of rhein from Rhizoma Rhei. Phytomedicine. 2011;18(6):470–478. doi: 10.1016/j.phymed.2010.10.006. [DOI] [PubMed] [Google Scholar]
  • 110.Hu F, Zhu D, Pei W, Lee I, Zhang X, Pan L, Xu J. Rhein inhibits ATP-triggered inflammatory responses in rheumatoid rat fibroblast-like synoviocytes. Int Immunopharmacol. 2019;75:105780. doi: 10.1016/j.intimp.2019.105780. [DOI] [PubMed] [Google Scholar]
  • 111.Hu F, Xing F, Zhu G, Xu G, Li C, Qu J, Lee I, Pan L. Rhein antagonizes P2X7 receptor in rat peritoneal macrophages. Sci Rep. 2015;5:14012. doi: 10.1038/srep14012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chtourou Y, Aouey B, Kebieche M, Fetoui H. Protective role of naringin against cisplatin induced oxidative stress, inflammatory response and apoptosis in rat striatum via suppressing ROS-mediated NF-κB and P53 signaling pathways. Chem Biol Interact. 2015;239:76–86. doi: 10.1016/j.cbi.2015.06.036. [DOI] [PubMed] [Google Scholar]
  • 113.Chen Q, Wu H, Tao J, Liu C, Deng Z, Liu Y, Chen G, Liu B, Xu C. Effect of naringin on gp120-induced injury mediated by P2X7 receptors in rat primary cultured microglia. PLoS One. 2017;12(8):e0183688. doi: 10.1371/journal.pone.0183688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Pu Z, Liu Y, Li C, Xu M, Xie H, Zhao J. Using network pharmacology for systematic understanding of geniposide in ameliorating inflammatory responses in colitis through suppression of NLRP3 inflammasome in macrophage by AMPK/Sirt1 dependent signaling. Am J Chin Med. 2020;48(7):1693–1713. doi: 10.1142/S0192415X20500846. [DOI] [PubMed] [Google Scholar]
  • 115.Fu C, Zhang X, Lu Y, Wang F, Xu Z, Liu S, Zheng H, Liu X. Geniposide inhibits NLRP3 inflammasome activation via autophagy in BV-2 microglial cells exposed to oxygen-glucose deprivation/reoxygenation. Int Immunopharmacol. 2020;84:106547. doi: 10.1016/j.intimp.2020.106547. [DOI] [PubMed] [Google Scholar]
  • 116.Li F, Li W, Li X, Li F, Zhang L, Wang B, Huang G, Guo X, Wan L, Liu Y, Zhang S, Kang S, Ma J. Geniposide attenuates inflammatory response by suppressing P2Y14 receptor and downstream ERK1/2 signaling pathway in oxygen and glucose deprivation-induced brain microvascular endothelial cells. J Ethnopharmacol. 2016;185:77–86. doi: 10.1016/j.jep.2016.03.025. [DOI] [PubMed] [Google Scholar]
  • 117.Mancuso C, Santangelo R. Ferulic acid: pharmacological and toxicological aspects. Food Chem Toxicol. 2014;65:185–195. doi: 10.1016/j.fct.2013.12.024. [DOI] [PubMed] [Google Scholar]
  • 118.Zhou Q, Gong X, Kuang G, Jiang R, Xie T, Tie H, Chen X, Li K, Wan J, Wang B. Ferulic acid protected from kidney ischemia reperfusion injury in mice: possible mechanism through increasing adenosine generation via HIF-1α. Inflammation. 2018;41(6):2068–2078. doi: 10.1007/s10753-018-0850-3. [DOI] [PubMed] [Google Scholar]
  • 119.Ding W, Ding Z, Wang Y, Zhu Y, Gao Q, Cao W, Du R. Evodiamine attenuates experimental colitis injury via activating autophagy and inhibiting NLRP3 inflammasome assembly. Front Pharmacol. 2020;11:573870. doi: 10.3389/fphar.2020.573870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lv Q, Xue Y, Li G, Zou L, Zhang X, Ying M, Wang S, Guo L, Gao Y, Li G, Xu H, Liu S, Xie J, Liang S. Beneficial effects of evodiamine on P2X(4)-mediated inflammatory injury of human umbilical vein endothelial cells due to high glucose. Int Immunopharmacol. 2015;28(2):1044–1049. doi: 10.1016/j.intimp.2015.08.020. [DOI] [PubMed] [Google Scholar]
  • 121.Xue Y, Guo T, Zou L, Gong Y, Wu B, Yi Z, Jia T, Zhao S, Shi L, Li L, Yuan H, Liu H, Gao Y, Li G, Liu S, Xu H, Zhang C, Liang S, Li G. Evodiamine attenuates P2X7-Mediated inflammatory injury of human umbilical vein endothelial cells exposed to high free fatty acids. Oxid Med Cell Longev. 2018;2018:5082817. doi: 10.1155/2018/5082817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Huang Q, Sun ML, Chen Y, Li XY, Wang YX. Concurrent bullatine A enhances morphine antinociception and inhibits morphine antinociceptive tolerance by indirect activation of spinal κ-opioid receptors. J Ethnopharmacol. 2017;196:151–159. doi: 10.1016/j.jep.2016.12.027. [DOI] [PubMed] [Google Scholar]
  • 123.Li J, Ren W, Huang XJ, Zou DJ, Hu X. Bullatine A, a diterpenoid alkaloid of the genus Aconitum, could attenuate ATP-induced BV-2 microglia death/apoptosis via P2X receptor pathways. Brain Res Bull. 2013;97:81–85. doi: 10.1016/j.brainresbull.2013.05.015. [DOI] [PubMed] [Google Scholar]
  • 124.Ranjbar A, Jamshidi M, Torabi S. Molecular modelling of the antiviral action of resveratrol derivatives against the activity of two novel SARS CoV-2 and 2019-nCoV receptors. Eur Rev Med Pharmacol Sci. 2020;24(14):7834–7844. doi: 10.26355/eurrev_202007_22288. [DOI] [PubMed] [Google Scholar]
  • 125.Huang TT, Lai HC, Chen YB, Chen LG, Wu YH, Ko YF, Lu CC, Chang CJ, Wu CY, Martel J, Ojcius DM, Chong KY, Young JD. cis-Resveratrol produces anti-inflammatory effects by inhibiting canonical and non-canonical inflammasomes in macrophages. Innate Immun. 2014;20(7):735–750. doi: 10.1177/1753425913507096. [DOI] [PubMed] [Google Scholar]
  • 126.Uddin SJ, Hasan MF, Afroz M, Sarker DK, Rouf R, Islam MT, Shilpi JA, Mubarak MS (2020) Curcumin and its multi-target function against pain and inflammation: an update of pre-clinical data. Curr Drug Targets 21. 10.2174/1389450121666200925150022 [DOI] [PubMed]
  • 127.Wang Z, Ren W, Zhao F, Han Y, Liu C, Jia K. Curcumin amends Ca2+ dysregulation in microglia by suppressing the activation of P2X7 receptor. Mol Cell Biochem. 2020;465(1-2):65–73. doi: 10.1007/s11010-019-03668-8. [DOI] [PubMed] [Google Scholar]
  • 128.de Vries S, Schaapveld M, Janus CPM, Daniëls LA, Petersen EJ, van der Maazen RWM, Zijlstra JM, Beijert M, Nijziel MR, Verschueren KMS, Kremer LCM, van Eggermond AM, Lugtenburg PJ, Krol ADG, Roesink JM, Plattel WJ, van Spronsen DJ, van Imhoff GW, de Boer JP, Aleman BMP, van Leeuwen FE (2020) Long-term cause-specific mortality in hodgkin lymphoma patients. J Natl Cancer Inst:djaa194
  • 129.Ding W, Feng H, Li WJ, Liao HH, Tang QZ. Research progress on the interaction between autophagy and energy homeostasis in cardiac remodeling. Front Pharmacol. 2020;11:587438. doi: 10.3389/fphar.2020.587438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Liu S, Wang X, Yu S, Yan M, Peng Y, Zhang G, Xu Z. A meta-analysis on the association between TNFSF4 polymorphisms (rs3861950 T > C and rs1234313 A > G) and susceptibility to coronary artery disease. Front Physiol. 2020;11:539288. doi: 10.3389/fphys.2020.539288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Erlinge D, Burnstock G. P2 receptors in cardiovascular regulation and disease. Purinergic Signal. 2008;4(1):1–20. doi: 10.1007/s11302-007-9078-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Harhun MI, Sukhanova K, Gordienko D, Dyskina Y. Molecular identification of P2X receptors in vascular smooth muscle cells from rat anterior, posterior, and basilar arteries. Pharmacol Rep. 2015;67(6):1055–1060. doi: 10.1016/j.pharep.2015.03.014. [DOI] [PubMed] [Google Scholar]
  • 133.Liu S, Yu S, Xu C, Peng L, Xu H, Zhang C, Li G, Gao Y, Fan B, Zhu Q, Zheng C, Wu B, Song M, Wu Q, Liang S. Puerarin alleviates aggravated sympathoexcitatory response induced by myocardial ischemia via regulating P2X3 receptor in rat superior cervical ganglia. Neurochem Int. 2014;70:39–49. doi: 10.1016/j.neuint.2014.03.004. [DOI] [PubMed] [Google Scholar]
  • 134.Zhao L, Xu Y, Tao L, Yang Y, Shen X, Li L, Luo P. Oxymatrine inhibits transforming growth factor β1 (TGF-β1)-induced cardiac fibroblast-to-myofibroblast transformation (fmt) by mediating the Notch signaling pathway in vitro. Med Sci Monit. 2018;24:6280–6288. doi: 10.12659/MSM.910142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Li G, Liu S, Yang Y, Xie J, Liu J, Kong F, Tu G, Wu R, Li G, Liang S. Effects of oxymatrine on sympathoexcitatory reflex induced by myocardial ischemic signaling mediated by P2X3 receptors in rat SCG and DRG. Brain Res Bull. 2011;84(6):419–424. doi: 10.1016/j.brainresbull.2011.01.011. [DOI] [PubMed] [Google Scholar]
  • 136.Zhang P, Hou J, Fu J, Li D, Zhang C, Liu J. Baicalin protects rat brain microvascular endothelial cells injured by oxygen-glucose deprivation via anti-inflammation. Brain Res Bull. 2013;97:8–15. doi: 10.1016/j.brainresbull.2013.05.005. [DOI] [PubMed] [Google Scholar]
  • 137.Sheng X, Wang J, Guo J, Xu Y, Jiang H, Zheng C, Xu Z, Zhang Y, Che H, Liang S, Zhu G, Li G. Effects of baicalin on diabetic cardiac autonomic neuropathy mediated by the P2Y12 receptor in rat stellate ganglia. Cell Physiol Biochem. 2018;46(3):986–998. doi: 10.1159/000488828. [DOI] [PubMed] [Google Scholar]
  • 138.He J, Zhou Y, Arredondo Carrera HM, Sprules A, Neagu R, Zarkesh SA, Eaton C, Luo J, Gartland A, Wang N. Inhibiting the P2X4 receptor suppresses prostate cancer growth in vitro and in vivo, suggesting a potential clinical target. Cells. 2020;9(11):2511. doi: 10.3390/cells9112511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Jones GD, Brandt WS, Shen R, Sanchez-Vega F, Tan KS, Martin A, Zhou J, Berger M, Solit DB, Schultz N, Rizvi H, Liu Y, Adamski A, Chaft JE, Riely GJ, Rocco G, Bott MJ, Molena D, Ladanyi M, Travis WD, Rekhtman N, Park BJ, Adusumilli PS, Lyden D, Imielinski M, Mayo MW, Li BT, Jones DR (2020) A genomic-pathologic annotated risk model to predict recurrence in early-stage lung adenocarcinoma. JAMA Surg:e205601 [DOI] [PMC free article] [PubMed]
  • 140.Vultaggio-Poma V, Sarti AC, Di Virgilio F. Extracellular ATP: A feasible target for cancer therapy. Cells. 2020;9(11):2496. doi: 10.3390/cells9112496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Nooshabadi VT, Arab S (2020) Targeting tumor-derived exosomes expressing CD73: new opportunities in the pathogenesis and treatment of cancer. Curr Mol Med 20. 10.2174/1566524020666201120142953 [DOI] [PubMed]
  • 142.Xu K, Wu X, Cheng Y, Yan J, Feng Y, Chen R, Zheng R, Li X, Song P, Wang Y, Zhang H. A biomimetic nanoenzyme for starvation therapy enhanced photothermal and chemodynamic tumor therapy. Nanoscale. 2020;12(45):23159–23165. doi: 10.1039/d0nr05097k. [DOI] [PubMed] [Google Scholar]
  • 143.Chen R, Huang L, Hu K. Natural products remodel cancer-associated fibroblasts in desmoplastic tumors. Acta Pharm Sin B. 2020;10(11):2140–2155. doi: 10.1016/j.apsb.2020.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zhang XG, Ma GY, Kou F, Liu WJ, Sun QY, Guo GJ, Ma XD, Guo SJ, Jian-Ning Z. Reynoutria japonica from traditional Chinese medicine: a source of competitive adenosine deaminase inhibitors for anticancer. Comb Chem High Throughput Screen. 2019;22(2):113–122. doi: 10.2174/1386207322666190415100618. [DOI] [PubMed] [Google Scholar]
  • 145.Jelassi B, Anchelin M, Chamouton J, Cayuela ML, Clarysse L, Li J, Goré J, Jiang LH, Roger S. Anthraquinone emodin inhibits human cancer cell invasiveness by antagonizing P2X7 receptors. Carcinogenesis. 2013;34(7):1487–1496. doi: 10.1093/carcin/bgt099. [DOI] [PubMed] [Google Scholar]
  • 146.Navarro M, Arnaez E, Moreira I, Hurtado A, Monge D, Monagas M. Polyphenolic composition and antioxidant activity of uncaria tomentosa commercial bark products. Antioxidants (Basel) 2019;8(9):339. doi: 10.3390/antiox8090339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Santos KF, Gutierres JM, Pillat MM, Rissi VB, Santos Araújo MD, Bertol G, Gonçalves PB, Schetinger MR, Morsch VM. Uncaria tomentosa extract alters the catabolism of adenine nucleotides and expression of ecto-5'-nucleotidase/CD73 and P2X7 and A1 receptors in the MDA-MB-231 cell line. J Ethnopharmacol. 2016;194:108–116. doi: 10.1016/j.jep.2016.08.051. [DOI] [PubMed] [Google Scholar]
  • 148.Goto S, Hasebe T, Takagi S. Platelets: small in size but essential in the regulation of vascular homeostasis-translation from basic science to clinical medicine. Circ J. 2015;79(9):1871–1881. doi: 10.1253/circj.CJ-14-1434. [DOI] [PubMed] [Google Scholar]
  • 149.Xu H, Lu H, Zhu X, Wang W, Zhang Z, Fu H, Ma S, Luo Y, Fu J. Inhibitory effects of luteolin-4'-O-β-D-glucopyranoside on P2Y12 and thromboxane A2 receptor-mediated amplification of platelet activation in vitro. Int J Mol Med. 2018;42(1):615–624. doi: 10.3892/ijmm.2018.3634. [DOI] [PubMed] [Google Scholar]
  • 150.Gerhards C, Uhlig S, Etemad M, Christodoulou F, Bieback K, Klüter H, Bugert P (2020) Expression of ADP receptor P2Y12, thromboxane A2 receptor and C-type lectin-like receptor 2 in cord blood-derived megakaryopoiesis. Platelets:1–8 [DOI] [PubMed]
  • 151.Ye Y, Zhao X, Tu C, Li Q, Zeng Y. Elevated serum levels of alkaline phosphatase and the risk of low responsiveness to clopidogrel. Int Heart J. 2020;61(6):1135–1141. doi: 10.1536/ihj.20-285. [DOI] [PubMed] [Google Scholar]
  • 152.Fang Y, Yu Y, Hou Q, Zheng X, Zhang M, Zhang D, Li J, Wu XR, Huang C. The Chinese herb isolate isorhapontigenin induces apoptosis in human cancer cells by down-regulating overexpression of antiapoptotic protein XIAP. J Biol Chem. 2012;287(42):35234–35243. doi: 10.1074/jbc.M112.389494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lu Y, Wang A, Shi P, Zhang H. A theoretical study on the antioxidant activity of piceatannol and isorhapontigenin scavenging nitric oxide and nitrogen dioxide radicals. PLoS One. 2017;12(1):e0169773. doi: 10.1371/journal.pone.0169773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Yeo SCM, Fenwick PS, Barnes PJ, Lin HS, Donnelly LE. Isorhapontigenin, a bioavailable dietary polyphenol, suppresses airway epithelial cell inflammation through a corticosteroid-independent mechanism. Br J Pharmacol. 2017;174(13):2043–2059. doi: 10.1111/bph.13803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ravishankar D, Albadawi DAI, Chaggar V, Patra PH, Williams HF, Salamah M, Vaiyapuri R, Dash PR, Patel K, Watson KA, Vaiyapuri S. Isorhapontigenin, a resveratrol analogue selectively inhibits ADP-stimulated platelet activation. Eur J Pharmacol. 2019;862:172627. doi: 10.1016/j.ejphar.2019.172627. [DOI] [PubMed] [Google Scholar]
  • 156.Bertoni APS, de Campos RP, Tamajusuku ASK, Stefani GP, Braganhol E, Battastini AMO, Wink MR. Biochemical analysis of ectonucleotidases on primary rat vascular smooth muscle cells and in silico investigation of their role in vascular diseases. Life Sci. 2020;256:117862. doi: 10.1016/j.lfs.2020.117862. [DOI] [PubMed] [Google Scholar]
  • 157.Akinyemi AJ, Thomé GR, Morsch VM, Bottari NB, Baldissarelli J, de Oliveira LS, Goularte JF, Belló-Klein A, Oboh G, Schetinger MR. Dietary supplementation of ginger and turmeric rhizomes modulates platelets ectonucleotidase and adenosine deaminase activities in normotensive and hypertensive rats. Phytother Res. 2016;30(7):1156–1163. doi: 10.1002/ptr.5621. [DOI] [PubMed] [Google Scholar]
  • 158.Boligon AA, Pimentel VC, Bagatini MD, Athayde ML. Effect of Scutia buxifolia Reissek in nucleotidase activities and inhibition of platelet aggregation. J Nat Med. 2015;69(1):46–54. doi: 10.1007/s11418-014-0858-4. [DOI] [PubMed] [Google Scholar]
  • 159.Boligon AA, da Rosa ML, Piana M, de Campos MM, Oliveira SM. Topical antiedematogenic and anti-inflammatory effect of Scutia buxifolia Reissek gel and stability study. J Photochem Photobiol B. 2017;167:29–35. doi: 10.1016/j.jphotobiol.2016.12.026. [DOI] [PubMed] [Google Scholar]
  • 160.Neto-Neves EM, da Silva Maia Bezerra Filho C, Dejani NN, de Sousa DP (2021) Ferulic acid and cardiovascular health: therapeutic and preventive potential. Mini Rev Med Chem 21. 10.2174/1389557521666210105122841 [DOI] [PubMed]
  • 161.Wang X, Hou Y, Li Q, Li X, Wang W, Ai X, Kuang T, Chen X, Zhang Y, Zhang J, Hu Y, Meng X. Rhodiola crenulata attenuates apoptosis and mitochondrial energy metabolism disorder in rats with hypobaric hypoxia-induced brain injury by regulating the HIF-1α/microRNA 210/ISCU1/2(COX10) signaling pathway. J Ethnopharmacol. 2019;241:111801. doi: 10.1016/j.jep.2019.03.028. [DOI] [PubMed] [Google Scholar]
  • 162.Hou Y, Tang Y, Wang X, Ai X, Wang H, Li X, Chen X, Zhang Y, Hu Y, Meng X, Zhang J. Rhodiola Crenulata ameliorates exhaustive exercise-induced fatigue in mice by suppressing mitophagy in skeletal muscle. Exp Ther Med. 2020;20(4):3161–3173. doi: 10.3892/etm.2020.9072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Ai X, Yu P, Hou Y, Song X, Luo J, Li N, Lai X, Wang X, Meng X. A review of traditional Chinese medicine on treatment of diabetic retinopathy and involved mechanisms. Biomed Pharmacother. 2020;132:110852. doi: 10.1016/j.biopha.2020.110852. [DOI] [PubMed] [Google Scholar]
  • 164.Yang D, Huang WY, Li YQ, Chen SY, Su SY, Gao Y, Meng XL, Wang P (2020) Acute and subchronic toxicity studies of rhein in immature and d-galactose-induced aged mice and its potential hepatotoxicity mechanisms. Drug Chem Toxicol:1–12 [DOI] [PubMed]
  • 165.Wang X, Hou Y, Ai X, Sun J, Xu B, Meng X, Zhang Y, Zhang S. Potential applications of microfluidics based blood brain barrier (BBB)-on-chips for in vitro drug development. Biomed Pharmacother. 2020;132:110822. doi: 10.1016/j.biopha.2020.110822. [DOI] [PubMed] [Google Scholar]
  • 166.Wang X, Liu Z, Fan F, Hou Y, Yang H, Meng X, Zhang Y, Ren F. Microfluidic chip and its application in autophagy detection. TrAC Trends Anal Chem. 2019;117:300–315. [Google Scholar]
  • 167.Huizing LRS, McDuffie J, Cuyckens F, van Heerden M, Koudriakova T, Heeren RMA, Vreeken RJ. Quantitative mass spectrometry imaging to study drug distribution in the intestine following oral dosing. Anal Chem. 2021;93(4):2144–2151. doi: 10.1021/acs.analchem.0c03956. [DOI] [PubMed] [Google Scholar]
  • 168.Zan B, Shi R, Wang T, Wu J, Ma Y, Cheng N. Simultaneous quantification of multiple active components from Xiexin decoction in rat plasma by LC-ESI-MS/MS: application in pharmacokinetics. Biomed Chromatogr. 2011;25(7):816–826. doi: 10.1002/bmc.1521. [DOI] [PubMed] [Google Scholar]
  • 169.Kuenzi BM, Park J, Fong SH, Sanchez KS, Lee J, Kreisberg JF, Ma J, Ideker T. Predicting drug response and synergy using a deep learning model of human cancer cells. Cancer Cell. 2020;38(5):672–684.e6. doi: 10.1016/j.ccell.2020.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.


Articles from Purinergic Signalling are provided here courtesy of Springer

RESOURCES