A3 adenosine receptor agonist, CF102, protects against hepatic ischemia/reperfusion injury following partial hepatectomy

  • Authors:
    • Gil Ohana
    • Shira Cohen
    • Lea Rath‑Wolfson
    • Pnina Fishman
  • View Affiliations

  • Published online on: September 19, 2016     https://doi.org/10.3892/mmr.2016.5746
  • Pages: 4335-4341
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ischemia/reperfusion (IR) injury during clinical hepatic procedures is characterized by inflammatory conditions and the apoptosis of hepatocytes. Nuclear factor‑κB (NF‑κB), nitric oxide and the expression levels of inflammatory cytokines, tumor necrosis factor‑α and interleukin‑1 were observed to increase following IR and mediate the inflammatory response in the liver. CF102 is a highly selective A3 adenosine receptor (A3AR) agonist, and has been identified to induce an anti‑inflammatory and protective effect on the liver via the downregulation of the NF‑κB signaling pathway. The present study aimed to determine the effect of CF102 on protecting the liver against IR injury. The potential protective effect of CF102 (100 µg/kg) was assessed using an IR injury model on 70% of the liver of Wistar rats, which was induced by clamping the hepatic vasculature for 30 min. The regenerative effect of CF102 was assessed by the partial hepatectomy of 70% of the liver during 10 min of IR. CF102 reduced the levels of liver enzymes following IR injury. A higher regeneration rate in the CF102 treatment group was observed compared with the control group, suggesting that CF102 had a positive effect on the proliferation of hepatocytes following hepatectomy. CF102 had a protective effect on the liver of Wistar rats subsequent to IR injury during hepatectomy. This may be due to an anti‑inflammatory and anti‑apoptotic effect mediated by the A3AR.

Introduction

Ischemia/reperfusion (IR) injury occurs when the cellular damage of a hypoxic organ is increased subsequent to the restoration of oxygen delivery. Warm IR injury may arise clinically in hepatic surgery, liver transplantation, hypovolemic shock and various types of toxic liver injury. The common denominator of the aforementioned conditions is the ‘low-flow’ state induced by liver hypoxia, which may lead to ischemic hepatitis. The Pringle maneuver has been established as the occlusion of the hepatic artery and the portal vein by cross-clamping the porta hepatis with a vascular clamp. It is frequently used to control bleeding during the repair of extensive liver wounds, hepatic resection and liver transplantation. However, this may result in hepatic IR injury, in the event that the clamping the hepatic vasculature is performed for a long period of time.

Two distinct phases of liver injury have been identified following warm IR injury. The initial phase (<2 h after reperfusion) is characterized by oxidant stress, which directly results in hepatocellular injury. The late phase (6–48 h after hepatic reperfusion) is an inflammatory disorder mediated by recruited neutrophils (1).

Cellular hypoxia has been identified as the key factor that leads to the activation of the transcriptional regulator, nuclear factor-κB (NF-κB), and triggers the release of other inflammatory mediators, including activated Kupffer cells and neutrophils, tumor necrosis factor-α (TNF-α), interleukin (IL)-1 and nitric oxide (NO), possibly in order to mediate the reperfusion injury process (2,3). The production of inflammatory mediators following the activation of Kupffer cells has been determined to be an important component for the pathophysiology of neutrophil-mediated injury during liver IR (46). In addition, previous studies have identified that apoptosis in hepatocytes is increased, and a decline in their regeneration during hepatic IR injury has been observed (4,7).

The importance of apoptotic cell death in hepatic IR has been investigated previously. According to previous studies, 40–60% of hepatocytes may undergo apoptosis during IR (810).

Ischemic preconditioning is a technique in which organs are exposed to brief periods of IR in order to prepare an organ for the subsequent effects of a longer period of IR exposure. Initially, it was used for coronary ischemia and protection against myocardial infarction; however, at present, it used in various organs, including the brain, intestine, skeletal muscle and liver (11). The underlying cytoprotective mechanisms remain to be elucidated and may include an increase in the adenosine levels in the cell microenvironment, a decrease in NO levels, a reduction in TNF-α release, changes in energy metabolism and accelerated cell-cycle progression (12).

Previous studies that conducted experimental models of partial liver IR injury by 5–15 min of ischemia followed by 10 min of reperfusion led to hepatoprotective conditions, as a significant reduction in areas of hepatocyte necrosis occurred and the increase in transaminase levels was limited (13,14). A previous study determined that this narrowed hepatoprotective time window for ischemia is defined by the relative tissue concentrations of adenosine and xanthine (15). Additionally, purinergic signaling may have a protective role during hepatic IR injury (16).

Adenosine accumulates in the extracellular space following ischemia by binding to selective G-protein-associated membrane receptors, termed A1, A2A, A2B and A3, and has been established to promote cytoprotection. In a model of IR injury in mice, adenosine administration was observed to induce a protective effect on the liver (17). Additionally, the activation of the A3 adenosine receptor (A3AR) has been identified to exert cardioprotection, neuroprotection and pulmonoprotection against IR injury (1822).

Upon activation of the A3AR, specific signal transduction pathways are modulated, including the Wnt and NF-κB signaling pathways (23,24). The activation of A3AR in inflammatory cells and tissues may lead to deregulation of the NF-κB signaling pathway and may result in reduced expression levels of phosphoinositide 3-kinase (PI3K), protein kinase B/Akt (PKB/Akt), IκB kinase, IκB and NF-κB, resulting in an anti-inflammatory effect (2426). Additionally, apoptosis of inflammatory cells was observed downstream of PKB/Akt modulation, leading to upregulation of caspase 3 expression levels (27).

It is of note that activation of the A3AR resulted in increased differentiation of tumor cells compared with normal cells (28). CF102, a selective agonist to A3AR, induced the differentiation of normal and tumor cells and inhibited their proliferation, along with the induction of apoptosis in inflammatory cells, whereas it had a protective effect on normal cells (29).

Based on the aforementioned previous studies, the present study aimed to determine the ability of CF102 to protect the liver against IR injury.

Materials and methods

Reagents

The A3AR agonist, CF102, a compound also termed 2-chloro-N6-(3-iodobenzyl)-adenosine-5′-N-methyl-uronamide (Cl-IB-MECA),was synthesized for Can-Fite BioPharma (Petah Tikva, Israel) by Albany Molecular Research, Inc. (Albany, NY, USA). A stock solution of 5 mg/ml was prepared in dimethylsulphoxide and further diluted in phosphate-buffered saline (PBS).

Animals

Animal handling was conducted according to the guidelines of the National Institutes of Health (Bethesda, MD, USA) and the Association for Assessment and Accreditation of Laboratory Animal Care (Frederick, MD, USA).

Male Wistar rats (n=76; age, 10 weeks; Harlan Laboratories, Inc., Jerusalem, Israel) were used in the present study. The body weights of the rats ranged from 275 to 300 g. The animals were provided with a nutritionally balanced rodent diet and water ad libitum. They were maintained under constant environmental conditions, temperatures range was 18–24°C and relative humidity range was 30–70% with 12 h light and 12 h dark cycle. The present study was performed following approval by the Can-Fite Biopharma, Institutional Ethic committee (Petah Tikva, Israel), and in compliance with the Israel Animal Welfare Act, approval nos. 029_a123 and 029_a128.

IR injury procedure

A total of 28 male Wistar rats (275–300 g) were used for the IR injury procedure in the following groups: i) Naïve (n=4), laparatomy was performed without IR procedure; ii) control (n=12) no treatment or surgery; and iii) CF102 (n=12) laparatomy and IR were performed, followed by treatment with CF102. Rats were fasted 12 h prior to surgery, then anesthetized with ketamine (90 mg/kg) and xylazine (10 mg/kg). Laparatomy was performed via a subcostal, bilateral incision. The main portal pedicle to the total liver was clamped for a period of 30 min, and the blood flow was restored by de-clamping. Subsequently, the rats were injected subcutaneously with 100 µg/kg CF102 (in 0.1 ml PBS) during reperfusion, and the compound was orally administered three times per day thereafter. The control rats received normal saline of the identical volume. All animals received 5 ml 5% glucose and 0.2 ml penicillin subcutaneously immediately following the operation. Postoperatively, the animals were maintained on a standardized pelleted diet and were supplied with water ad libitum. The present study was conducted over the course of 48 h.

The rats were anesthetized using isoflurane (Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) 48 h after the operation, 2 ml blood samples were collected from the inferior vena cava and were centrifuged at 24, 000 × g for 15 min at 25°C. The supernatant was collected and serum aspartate aminotransferase (SGOT) and alanine aminotransferase (SGPT) levels were analyzed in the Medical Center Laboratories (Herzelia, Israel) to evaluate the extent of liver injury. The rats were sacrificed using CO2 in a gas chamber. The livers were removed and fixed in 10% buffered formalin for pathological analysis and staining.

Partial hepatectomy

A total of 48 male Wistar rats (300–350 g) were used in the present study: The control group contained 22 individuals, the CF102 group had 22 and the naive group had 4 rats. Rats were fasted 12 h prior to surgery and anesthetized by intraperitoneal injection of ketamine (45 mg/kg) and xylazine (5 mg/kg). Laparatomy was performed via a subcostal, bilateral incision. The primary portal pedicle to the liver was identified and clamped prior to the hepatectomy procedure. A period of 10 min total liver ischemia was initiated, during which a 70% hepatectomy was performed. This was achieved by resection of the median and left lateral lobes, according to the technique previously described by Higgins and Anderson (30). Following 10 min of ischemia and completion of 70% hepatectomy, blood flow to the remaining liver was restored by de-clamping. All animals received 5 ml 5% glucose and 0.2 ml penicillin subcutaneously immediately following the operation. Postoperatively, the animals were maintained on a standardized pelleted diet and were supplied with tap-water. Rats were injected subcutaneously with 100 µg/kg CF102 (in 0.1 ml PBS) during reperfusion, and the compound was administered orally three times per day thereafter. The control rats received normal saline at the identical volume. At 2, 4 and 48 h after surgery, rats were anesthetized with isoflurane and blood samples (2 ml) were collected from the rats' tails for analysis of SGOT and SGPT levels. Rats were sacrificed using CO2 after 24 and 48 h, and the liver tissues were resected and weighed in order to calculate the hepatic regeneration rate, with subsequent fixing in 10% buffered formalin for further pathological analysis and staining.

Immunohistochemical staining and histology

Liver tissue specimens were fixed in 10% formalin, embedded in paraffin and cut into 5 mm thick sections.

Hematoxylin and eosin staining was performed in order to obtain a mitotic index (MI) by dividing the number of cells undergoing mitosis by the total cell number in 30 high-power fields. Additionally, the presence of hepatic injury and necrosis was also observed.

Apoptosis was examined by observation of DNA fragmentation using the ApopTag Peroxidase in Situ Detection kit (Chemicon; EMD Millipore, Billerica, MA, USA) for a terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay following the manufacturer's protocol. Next, the sections were deparaffinized, re-hydrated and endogenous peroxidase was quenched with 20% hydrogen peroxidase in methanol for 20 min. The slides were then pretreated with proteinase K (BioVision, Inc., Milpitas, CA, USA) for 15 min at room temperature, and treated with an equilibration buffer for 10 s and terminal deoxynucleotidyl transferase enzyme (Sigma-Aldrich; Merck Millipore) for 1 h. Sections were incubated with an anti-digoxigenin conjugated antibody (cat. no. ab119345; 1:1,000; Abcam, Cambridge, UK), washed with PBS and developed with 3,3′-diaminobenzidine, counterstained with hematoxylin and eosin and then mounted.

To calculate the proliferation cell nuclear antigen (PCNA) labeling index, 5 mm sections were incubated with anti-PCNA antibody (cat. no. ab29; 1:1,000; Abcam). The proliferation of hepatocytes was calculated as the number of cells in the S-phase. PCNA-positive cells exhibited brownish-yellow or yellow nuclei. If brown particles were observed in the nucleus, the cells were considered to be positive.

Hepatic regeneration rate

The growth of residual liver lobes was assessed using the following equation: Hepatic regeneration rate (%) = C-(A-B) / A ×100, where A is the estimated total liver weight prior to the hepatectomy (A was considered to be 3.4% of a rat's total weight) (31), B is the weight of the resected liver during the hepatectomy and C is the weight of the regenerated liver at the final resection (32).

Statistical analysis

The results were evaluated using Student's t-test. Comparisons between the mean values of the different experiments were performed. Data are presented as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

CF102 attenuates liver IR injury in rats

In the present study, rats were exposed to 30 min of ischemia. The serum levels of SGOT and SGPT, which may be used as a proxy of the extent of liver damage, were significantly reduced in the CF102 treatment group when compared with the control group 48 h after surgery (P<0.05;Fig. 1).

Pathological analyses were performed in order to obtain additional qualitative assessment of liver injury. Paraffin-embedded slides of liver tissues were stained by hematoxylin and eosin 48 h after liver reperfusion. The upper panel of Fig. 2 shows healthy hepatocytes and no signs of necrosis or inflammation in the naïve group, whereas large areas of necrosis surrounded by numerous inflammatory cells and pyknotic nuclei were observed in liver tissues from the control group (middle panel). By contrast, liver tissues from the CF102-treated group did not exhibit areas of necrosis, and only a few inflammatory cells were visible (lower panel). These findings confirm that the A3AR agonist, CF102, allowed for substantial protection against IR injury from occurring (Fig. 2).

CF102 protects against IR injury in hepatectomized rats

Liver hepatectomy was performed on 70% of the rat's liver, whilst being exposed to 10 min of ischemia. Liver enzymic analysis revealed a marked increase in the levels of SGOT and SGPT 2 h after the hepatectomy, which peaked at 4 h and started to decrease after 48 h in the control group. CF102 treatment applied following reperfusion induced a reduction in the levels of SGOT and SGPT, which was maintained for 48 h (Fig. 3).

A3AR agonist enhances liver regeneration following partial hepatectomy

Hepatic regeneration rates were higher in the CF102 treatment group compared with the saline-treated control group 48 h after the hepatectomy (Fig. 4). CF102 treatment increased the regeneration rate of the remnant liver by 45% at 48 h following the 70% hepatectomy (Fig. 4). Additionally, in the CF102 treatment group, the PCNA labeling index (Fig. 5) and the MI (P=0.013; Fig. 6) were higher compared with the saline-treated control group.

Discussion

The present study determined that the selective A3AR agonist, CF102, when administered during liver reperfusion, may attenuate cellular injury, apoptosis and the extent of necrosis in liver subjected to IR. Additionally, CF102 stimulated liver regeneration, which favored the survival of hepatocytes during IR injury. A rat model combining 70% partial hepatectomy with 10 min of Pringle maneuver was used, in order to imitate the surgical procedure performed in humans. In this model, 30% of the liver remained, which was exposed to IR injury. Following partial hepatectomy and liver reperfusion, serum levels of SGOT and SGPT, which may be used as a proxy of liver damage, were reduced in the CF102 treatment group when compared with the control group at 2, 4, and 48 h after the hepatectomy. Reduced liver necrosis and cell apoptosis were observed in the remaining liver obtained from the CF102-treated rats 48 h after the operation when compared with the control group. It is of note that the remnant liver in the CF102-treated rats exhibited a 45% increase in the hepatic regeneration rate when compared with the untreated control group rats. This was confirmed, as the MI and the PCNA labeling index were higher in the CF102 treatment group compared with the control (Figs. 5 and 6). Therefore, CF102, a highly selective A3AR agonist with high receptor affinity, may be able to protect against liver IR injury at low concentrations. Previous studies have extensively reported that A3AR may be used as a target to mediate different protective functions, including neuroprotection, chemoprotection and cardioprotection (21,33,34). Auchampach et al (33) revealed that IB-MECA (CF102), administered at a concentration of 100 µg/kg intravenously 5 min prior to the onset of reperfusion, reduced the size of myocardial infarct in dogs (33). Similarly, 2-CL-IB-MECA administered during reperfusion reduced infarct size in isolated rat hearts and prevented apoptosis in isolated rat cardiomyocytes (35). The PI3K-PKB/Akt signal transduction pathway was identified to be responsible for IB-MECA-induced glycogen synthase kinase-3b inactivation, which is considered to be involved in cardioprotection (15).

Inhibition of the apoptotic response to IR injury in the lungs by the activation of A3AR has also been previously described (36). Rivo et al (36) have established a spontaneously breathing cat model where the administration of IB-MECA (300 µg/kg) during reperfusion markedly attenuated indices of injury and apoptosis (36). Adenosine has been established as a regulator of cell viability (37), and A3AR has been specifically identified to contribute to the modulation of cell survival. Activation of A3AR may induce cell protection or death, depending on the degree of receptor activation (37,38).

Activation of the A3AR has also been described to trigger neuroprotection. Chen et al (21) have determined that, when A3R knockout mice are treated with a high dose of CL-IB-MECA (0.2 mg/kg), reduced cerebral infarction through the activation of A3AR and suppression of apoptosis occurred (21).

Apoptosis has been implicated in the pathophysiology of IR liver injury (39); however, an association with A3AR activation remains to be described.

The present study determined that treatment with CF102 accelerated liver regeneration following a 70% hepatectomy and IR injury. Zhang et al (40) have determined that, in a regenerative state, the liver was protective against carbon tetrachloride-induced hepatotoxicity via a mechanism involving increased mitochondrial respiratory activity and plasma membrane fluidity (40).

Our previous study determined that activation of A3AR initiates a molecular mechanism that involves the deregulation of the PI3K-NF-κB signaling pathway, which results in an effective anti-inflammatory effect (26). Therefore, this may be the possible mechanism through which CF102 exerts its hepatoprotection against IR injury, as the late phase of IR liver injury involved an inflammatory process.

In conclusion, the ability of CF102, an A3AR agonist, to protect against hepatic IR injury, may include an anti-inflammatory and an anti-apoptotic effect, combined with an increased rate of liver regeneration. However, further studies should be conducted in order to elucidate the molecular basis for these clinically important effects of CF102.

Glossary

Abbreviations

Abbreviations:

A3AR

A3 adenosine receptor

IL-1

interleukin 1

I/R

ischemia-reperfusion injury

NF-κB

nuclear factor-κB

NO

nitric oxide

PKB/Akt

protein kinase B/Akt

PCNA

proliferation cell nuclear antigen

SGOT

aspartate aminotransferase

SGPT

alanine aminotransferase

TNF-α

tumor necrosis factor-α

References

1 

Lentsch AB, Atsushi K, Yoshidome H, MacMasters KM and Edwards MJ: Inflammatory mechanisms and therapeutic strategies for warm hepatic ischemia/reperfusion injury. Hepatology. 32:169–173. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Semenza GL: HIF-1: Mediator of physiological and pathophysiological response to hypoxia. J Appl Physiol (1985). 88:1474–1480. 2000.PubMed/NCBI

3 

Li C and Jackson RM: Reactive species mechanisms of cellular hypoxia-reoxygenation injury. Am J Physiol Cell Physiol. 282:C227–C241. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Jaeschke H: Mechanisms of reperfusion injury after warm ischemia of the liver. J Hepatobiliary Pancreat Surg. 5:402–408. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Okuaki Y, Miyazaki H, Zeniya M, Ishikawa T, Ohkawa Y, Teuno S, Sakaguchi M, Hara M, Takahashi H and Toda G: Splenectomy-reduced hepatic injury induced by ischemia/reperfusion in the rat. Liver. 16:188–194. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Zwacka RM, Zhang Y, Halldorson J, Schlossberg H, Dudus L and Ehgelhardt JF: CD4(+) T-lymphocytes mediate ischemia/reperfusion-induced inflammatory responses in mouse liver. J Clin Invest. 100:279–289. 1997. View Article : Google Scholar : PubMed/NCBI

7 

Liu P, McGuire GM, Fischer MA, Farhood A, Smith CW and Jaeschke H: Activation of Kupffer cells and neutrophils for reactive oxygen formation is responsible for endotoxin-enhanced liver injury after hepatic ischemia. Shock. 3:56–62. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Gao W, Bentely RC, Madden JF and Clavien PA: Apoptosis of sinusoidal endothelial cells is a critical mechanism of prevention of injury in rat liver transplantation. Hepatology. 27:1652–1660. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Cursio R, Gugenheim J, Ricci JE, Crenesse D, Rostagno P, Maulon L, Saint-Paul MC, Ferrua B and Auberger AP: A caspase inhibitor fully protects rats against lethal normothermic liver ischemia by inhibition of liver apoptosis. FASEB J. 13:253–261. 1999.PubMed/NCBI

10 

Kohli V, Selzner M, Madden JF, Bentley RC and Clavien PA: Endothelial cell and hepatocyte deaths occur by apoptosis after ischemia-reperfusion injury in the rat liver. Transplantation. 67:1099–1105. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Murry CE, Jenning RB and Reimer KA: Preconditioning with ischemia: A delay of lethal cell injury in ischemic myocardium. Circulation. 74:1124–1136. 1986. View Article : Google Scholar : PubMed/NCBI

12 

Teoh NC and Farrell GC: Hepatic ischemia reperfusion injury: Pathogenic mechanisms and basis for hepatoprotection. J Gastroenterol Hepatol. 18:891–902. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Peralta C, Closa D, Xaus C, Gelpi E, Roselló-Catafau J and Hotter G: Hepatic preconditioning in rats is defined by a balance of adenosine and xanthine. Hepatology. 28:768–773. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Teoh N, Pena A Dela and Farrel G: Hepatic ischemia preconditioning in mice is associated with activation of NF-kappaB, p38 kinase, and cell cycle entry. Hepatology. 36:94–102. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Gross ER, Hsu AK and Gross GJ: Opioid-induced cardioprotection occurs via glycogen synthase kinase beta inhibition during reperfusion in intact rat hearts. Circ Res. 94:960–966. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Vaughn BP, Robson SC and Longhi MS: Purinergic signaling in liver disease. Dig Dis. 32:516–24. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Sun X, Imai M, Nowak-Machen M, Guckelberger O, Enjyoj K, Wu Y, Khalpey Z, Berberat P, Munasinghe J and Robson SC: Liver damage and systemic inflammatory responses are exacerbated by the genetic deletion of CD39 in total hepatic ischemia. Purinergic Signal. 7:427–434. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Black RG Jr, Guo Y, Ge ZD, Murphree SS, Prabhu SD, Jones WK, Bolli R and Auchampach JA: Gene dosage-dependent effects of cardiac-specific overexpression of the A3 adenosine receptor. Circ Res. 91:165–172. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Cross HR, Murphy E, Black RG, Auchampach J and Steenbergen C: Overexpression of A(3) adenosine receptors decreases heart rate, preserves energetics, and protects ischemic hearts. Am J Physiol Heart Circ Physiol. 283:H1562–H1568. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Matot I, Weiniger CF, Zeira E, Galun E, Joshi BV and Jacobson KA: A3 adenosine receptors and mitogen-activated protein kinases in lung injury following in vivo reperfusion. Crit Care. 10:R652006. View Article : Google Scholar : PubMed/NCBI

21 

Chen GJ, Harvey BK, Shen H, Chou J, Victor A and Wang Y: Activation of A3 adenosine receptors reduces ischemic brain injury in rodents. J Neurosci Res. 84:1848–1855. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Borea PA, Varani K, Vincenzi F, Baraldi PG, Tabrizi MA, Merighi S and Gessi S: The A3 adenosine receptor: History and perspectives. Pharmacol Rev. 67:74–102. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Fishman P, Bar-Yehuda S, Ohana G, Barer F, Ochaion A, Erlanger A and Madi L: An agonist to the A3 adenosine receptor inhibits colon carcinoma growth in mice via modulation of GSK-3 beta and NF-kappa B. Oncogene. 23:2465–2471. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Fishman P, Madi L, Bar-Yehuda S, Barer F, Del Valle L and Khalili K: Evidence for involvement of Wnt signaling pathway in IB-MECA mediated suppression of melanoma cells. Oncogene. 21:4060–4064. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Hanada T and Yoshimura A: Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev. 13:413–421. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Fishman P, Bar-Yehuda S, Madi L, Rath-Wolfson L, Ochaion A, Cohen S and Baharav E: The PI3K-NF-kappa B signal transduction pathway is involved in mediating the anti-inflammatory effect of IB-MECA in adjuvant-induced arthritis. Arthritis Res Ther. 8:R332006. View Article : Google Scholar : PubMed/NCBI

27 

Shackelford RE, Alford BP, Xue Y, Thai SF, Adams DO and Pizzo S: Aspirin inhibits tumor necrosis factor-alpha gene expression in murine tissue macrophages. Mol Pharmacol. 52:421–429. 1997.PubMed/NCBI

28 

Ohana G, Bar-Yehuda S, Barer F and Fishman P: Differential effect of adenosine on tumor and normal cell growth: Focus on the A3 adenosine receptor. J Cell Physiol. 186:19–23. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Cohen S, Stemmer SM, Zozulya G, Ochaion A, Patoka R, Barer F, Bar-Yehuda S, Rath-Wolfson L, Jacobson KA and Fishman P: CF102 an A3 adenosine receptor agonist mediates anti-tumor and anti-inflammatory effects in the liver. J Cell Physiol. 226:2438–2447. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Higgins G and Anderson R: Experimental pathology of the liver: restoration of the liver of the white rat following partial surgical removal. Arch Pathol. 12:186–202. 1931.

31 

Kogure K, Zhang YQ, Shibata H and Kojima I: Immediate onset of DNA synthesis in remnant rat liver after 90% hepatectomy by administration of follistatin. J Hepatol. 29:977–984. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Selzner M and Clavien PA: Failure of regeneration of the steatotic rat liver: Disruption at two different levels in the regeneration pathway. Hepatology. 31:35–42. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Auchampach JA, Ge ZD, Wan TC, Moore J and Gross GJ: A3 adenosine receptor agonist IB-MECA reduces myocardial ischemia-reperfusion injury in dogs. Am J Physiol Heart Circ Physiol. 285:H607–H613. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Fishman P, Bar-Yehuda S, Barer F, Madi L, Multani AS and Pathak S: The A3 adenosine receptor as a new target for cancer therapy and chemoprotection. Exp Cell Res. 269:230–236. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Maddock HL, Mocanu MM and Yellon DM: Adenosine A(3) receptor activation protects the myocardium from reperfusion/reoxygenation injury. Am J Physiol Heart Circ Physiol. 283:H1307–H1313. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Rivo J, Zeira E, Galun E and Matot I: Activation of A3 adenosine receptors provides lung protection against ischemia reperfusion injury associated with reduction in apoptosis. Am J Transplant. 4:1941–1948. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Jacobson KA, Hoffmann C, Cattabeni F and Abbracchio MP: Adenosine-induced cell death: Evidence for receptor-mediated signaling. Apoptosis. 4:197–211. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Abbracchio MP, Ceruti S, Brambilla R, Franceschi C, Malorni W, Jacobson KA, von Lubitz DK and Cattabeni F: Modulation of apoptsis by adenosine in the central nervous system: A possible role for the A3 adenosine receptor. Pathophysiological significance and therapeutic implications for neurodegenerative disorders. Ann NY Acad Sci. 825:11–22. 1997. View Article : Google Scholar : PubMed/NCBI

39 

Borghi-Scoazec G, Scoazec JY, Durand F, Bernuau J, Belghiti J, Feldmann G, Henin D and Degott C: Apoptosis after ischemia-reperfusion in human liver allografts. Liver Transpl Surg. 3:407–415. 1997. View Article : Google Scholar : PubMed/NCBI

40 

Zhang BH, Gong DZ and Mei MH: Protection of regenerating liver after partial hepatectomy from carbon tetrachloride hepatotoxicity in rats: Role of hepatic stimulator substance. J Gastroenterol Hepatol. 14:1010–1017. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ohana G, Cohen S, Rath‑Wolfson L and Fishman P: A3 adenosine receptor agonist, CF102, protects against hepatic ischemia/reperfusion injury following partial hepatectomy. Mol Med Rep 14: 4335-4341, 2016
APA
Ohana, G., Cohen, S., Rath‑Wolfson, L., & Fishman, P. (2016). A3 adenosine receptor agonist, CF102, protects against hepatic ischemia/reperfusion injury following partial hepatectomy. Molecular Medicine Reports, 14, 4335-4341. https://doi.org/10.3892/mmr.2016.5746
MLA
Ohana, G., Cohen, S., Rath‑Wolfson, L., Fishman, P."A3 adenosine receptor agonist, CF102, protects against hepatic ischemia/reperfusion injury following partial hepatectomy". Molecular Medicine Reports 14.5 (2016): 4335-4341.
Chicago
Ohana, G., Cohen, S., Rath‑Wolfson, L., Fishman, P."A3 adenosine receptor agonist, CF102, protects against hepatic ischemia/reperfusion injury following partial hepatectomy". Molecular Medicine Reports 14, no. 5 (2016): 4335-4341. https://doi.org/10.3892/mmr.2016.5746