Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2736 word(s) 2736 2021-08-17 10:10:51 |
2 format correct Meta information modification 2736 2021-09-29 11:18:25 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Allegrini, S. Purine-Metabolising Enzymes. Encyclopedia. Available online: https://encyclopedia.pub/entry/14723 (accessed on 28 March 2024).
Allegrini S. Purine-Metabolising Enzymes. Encyclopedia. Available at: https://encyclopedia.pub/entry/14723. Accessed March 28, 2024.
Allegrini, Simone. "Purine-Metabolising Enzymes" Encyclopedia, https://encyclopedia.pub/entry/14723 (accessed March 28, 2024).
Allegrini, S. (2021, September 29). Purine-Metabolising Enzymes. In Encyclopedia. https://encyclopedia.pub/entry/14723
Allegrini, Simone. "Purine-Metabolising Enzymes." Encyclopedia. Web. 29 September, 2021.
Purine-Metabolising Enzymes
Edit

The enzymes of both de novo and salvage pathways for purine nucleotide synthesis are regulated to meet the demand of nucleic acid precursors during proliferation. Among them, the salvage pathway enzymes seem to play the key role in replenishing the purine pool in dividing and tumour cells that require a greater amount of nucleotides. An imbalance in the purine pools is fundamental not only for preventing cell proliferation, but also, in many cases, to promote apoptosis. It is known that tumour cells harbour several mutations that might lead to defective apoptosis-inducing pathways, and this is probably at the basis of the initial expansion of the population of neoplastic cells. Therefore, knowledge of the molecular mechanisms that lead to apoptosis of tumoural cells is key to predicting the possible success of a drug treatment and planning more effective and focused therapies. 

purine salvage apoptosis CD73 cN-II ADA PNP HPRT IMPDH SAMHD1 MTH1

1. Introduction

Intracellular purine nucleotide concentration is determined and maintained through two distinct pathways, both depending on a common metabolite: 5-phosphoribosyl-1-pyrophosphate (PRPP) (Figure 1). The de novo pathway consists of 10 reactions catalysed by six enzymes, some of which are allosterically regulated mainly by PRPP and purine nucleotides [1]. Furthermore, the six enzymes can cluster near mitochondria and microtubules to form dynamic multienzyme complexes referred to as “purinosomes” [2]. The purinosome formation causes a strong increase in the rate of purine synthesis. This regulatory mechanism ensures that, during proliferation and in the absence of preformed purine ring to be salvaged, the supply of purine compounds is secured. When preformed purine rings are available, they can be converted in one step into the corresponding nucleoside monophosphates through the action of the salvage pathway enzymes adenine phosphoribosyltransferase (APRT) and hypoxanthine-guanine phosphoribosyltransferase (HPRT) which utilize PRPP as a co-substrate (Figure 1) [1]. The availability of preformed purine ring prevents purinosome formation [2]. Regulation both at genetic and protein levels ensures the correct amount of nucleotides to sustain replicative and metabolic needs [3]. Therefore, it is not surprising that alterations of replicative rate and eventually activation of apoptosis are a consequence of dysfunctions of purine metabolism. Furthermore, an imbalance in purine supply can affect also mitochondrial proliferation and function, leading to metabolic changes and apoptosis [4]. Nevertheless, in some cases, the relationship between enzyme dysfunction and proliferation rate, metabolic alterations, and apoptosis is not as simple and direct as expected. The knowledge of the consequences of the alteration of enzymes involved in purine metabolism is essential not only for the understanding of “where and how” these enzymes impact metabolic pathways, but also to uncover whether they can be targets of antineoplastic drugs or be responsible for drug resistance. This review presents the most recent reports on the impact of purine catabolism and salvage enzymes (Figure 2) in the activation of apoptosis and the implied molecular mechanisms. Furthermore, the possible applications of this knowledge to anti-tumour therapy are discussed.
Figure 1. De novo and salvage pathways for purine nucleotide biosynthesis. Cyan background: de novo synthesis; yellow background: salvage synthesis. The figure outlines the central role played by PRPP, needed for both de novo and salvage pathways. Hyp: hypoxanthine; Gua: guanine; Ade: adenine; Rib-5-P: ribose-5-phosphate; PRPP: 5-phosphoribosyl-1-pyrophosphate; Gln: glutamine; Gly: glycine; THF: tetrahydrofolate; Asp: aspartate; S-AMP: succinyl-AMP; XMP: xanthosine-5′-monophosphate; IMP: inosine-5′-monophosphate; GMP: guanosine-5′-monphosphate; AMP: adenosine-5′-monophosphate.
Figure 2. Pathways of purine metabolism. 1: Ectosolic 5′-nucleotidase; 2: Adenosine deaminase; 3: Cytosolic 5′-nucleotidase II; 4: Purine nucleoside phosphorylase; 5: Hypoxanthine-guanine phosphoribosyltransferase; 6: IMP dehydrogenase. Inset: deoxyribonucleoside triphosphates (dNTP) are converted into the respective deoxynucleosides (dNs) by a one-step reaction catalysed by a sterile alpha motif and histidine-aspartate (HD) domain-containing protein 1 (enzyme 7). An increase in reactive oxygen species (ROS) brings about an increase in 8-oxo-dGTP, converted into the monophosphate by human MutT homolog 1 (enzyme 8). Ado: adenosine; Guo: guanosine; Ino: inosine.

2. Ectosolic 5′-Nucleotidase

Ectosolic 5′-nucleotidase (CD73) (EC 3.1.3.5) is a 70-kDa glycosylated protein bound to the outer surface of the plasma membrane by a glycosylphosphatidylinositol anchor. The enzyme is overexpressed in a variety of tumours [5] and in some of them, is associated with a highly invasive cancer phenotype, drug resistance, and tumour-promoting functions [6]. CD73 catalyses the dephosphorylation of extracellular AMP to adenosine (Ado), which plays important roles in many physiological and pathophysiological conditions through G-protein coupled Ado receptors (A1, A2A, A2B, and A3) [7][8]. The roles played by Ado are complex since its interaction with different receptors achieves different results; in particular, A1 and A3 receptors are coupled with Gi-proteins determining a decrease in cyclic AMP (cAMP), while A2A and A2B receptors are coupled with Gs-proteins causing an increase in intracellular cAMP [9] (Figure 3). Therefore, the role played by CD73 depends on the nature and distribution of the Ado receptors in a particular kind of cell. In addition, CD73 plays also many different roles in cell physiology not related to its catalytic activity [10]. Analysing enzymatic and non-enzymatic functions of CD73, it was concluded that both faces were involved in the aggressive behaviour of cancer cells. The enzymatic function seems to be primarily involved in invasion, whereas the non-enzymatic action of the protein contributes to cell adhesion and migration through activation of focal adhesion kinase [10]. Among the tumours in which CD73 is upregulated, breast cancer is the most studied [11]. In some cases of this type of cancer, an involvement of CD73 enzymatic activity was demonstrated, since supply of Ado could be a substitute for enzyme upregulation in promoting proliferation and motility [12]. In other cases, the involvement of different not yet described mechanisms independent of enzyme activity was demonstrated [13]. In all cases, an activation of the Akt/GSK-3β pathway was found to be involved in the tumour growth and motility promoted by CD73 [13]. In human cervical cancer cells, an increase in cell proliferation and motility was associated to CD73 overexpression, but the mechanism was independent of enzyme activity. In fact, CD73 inhibitors were unable to prevent the increase in proliferation in cells that overexpressed the enzyme. In addition, an increase in Ado, that could be expected when the enzyme is overexpressed, induced a decreased cell proliferation [14]. In this type of tumour, an activation of the Akt pathway was demonstrated as well [14]. In colorectal cancer, a downregulation of miR-30a was shown to determine an increase of CD73 expression in tumour cells, which promoted proliferation and inhibited apoptosis. MiR-30a is one of the most important tumour-suppressor factors in various human cancers and its level is significantly decreased in several tumours [15]. Since, as stated before, many effects exerted by the expression of CD73 on tumours are mediated by the participation of the enzyme activity in the conversion of extracellular ATP into Ado, such effects are dependent on the amount and nature of the Ado receptors expressed by tumour cells and other cells present in the tumour microenvironment. In this regard, in contrast to reports for other cancer types (see above), in an in vivo study on medulloblastoma, overexpression of CD73 reduced tumour growth and vascularization, and also promoted differentiation and initiated apoptosis, supposedly by the accumulation of Ado which interacted with A1 receptor [16]. The level of CD73 expression is very important not only for tumour growth and motility, but also for the success of the therapeutic approach. In fact, during chemotherapy, various immunogenic mediators accumulate in the tumour microenvironment, included ATP. Extracellular ATP, in this case, is released by the cells undergoing intrinsically or extrinsically activated apoptosis, through pannexin-1 channels and functions as a “find me” signal for P2Y family receptors expressed by macrophages and dendritic cells [17]. In this mechanism, the rapid ATP degradation catalysed by several extracellular enzymes is a determinant for the immunogenic activation, since the accumulated AMP, which cannot interact with P2Y receptors, can generate Ado through CD73, which mainly mediates the immune-escape of tumour cells interacting with A2A receptors [18]. Moreover, also in the case of regulatory T-cells (Tregs) in a tumour environment, the presence of several signals triggers apoptosis, and apoptotic Treg cells achieved superior immuno-suppression via an oxidative stress-associated mechanism [19]. Therefore, the induction of apoptosis in a tumour environment in which cells highly expressing CD73 and Ado receptors A2A are present is not always a successful therapeutic approach. Recently, CD73 has been targeted for the synthesis of new inhibitory compounds which prevent extracellular Ado formation from AMP and the consequent immune-escape [20].
Figure 3. Effectors of adenosine receptor-mediated apoptosis. The figure shows the different types of G-proteins associated with the four adenosine receptors and illustrates the apoptosis effectors found in several models by using agonists and antagonists of the receptors, or receptor silencing. The numbers in brackets refer to the respective reference. Note that adenosine receptors can be involved in survival in other cell types [21].

3. Cytosolic 5′-Nucleotidase II

Cytosolic 5′-nucleotidase II (cN-II) (EC 3.1.3.5) is an ubiquitously expressed and highly conserved enzyme that hydrolyses purine nucleoside monophosphates (preferentially IMP and GMP) into their corresponding nucleosides and inorganic phosphate. Its enzymatic activity and biochemical features have been well characterised, and the reader is referred to a recent review for further investigation of these issues [22]. The first observation on its prognostic value in nucleoside analogue-treated patients with acute myeloid leukaemia indicated that patients with high cN-II expression in leukaemic blasts showed a poorer outcome with respect to those with a lower expression [23]. Since then, the implication of cN-II in cancer cells and in the response to anticancer treatment has been extensively demonstrated [24][25]. Indeed, shRNA-based cell models with downregulated cN-II are more sensitive to purine nucleoside and nucleobase analogues, as compared to control cells [26]. The enzyme is highly expressed in tumour cells, and cN-II expression in human neuroblastoma cells and in lung cancer cells correlated with cell proliferation [27][28], whereas its inhibition in human breast cancer cells was associated with a better defence towards reactive oxygen species (ROS) and a better adaptability to glucose deprivation in culture media [29]. In a cell model of lung cancer (A549), an activation of p53 and inactivation of Akt following cN-II partial silencing was demonstrated [28]. Indeed, in an astrocytoma cell line (ADF), transitory cN-II silencing was followed by caspase-3 activation and apoptosis [30]. Furthermore, the siRNA-mediated inhibition of cN-II expression in murine skeletal muscles induced an increase in the AMP/ATP ratio and a subsequent activation of AMP-activated protein kinase (AMPK) [31], even though this was not confirmed in cN-II deficient mice [32]. In parallel to these biological studies, several genetic as well as genome wide association studies have identified the cN-II encoding gene NT5C2 or some genetic variants as being associated to various pathological conditions such as hereditary spastic paraplegia 45, psychiatric disorders, a disturbance of blood pressure, and a decrease in body mass index [33]. Fluctuation of cN-II expression has been demonstrated to influence the concentration of intracellular nucleotides, depending on the type of cell and the technique utilised for enzyme silencing. In some cases, the alteration of nucleotide concentration is not statistically significant, but the effect on metabolic and proliferative features is still present [28][32][34][35]. These observations indicate that cN-II regulates several cellular pathways through a mechanism at least partially independent of its impact on intracellular nucleotide concentration.

4. Adenosine Deaminase

Adenosine deaminase (ADA) (E.C.3.5.4.4) catalyses the deamination of Ado and deoxyadenosine (dAdo) to inosine and deoxyinosine, respectively. There are two isoenzymes of ADA in human tissues, ADA1 and ADA2 [36]. ADA1 is ubiquitous, has a similar affinity for both substrates, and can interact with membrane proteins such as the dipeptidyl dipeptidase-4 (CD26), A1 [37] and A2A receptors [38]. ADA1 also acts as an ectoenzyme which catalyses the deamination of Ado and dAdo in the extracellular space. ADA2 is the main ADA isoenzyme found in human serum [36]. Low ADA activity was found in prostate [39] and gastric tumour tissues [40] and in lymphocytes of patients suffering from different pathologies, such as gynaecological [41], renal [42], head and neck [43], and gastric tumours [44][45], as well as Hodgkin’s lymphoma [46]. Low ADA activity in lymphocytes, as outlined later on in this section, could account for the decreased cellular immune function in cancer patients. Conversely, ADA activity was increased in cancerous tissue from breast [47][48][49], kidney [50], and colorectal tumours [51], in serum of patients with bladder [52] ovarian [53], laryngeal [54], and head and neck squamous cell carcinomas [55][56] and in lymphocytes of patients with chronic lymphocytic leukaemia (CLL) [57]. High ADA activity might be advantageous to the cancer cells by causing, in association with purine nucleoside phosphorylase (PNP), an increase in hypoxanthine, a substrate for the salvage pathway (Figure 2). In addition, increased ADA activity might be a compensatory mechanism against toxic accumulation of its substrates. Indeed, Ado and dAdo are known to induce apoptosis, and ADA inhibition is an antitumoural strategy. Insights from the apoptosis-induced effect of ADA deficiency were obtained from studies regarding severe combined immunodeficiency (SCID) caused by mutations in the ADA1 gene. The primary cause of lymphotoxicity in ADA-SCID is considered to be the accumulation of dAdo and dATP. In the absence of ADA activity, Ado and dAdo accumulate both in the extracellular compartments and inside the cells. dAdo is then phosphorylated by deoxycytidine kinase (dCK) and/or ADK to dAdo monophosphate, which in turn is converted to dAdo triphosphate (dATP). Intracellular dATP might generate DNA strand breaks and inhibit ribonucleotide reductase, leading to DNA synthesis impairment and apoptosis [58]. Deoxycoformycin (dCF), a powerful inhibitor of ADA [59], has been used alone or in combination with other drugs for the treatment of several types of lymphatic leukaemia [60][61][62][63]. dATP accumulation induced by dCF in hairy cell leukaemia led to activation of p53, release of cytochrome c from mitochondria and activation of apoptotic protease-activating factor 1 (Apaf-1), and therefore caspase-9 and caspase-3 activation [64].
The combination of dAdo and dCF was found to be toxic for several tumoural cell lines such as rat hepatoma cells [65], and human colon carcinoma cell lines LoVo and HT29 [66][67][68]. The treatment with dAdo and dCF in combination resulted in the activation of the apoptotic mitochondrial pathway in LoVo, human astrocytoma, and neuroblastoma cell lines [68][69][70][71] with cytochrome c release and caspase-3 activation. Activation of caspase-8, and of both caspase-9 and -8 has also been found in astrocytoma and in neuroblastoma cells, respectively [70][71]. In astrocytoma cells, but not in neuroblastoma cells, a reduction in the production of lactate preceded the effect of dAdo and dCF on cell viability, suggesting a decreased glycolytic capacity. In both cell lines, dAdo must be phosphorylated in order to exert its cytotoxic effect; however, a decrease in the energy charge was observed in astrocytoma, but not in neuroblastoma cells [70][71].
The involvement of the four Ado receptors in apoptosis of cancer cells has been reported and reviewed recently [21][72]. Apoptosis can occur through A1 [73][74][75], A2A [76][77][78], A2B [79][80], and A3 [81][82][83][84][85][86][87][88][89] receptors (Figure 3). Extracellular Ado can enter the cells through Ado transporters, and as intracellular Ado is converted by ADK to AMP which can activate AMPK, an energy sensor of the cells involved both in survival and cancer suppression (for a recent review see [90]). AMPK is responsible for apoptosis in some human gastric [91][92] cancer cells and astrocytoma cells among others [74]. In human mesothelioma cells this pathway led to upregulation of p53 [93]. p53 exerts its pro-apoptotic effect by transcription-dependent and transcription-independent actions. The targets of p53 transactivation are pro-apoptotic members of the Bcl-2 family (Bax, Bid, Puma, Noxa), as well as other apoptotic effector proteins (Apaf-1, caspase-8, caspase-6), cell death receptors, and cell death ligands. The p53 protein also acts directly in the mitochondria facilitating the oligomerization of Bax and Bak and interacting with anti-apoptotic Bcl-2, Bcl-xL, and Mcl-1 proteins [94]. The ADA inhibitor erythro-9-(2-hydroxy-3-nonyl)adenine (EHNA) but not dCF, induced apoptosis in malignant pleural mesothelioma [95] by increasing intracellular Ado that needed to be converted to AMP, since ADK inhibition neutralised the effect. EHNA also inhibits cyclic nucleotide phosphodiesterase [96], therefore, it was postulated that this enzyme might have a role in the cytotoxicity observed in these cells. In addition, Ado-induced apoptosis involves the intrinsic and extrinsic pathways resulting in caspase-3 activation, and caspase-independent pathways leading to accumulation of Apoptosis Inducing Factor (AIF) (or its homologous AIF-homologous mitochondrion-associated inducer of death, AMID) in the nucleus [21][97]. AIF can be released from mitochondria, migrates to the nucleus where it can recruit nucleases, or organize a DNA-degrading complex [98]. In human hepatoma cells, Ado upregulated AMID, and promoted its translocation to the nucleus where it may induce DNA degradation [99]. Ado accumulation also reverses the action of S-adenosylhomocysteine hydrolase, increasing S-adenosylhomocysteine, which inhibits the transmethylation reactions. S-adenosylhomocysteine has been suggested to be involved in Ado-induced apoptosis in hepatoma HepG2 by altering gene expression [100]. It is interesting to note that dCF, through Ado receptor-dependent mechanisms, was also able to decrease the aggressiveness of cancer cells by modulating migration and invasion and by regulating endothelial cell permeability [101].

References

  1. Ipata, P.L.; Balestri, F.; Camici, M.; Tozzi, M.G. Molecular mechanisms of nucleoside recycling in the brain. Int. J. Biochem. Cell Biol. 2011, 43, 140–145.
  2. An, S.; Kumar, R.; Sheets, E.D.; Benkovic, S.J. Reversible compartmentalization of de novo purine biosynthetic complexes in living cells. Science 2008, 320, 103–106.
  3. Pedley, A.M.; Benkovic, S.J. A new view into the regulation of purine metabolism: The purinosome. Trends Biochem. Sci. 2017, 42, 141–154.
  4. Rampazzo, C.; Miazzi, C.; Franzolin, E.; Pontarin, G.; Ferraro, P.; Frangini, M.; Reichard, P.; Bianchi, V. Regulation by degradation, a cellular defense against deoxyribonucleotide pool imbalances. Mutat. Res. 2010, 703, 2–10.
  5. Zhang, B. CD73 promotes tumor growth and metastasis. Oncoimmunology 2012, 1, 67–70.
  6. Gao, Z.W.; Dong, K.; Zhang, H.Z. The roles of CD73 in cancer. Biomed. Res. Int. 2014, 2014, 460654.
  7. Regateiro, F.S.; Cobbold, S.P.; Waldmann, H. CD73 and adenosine generation in the creation of regulatory microenvironments. Clin. Exp. Immunol. 2013, 171, 1–7.
  8. Kazemi, M.H.; Raoofi Mohseni, S.; Hojjat-Farsangi, M.; Anvari, E.; Ghalamfarsa, G.; Mohammadi, H.; Jadidi-Niaragh, F. Adenosine and adenosine receptors in the immunopathogenesis and treatment of cancer. J. Cell Physiol. 2018, 233, 2032–2057.
  9. Sousa, J.B.; Fresco, P.; Diniz, C.; Goncalves, J. Adenosine receptor ligands on cancer therapy: A review of patent literature. Recent Pat. Anticancer Drug Discov. 2018, 13, 40–69.
  10. Sadej, R.; Skladanowski, A.C. Dual, enzymatic and non-enzymatic, function of ecto-5’-nucleotidase (eN, CD73) in migration and invasion of A375 melanoma cells. Acta Biochim. Pol. 2012, 59, 647–652.
  11. Stagg, J.; Divisekera, U.; McLaughlin, N.; Sharkey, J.; Pommey, S.; Denoyer, D.; Dwyer, K.M.; Smyth, M.J. Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2010, 107, 1547–1552.
  12. Yang, X.; Pei, S.; Wang, H.; Jin, Y.; Yu, F.; Zhou, B.; Zhang, H.; Zhang, D.; Lin, D. Tiamulin inhibits breast cancer growth and pulmonary metastasis by decreasing the activity of CD73. BMC Cancer 2017, 17, 255.
  13. Yu, J.; Wang, X.; Lu, Q.; Wang, J.; Li, L.; Liao, X.; Zhu, W.; Lv, L.; Zhi, X.; Yu, J.; et al. Extracellular 5’-nucleotidase (CD73) promotes human breast cancer cells growth through AKT/GSK-3beta/beta-catenin/cyclinD1 signaling pathway. Int. J. Cancer 2018, 142, 959–967.
  14. Gao, Z.W.; Wang, H.P.; Lin, F.; Wang, X.; Long, M.; Zhang, H.Z.; Dong, K. CD73 promotes proliferation and migration of human cervical cancer cells independent of its enzyme activity. BMC Cancer 2017, 17, 135.
  15. Lu, N.; Lin, T.; Wang, L.; Qi, M.; Liu, Z.; Dong, H.; Zhang, X.; Zhai, C.; Wang, Y.; Liu, L.; et al. Association of SOX4 regulated by tumor suppressor miR-30a with poor prognosis in low-grade chondrosarcoma. Tumour Biol. 2015, 36, 3843–3852.
  16. Cappellari, A.R.; Pillat, M.M.; Souza, H.D.; Dietrich, F.; Oliveira, F.H.; Figueiro, F.; Abujamra, A.L.; Roesler, R.; Lecka, J.; Sevigny, J.; et al. Ecto-5’-Nucleotidase overexpression reduces tumor growth in a xenograph medulloblastoma model. PLoS ONE 2015, 10, e0140996.
  17. Boyd-Tressler, A.M.; Lane, G.S.; Dubyak, G.R. Up-Regulated ectonucleotidases in fas-associated death domain protein- and receptor-interacting protein kinase 1-deficient jurkat leukemia cells counteract extracellular ATP/AMP accumulation via Pannexin-1 channels during chemotherapeutic drug-induced apoptosis. Mol. Pharm. 2017, 92, 30–47.
  18. Antonioli, L.; Blandizzi, C.; Pacher, P.; Hasko, G. Immunity, inflammation and cancer: A leading role for adenosine. Nat. Rev. Cancer 2013, 13, 842–857.
  19. Maj, T.; Wang, W.; Crespo, J.; Zhang, H.; Wang, W.; Wei, S.; Zhao, L.; Vatan, L.; Shao, I.; Szeliga, W.; et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat. Immunol. 2017, 18, 1332–1341.
  20. Dumontet, C.; Peyrottes, S.; Rabeson, C.; Cros-Perrial, E.; Geant, P.Y.; Chaloin, L.; Jordheim, L.P. CD73 inhibition by purine cytotoxic nucleoside analogue-based diphosphonates. Eur. J. Med. Chem. 2018, 157, 1051–1055.
  21. Soleimani, A.; Bahreyni, A.; Roshan, M.K.; Soltani, A.; Ryzhikov, M.; Shafiee, M.; Soukhtanloo, M.; Jaafari, M.R.; Mashkani, B.; Hassanian, S.M. Therapeutic potency of pharmacological adenosine receptors agonist/antagonist on cancer cell apoptosis in tumor microenvironment, current status, and perspectives. J. Cell. Physiol. 2019, 234, 2329–2336.
  22. Tozzi, M.G.; Pesi, R.; Allegrini, S. On the physiological role of cytosolic 5’-nucleotidase II (cN-II): Pathological and therapeutical implications. Curr. Med. Chem. 2013, 20, 4285–4291.
  23. Galmarini, C.M.; Graham, K.; Thomas, X.; Calvo, F.; Rousselot, P.; El Jafaari, A.; Cros, E.; Mackey, J.R.; Dumontet, C. Expression of high Km 5’-nucleotidase in leukemic blasts is an independent prognostic factor in adults with acute myeloid leukemia. Blood 2001, 98, 1922–1926.
  24. Cividini, F.; Pesi, R.; Chaloin, L.; Allegrini, S.; Camici, M.; Cros-Perrial, E.; Dumontet, C.; Jordheim, L.P.; Tozzi, M.G. The purine analog fludarabine acts as a cytosolic 5’-nucleotidase II inhibitor. Biochem. Pharm. 2015, 94, 63–68.
  25. Jordheim, L.P.; Chaloin, L. Therapeutic perspectives for cN-II in cancer. Curr. Med. Chem. 2013, 20, 4292–4303.
  26. Jordheim, L.P.; Puy, J.Y.; Cros-Perrial, E.; Peyrottes, S.; Lefebvre, I.; Perigaud, C.; Dumontet, C. Determination of the enzymatic activity of cytosolic 5’-nucleotidase cN-II in cancer cells: Development of a simple analytical method and related cell line models. Anal. Bioanal. Chem. 2015, 407, 5747–5758.
  27. Cividini, F.; Cros-Perrial, E.; Pesi, R.; Machon, C.; Allegrini, S.; Camici, M.; Dumontet, C.; Jordheim, L.P.; Tozzi, M.G. Cell proliferation and drug sensitivity of human glioblastoma cells are altered by the stable modulation of cytosolic 5’-nucleotidase II. Int. J. Biochem. Cell Biol. 2015, 65, 222–229.
  28. Pesi, R.; Petrotto, E.; Colombaioni, L.; Allegrini, S.; Garcia-Gil, M.; Camici, M.; Jordheim, L.P.; Tozzi, M.G. Cytosolic 5’-Nucleotidase II silencing in a human lung carcinoma cell line opposes cancer phenotype with a concomitant increase in p53 phosphorylation. Int. J. Mol. Sci. 2018, 19, 2115.
  29. Bricard, G.; Cadassou, O.; Cassagnes, L.E.; Cros-Perrial, E.; Payen-Gay, L.; Puy, J.Y.; Lefebvre-Tournier, I.; Tozzi, M.G.; Dumontet, C.; Jordheim, L.P. The cytosolic 5’-nucleotidase cN-II lowers the adaptability to glucose deprivation in human breast cancer cells. Oncotarget 2017, 8, 67380–67393.
  30. Careddu, M.G.; Allegrini, S.; Pesi, R.; Camici, M.; Garcia-Gil, M.; Tozzi, M.G. Knockdown of cytosolic 5’-nucleotidase II (cN-II) reveals that its activity is essential for survival in astrocytoma cells. Biochim. Biophys. Acta 2008, 1783, 1529–1535.
  31. Kulkarni, S.S.; Karlsson, H.K.; Szekeres, F.; Chibalin, A.V.; Krook, A.; Zierath, J.R. Suppression of 5’-nucleotidase enzymes promotes AMP-activated protein kinase (AMPK) phosphorylation and metabolism in human and mouse skeletal muscle. J. Biol. Chem. 2011, 286, 34567–34574.
  32. Kviklyte, S.; Vertommen, D.; Yerna, X.; Andersen, H.; Xu, X.; Gailly, P.; Bohlooly, Y.M.; Oscarsson, J.; Rider, M.H. Effects of genetic deletion of soluble 5’-nucleotidases NT5C1A and NT5C2 on AMPK activation and nucleotide levels in contracting mouse skeletal muscles. Am. J. Physiol. Endocrinol. Metab. 2017, 313, e48–e62.
  33. Jordheim, L.P. Expanding the clinical relevance of the 5’-nucleotidase cN-II/NT5C2. Purinergic Signal. 2018, 14, 321–329.
  34. Cividini, F.; Filoni, D.N.; Pesi, R.; Allegrini, S.; Camici, M.; Tozzi, M.G. IMP-GMP specific cytosolic 5’-nucleotidase regulates nucleotide pool and prodrug metabolism. Biochim. Biophys. Acta 2015, 1850, 1354–1361.
  35. Allegrini, S.; Filoni, D.N.; Galli, A.; Collavoli, A.; Pesi, R.; Camici, M.; Tozzi, M.G. Expression of bovine cytosolic 5’-Nucleotidase (cN-II) in yeast: Nucleotide pools disturbance and its consequences on growth and homologous recombination. PLoS ONE 2013, 8, e63914.
  36. Gakis, C. Adenosine deaminase (ADA) isoenzymes ADA1 and ADA2: Diagnostic and biological role. Eur. Respir. J. 1996, 9, 632–633.
  37. Franco, R.; Casado, V.; Ciruela, F.; Saura, C.; Mallol, J.; Canela, E.I.; Lluis, C. Cell surface adenosine deaminase: Much more than an ectoenzyme. Prog. Neurobiol. 1997, 52, 283–294.
  38. Moreno, E.; Canet, J.; Gracia, E.; Lluis, C.; Mallol, J.; Canela, E.I.; Cortes, A.; Casado, V. Molecular evidence of adenosine deaminase linking adenosine A2A receptor and CD26 proteins. Front. Pharm. 2018, 9, 106.
  39. Biri, H.; Ozturk, S.; Kacmaz, M.; Karaca, K.; Tokucoglu, H.; Durak, I. Activities of DNA turnover and free radical metabolizing enzymes in cancerous human prostate tissue. Cancer Investig. 1999, 17, 314–319.
  40. Namiot, Z.; Stasiewicz, J.; Namiot, A.; Kemona, A.; Kralisz, M.; Gorski, J. Adenosine deaminase activity in patients with the intestinal type of gastric carcinoma. Cancer Lett. 1996, 109, 199–202.
  41. Specchia, G.; Pavone, V.; Maggio, F.; Lojudice, L.; Iacobazzi, A.; Detullio, L.; Cagnazzo, G.; Liso, V. Adenosine-Deaminase activity in peripheral lymphocytes of patients with gynecologic malignancies. Boll. Inst. Sieroter. Milan. 1985, 64, 404–407.
  42. Sufrin, G.; Tritsch, G.L.; Mittelman, A.; Moore, R.H.; Murphy, G.P. Adenosine-Deaminase activity in patients with renal adenocarcinoma. Cancer 1977, 40, 796–802.
  43. Dasmahapatra, K.S.; Hill, H.Z.; Dasmahapatra, A.; Suarez, S. Evaluation of adenosine-deaminase activity in patients with head and neck-cancer. J. Surg. Res. 1986, 40, 368–373.
  44. Kojima, O.; Majima, T.; Uehara, Y.; Yamane, T.; Fujita, Y.; Takahashi, T.; Majima, S. Alteration of adenosine-deaminase levels in peripheral-blood lymphocytes of patients with gastric-cancer. Jpn. J. Surg. 1985, 15, 130–133.
  45. Russo, M.; Giancane, R.; Apice, G.; Galanti, B. Adenosine-Deaminase and purine nucleoside phosphorylase activities in peripheral lymphocytes from patients with solid tumors. Br. J. Cancer 1981, 43, 196–200.
  46. Murray, J.L.; Perezsoler, R.; Bywaters, D.; Hersh, E.M. Decreased adenosine-deaminase (Ada) and 5’nucleotidase (5nt) activity in peripheral-blood T-Cells in Hodgkin disease. Am. J. Hematol. 1986, 21, 57–66.
  47. Camici, M.; Tozzi, M.G.; Allegrini, S.; Delcorso, A.; Sanfilippo, O.; Daidone, M.G.; Demarco, C.; Ipata, P.L. Purine salvage enzyme-activities in normal and neoplastic human tissues. Cancer Biochem. Bioph. 1990, 11, 201–209.
  48. Aghaei, M.; Karami-Tehrani, F.; Salami, S.; Atri, M. Adenosine deaminase activity in the serum and malignant tumors of breast cancer: The assessment of isoenzyme ADA1 and ADA2 activities. Clin. Biochem. 2005, 38, 887–891.
  49. Mahajan, M.; Tiwari, N.; Sharma, R.; Kaur, S.; Singh, N. Oxidative stress and its relationship with adenosine deaminase activity in various stages of breast cancer. Indian J. Clin. Biochem. 2013, 28, 51–54.
  50. Durak, I.; Beduk, Y.; Kavutcu, M.; Suzer, O.; Yaman, O.; Ozturk, H.S.; Canbolat, O.; Ulutepe, S. Activity of the enzymes participating in purine metabolism of cancerous and noncancerous human kidney tissues. Cancer Invest. 1997, 15, 212–216.
  51. Eroglu, A.; Canbolat, O.; Demirci, S.; Kocaoglu, H.; Eryavuz, Y.; Akgul, H. Activities of adenosine deaminase and 5 ‘-nucleotidase in cancerous and noncancerous human colorectal tissues. Med. Oncol. 2000, 17, 319–324.
  52. Pirincci, N.; Gecit, I.; Gunes, M.; Yuksel, M.B.; Kaba, M.; Tanik, S.; Demir, H.; Aslan, M. Serum adenosine deaminase, catalase and carbonic anhydrase activities in patients with bladder cancer. Clinics 2012, 67, 1443–1446.
  53. Urunsak, I.F.; Gulec, U.K.; Paydas, S.; Seydaoglu, G.; Guzel, A.B.; Vardar, M.A. Adenosine deaminase activity in patients with ovarian neoplasms. Arch. Gynecol. Obs. 2012, 286, 155–159.
  54. Sharma, S.D.; Desai, P.B.; Metgudmath, R.B. Evaluation of serum adenosine deaminase and retinol in patients with laryngeal cancer. Indian J. Pharm. Biol. Res. 2013, 1, 5.
  55. Lal, H.; Munjal, S.K.; Wig, U.; Saini, A.S. Serum enzymes in head and neck cancer III. J. Laryngol. Otol. 1987, 101, 1062–1065.
  56. Mishra, R.; Agarwal, M.K.; Chansuria, J.P. Serum adenosine deaminase levels as an index of tumor growth in head and neck malignancy. Indian J. Otolaryngol. Head Neck Surg. 2000, 52, 360–363.
  57. Ghaderi, B.; Amini, S.; Maroofi, F.; Jalali, C.; Javanmardi, M.; Roshani, D.; Abdi, M. Adenosine deaminase activity in chronic lymphocytic leukemia and healthy subjects. Iran. J. Cancer Prev. 2016, 9, e5069.
  58. Whitmore, K.V.; Gaspar, H.B. Adenosine deaminase deficiency—More than just an immunodeficiency. Front. Immunol. 2016, 7, 314.
  59. Agarwal, R.P. Recovery of 2’-deoxycoformycin-inhibited adenosine deaminase of mouse erythrocytes and leukemia L1210 in vivo. Cancer Res. 1979, 39, 1425–1427.
  60. Dohner, H.; Ho, A.D.; Thaler, J.; Stryckmans, P.; Sonneveld, P.; de Witte, T.; Lechner, K.; Lauria, F.; Bodewadt-Radzun, S.; Suciu, S.; et al. Pentostatin in prolymphocytic leukemia: Phase II trial of the European organization for research and treatment of cancer leukemia cooperative study group. J. Natl. Cancer Inst. 1993, 85, 658–662.
  61. Willis, C.R.; Goodrich, A.; Park, K.; Waselenko, J.K.; Lucas, M.; Reese, A.; Diehl, L.F.; Grever, M.R.; Byrd, J.C.; Flinn, I.W. A phase I/II study examining pentostatin, chlorambucil, and theophylline in patients with relapsed chronic lymphocytic leukemia and non-Hodgkin’s lymphoma. Ann. Hematol. 2006, 85, 301–307.
  62. Kay, N.E.; LaPlant, B.R.; Pettinger, A.M.; Call, T.G.; Leis, J.F.; Ding, W.; Parikh, S.A.; Conte, M.J.; Bowen, D.A.; Shanafelt, T.D. Cumulative experience and long term follow-up of pentostatin-based chemoimmunotherapy trials for patients with chronic lymphocytic leukemia. Expert Rev. Hematol. 2018, 11, 337–349.
  63. Tedeschi, A.; Rossi, D.; Motta, M.; Quaresmini, G.; Rossi, M.; Coscia, M.; Anastasia, A.; Rossini, F.; Cortelezzi, A.; Nador, G.; et al. A phase II multi-center trial of pentostatin plus cyclophosphamide with ofatumumab in older previously untreated chronic lymphocytic leukemia patients. Haematologica 2015, 100, e501–e504.
  64. Johnston, J.B. Mechanism of action of pentostatin and cladribine in hairy cell leukemia. Leuk. Lymphoma 2011, 52, 43–45.
  65. Hunt, S.W., 3rd; Hoffee, P.A. Adenosine deaminase from deoxycoformycin-sensitive and -resistant rat hepatoma cells. Purification and characterization. J. Biol. Chem. 1982, 257, 14239–14244.
  66. Camici, M.; Turriani, M.; Tozzi, M.G.; Turchi, G.; Cos, J.; Alemany, C.; Miralles, A.; Noe, V.; Ciudad, C.J. Purine enzyme profile in human colon-carcinoma cell-lines and differential sensitivity to deoxycoformycin and 2’-Deoxyadenosine in combination. Int. J. Cancer 1995, 62, 176–183.
  67. Bemi, V.; Tazzini, N.; Banditelli, S.; Giorgelli, F.; Pesi, R.; Turchi, C.; Mattana, A.; Sgarrella, F.; Tozzi, M.G.; Camici, M. Deoxyadenosine metabolism in a human colon-carcinoma cell line (LoVo) in relation to its cytotoxic effect in combination with deoxycoformycin. Int. J. Cancer 1998, 75, 713–720.
  68. Giannecchini, M.; D’Innocenzo, B.; Pesi, R.; Sgarrella, F.; Iorio, M.; Collecchi, P.; Tozzi, M.G.; Camici, M. 2 ‘-deoxyadenosine causes apoptotic cell death in a human colon carcinoma cell line. J. Biochem. Mol. Toxic 2003, 17, 329–337.
  69. Garcia-Gil, M.; Tozzi, M.G.; Allegrini, S.; Folcarelli, S.; Della Sala, G.; Voccoli, V.; Colombaioni, L.; Camici, M. Novel metabolic aspects related to adenosine deaminase inhibition in a human astrocytoma cell line. Neurochem. Int. 2012, 60, 523–532.
  70. Garcia-Gil, M.; Tozzi, M.G.; Balestri, F.; Colombaioni, L.; Camici, M. Mitochondrial damage and apoptosis induced by adenosine deaminase inhibition and deoxyadenosine in human neuroblastoma cell lines. J. Cell Biochem. 2016, 117, 1671–1679.
  71. Garcia-Gil, M.; Tozzi, M.G.; Varani, S.; Della Verde, L.; Petrotto, E.; Balestri, F.; Colombaioni, L.; Camici, M. The combination of adenosine deaminase inhibition and deoxyadenosine induces apoptosis in a human astrocytoma cell line. Neurochem. Int. 2015, 80, 14–22.
  72. Thirupathi, A.; Chang, Y.Z. Role of AMPK and its molecular intermediates in subjugating cancer survival mechanism. Life Sci. 2019, 227, 30–38.
  73. Saito, M.; Yaguchi, T.; Yasuda, Y.; Nakano, T.; Nishizaki, T. Adenosine suppresses CW2 human colonic cancer growth by inducing apoptosis via A(1) adenosine receptors. Cancer Lett. 2010, 290, 211–215.
  74. Sai, K.; Yang, D.; Yamamoto, H.; Fujikawa, H.; Yamamoto, S.; Nagata, T.; Saito, M.; Yamamura, T.; Nishizaki, T. A(1) adenosine receptor signal and AMPK involving caspase-9/-3 activation are responsible for adenosine-induced RCR-1 astrocytoma cell death. Neurotoxicology 2006, 27, 458–467.
  75. Chen, Y.; Yang, S.H.; Hueng, D.Y.; Syu, J.P.; Liao, C.C.; Wu, Y.C. Cordycepin induces apoptosis of C6 glioma cells through the adenosine 2A receptor-p53-caspase-7-PARP pathway. Chem. Biol. Interact. 2014, 216, 17–25.
  76. Merighi, S.; Mirandola, P.; Milani, D.; Varani, K.; Gessi, S.; Klotz, K.N.; Leung, E.; Baraldi, P.G.; Borea, P.A. Adenosine receptors as mediators of both cell proliferation and cell death of cultured human melanoma cells. J. Invest. Derm. 2002, 119, 923–933.
  77. Tamura, K.; Kanno, T.; Fujita, Y.; Gotoh, A.; Nakano, T.; Nishizaki, T. A(2a) adenosine receptor mediates HepG2 cell apoptosis by downregulating Bcl-X(L) expression and upregulating Bid expression. J. Cell Biochem. 2012, 113, 1766–1775.
  78. Yasuda, Y.; Saito, M.; Yamamura, T.; Yaguchi, T.; Nishizaki, T. Extracellular adenosine induces apoptosis in Caco-2 human colonic cancer cells by activating caspase-9/-3 via A(2a) adenosine receptors. J. Gastroenterol. 2009, 44, 56–65.
  79. Hajiahmadi, S.; Panjehpour, M.; Aghaei, M.; Shabani, M. Activation of A2b adenosine receptor regulates ovarian cancer cell growth: Involvement of Bax/Bcl-2 and caspase-3. Biochem. Cell Biol. 2015, 93, 321–329.
  80. Jafari, S.M.; Joshaghani, H.R.; Panjehpour, M.; Aghaei, M. A2B adenosine receptor agonist induces cell cycle arrest and apoptosis in breast cancer stem cells via ERK1/2 phosphorylation. Cell Oncol. 2018, 41, 61–72.
  81. Abedi, H.; Aghaei, M.; Panjehpour, M.; Hajiahmadi, S. Mitochondrial and caspase pathways are involved in the induction of apoptosis by IB-MECA in ovarian cancer cell lines. Tumour Biol. 2014, 35, 11027–11039.
  82. Jafari, S.M.; Panjehpour, M.; Aghaei, M.; Joshaghani, H.R.; Enderami, S.E. A3 adenosine receptor agonist inhibited survival of breast cancer stem cells via GLI-1 and ERK1/2 pathway. J. Cell Biochem. 2017, 118, 2909–2920.
  83. Cohen, S.; Stemmer, S.M.; Zozulya, G.; Ochaion, A.; Patoka, R.; Barer, F.; Bar-Yehuda, S.; Rath-Wolfson, L.; Jacobson, K.A.; Fishman, P. CF102 an A3 adenosine receptor agonist mediates anti-tumor and anti-inflammatory effects in the liver. J. Cell. Physiol. 2011, 226, 2438–2447.
  84. Kanno, T.; Gotoh, A.; Fujita, Y.; Nakano, T.; Nishizaki, T. A(3) adenosine receptor mediates apoptosis in 5637 human bladder cancer cells by G(q) protein/PKC-dependent AIF upregulation. Cell. Physiol. Biochem. 2012, 30, 1159–1168.
  85. Kanno, T.; Nakano, T.; Fujita, Y.; Gotoh, A.; Nishizaki, T. Adenosine induces apoptosis in SBC-3 human lung cancer cells through A(3) adenosine receptor-dependent AMID upregulation. Cell. Physiol. Biochem. 2012, 30, 666–677.
  86. Nagaya, H.; Gotoh, A.; Kanno, T.; Nishizaki, T. A3 adenosine receptor mediates apoptosis in in vitro RCC4-VHL human renal cancer cells by up-regulating AMID expression. J. Urol. 2013, 189, 321–328.
  87. Aghaei, M.; Panjehpour, M.; Karami-Tehrani, F.; Salami, S. Molecular mechanisms of A3 adenosine receptor-induced G1 cell cycle arrest and apoptosis in androgen-dependent and independent prostate cancer cell lines: Involvement of intrinsic pathway. J. Cancer Res. Clin. Oncol. 2011, 137, 1511–1523.
  88. Otsuki, T.; Kanno, T.; Fujita, Y.; Tabata, C.; Fukuoka, K.; Nakano, T.; Gotoh, A.; Nishizaki, T. A3 adenosine receptor-mediated p53-dependent apoptosis in Lu-65 human lung cancer cells. Cell. Physiol. Biochem. 2012, 30, 210–220.
  89. Kim, S.G.; Ravi, G.; Hoffmann, C.; Jung, Y.J.; Kim, M.; Chen, A.; Jacobson, K.A. p53-Independent induction of Fas and apoptosis in leukemic cells by an adenosine derivative, Cl-IB-MECA. Biochem. Pharm. 2002, 63, 871–880.
  90. Jiang, X.; Tan, H.Y.; Teng, S.; Chan, Y.T.; Wang, D.; Wang, N. The role of AMP-Activated protein kinase as a potential target of treatment of hepatocellular carcinoma. Cancers 2019, 11, 647.
  91. Tsuchiya, A.; Nishizaki, T. Anticancer effect of adenosine on gastric cancer via diverse signaling pathways. World J. Gastroenterol. 2015, 21, 10931–10935.
  92. Saitoh, M.; Nagai, K.; Nakagawa, K.; Yamamura, T.; Yamamoto, S.; Nishizaki, T. Adenosine induces apoptosis in the human gastric cancer cells via an intrinsic pathway relevant to activation of AMP-Activated protein kinase. Biochem. Pharmacol. 2004, 67, 2005–2011.
  93. Nogi, Y.; Kanno, T.; Nakano, T.; Fujita, Y.; Tabata, C.; Fukuoka, K.; Gotoh, A.; Nishizaki, T. AMP converted from intracellularly transported adenosine upregulates p53 expression to induce malignant pleural mesothelioma cell apoptosis. Cell. Physiol. Biochem. 2012, 30, 61–74.
  94. Zuckerman, V.; Wolyniec, K.; Sionov, R.V.; Haupt, S.; Haupt, Y. Tumour suppression by p53: The importance of apoptosis and cellular senescence. J. Pathol. 2009, 219, 3–15.
  95. Nakajima, Y.; Kanno, T.; Nagaya, T.; Kuribayashi, K.; Nakano, T.; Gotoh, A.; Nishizaki, T. Adenosine deaminase inhibitor EHNA exhibits a potent anticancer effect against malignant pleural mesothelioma. Cell. Physiol. Biochem. 2015, 35, 51–60.
  96. Haynes, J.; Killilea, D.W.; Peterson, P.D.; Thompson, W.J. Erythro-9-(2-hydroxy-3-nonyl)adenine inhibits cyclic-3’,5’-guanosine monophosphate-stimulated phosphodiesterase to reverse hypoxic pulmonary vasoconstriction in the perfused rat lung. J. Pharm. Exp. 1996, 276, 752–757.
  97. Tsuchiya, A.; Kanno, T.; Saito, M.; Miyoshi, Y.; Gotoh, A.; Nakano, T.; Nishizaki, T. Intracellularly transported adenosine induces apoptosis in MCF-7 human breast cancer cells by accumulating AMID in the nucleus. Cancer Lett. 2012, 321, 65–72.
  98. Bano, D.; Prehn, J.H.M. Apoptosis-Inducing factor (AIF) in physiology and disease: The tale of a repented natural born killer. EBioMedicine 2018, 30, 29–37.
  99. Yang, D.; Yaguchi, T.; Nagata, T.; Gotoh, A.; Dovat, S.; Song, C.; Nishizaki, T. AMID mediates adenosine-induced caspase-independent HuH-7 cell apoptosis. Cell Physiol. Biochem. 2011, 27, 37–44.
  100. Hermes, M.; Osswald, H.; Kloor, D. Role of S-adenosylhomocysteine hydrolase in adenosine-induced apoptosis in HepG2 cells. Exp. Cell Res. 2007, 313, 264–283.
  101. Kutryb-Zajac, B.; Koszalka, P.; Mierzejewska, P.; Bulinska, A.; Zabielska, M.A.; Brodzik, K.; Skrzypkowska, A.; Zelazek, L.; Pelikant-Malecka, I.; Slominska, E.M.; et al. Adenosine deaminase inhibition suppresses progression of 4T1 murine breast cancer by adenosine receptor-dependent mechanisms. J. Cell Mol. Med. 2018, 22, 5939–5954.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 375
Revisions: 2 times (View History)
Update Date: 29 Sep 2021
1000/1000