334
Views
15
CrossRef citations to date
0
Altmetric
Original

EFFECT OF PENTOXIFYLLINE ON ISCHEMIC ACUTE RENAL FAILURE IN RABBITS

, , , , &
Pages 757-772 | Published online: 07 Jul 2009

Abstract

Previous studies have demonstrated that levels of tumor necrosis factor-α (TNF-α) or its mRNA expression are increased in acute renal failure of various types including ischemia/reperfusion injury. This study was undertaken to determine whether pentoxifylline (PTX), an inhibitor of TNF-α production, provides a protective effect against ischemic acute renal failure in rabbits. Renal ischemia was induced by clamping bilateral renal arteries for 60 min. Animals were pretreated with PTX (30 mg/kg, i.v.) 10 min before release of clamp. At 24 h of reperfusion of blood after ischemia, changes in renal function, renal blood flow, and the expression of TNF-α mRNA were evaluated. Ischemia/reperfusion caused a marked reduction in GFR, which was accompanied by an increase of serum creatinine levels. Such changes were significantly attenuated by PTX pretreatment. PTX ameliorated the impairment of renal tubular function, but it had no effect on the reduction of renal blood flow induced by ischemia/reperfusion. The protective effect of PTX on functional changes was supported by morphological studies. The impairment of glucose and phosphate reabsorption in postischemic kidneys was associated with a depression in the expression of Na+-glucose and Na+-Pi transporters. The expression of TNF-α mRNA was increased after reperfusion, which was inhibited by PTX pretreatment. The PTX pretreatment in vitro prevented the release of lactate dehydrogenase induced by an oxidant t-butylhydroperoxide in rabbit renal cortical slices, but it did not produce any effect on the oxidant-induced lipid peroxidation, suggesting that PTX protection is not resulted from its antioxidant action. These results suggest that PTX may exert a protective effect against ischemic acute renal failure by inhibiting the production of TNF-α in rabbits.

INTRODUCTION

Tumor necrosis factor-α (TNF-α) is a cytokine with pleiotropic actions that participates in inflammation and immunity. Previous studies have demonstrated that levels of TNF-α or its mRNA expression are increased in acute nephrosis induced by aminonucleoside Citation[[1]] and adriamycin Citation[[2]], immune glomerulonephritis Citation[3-4], diabetic nephropathy Citation[[5]], and endotoxin-induced acute renal failure Citation[[6]]. These reports suggest that TNF-α is linked to the pathogenesis of diverse renal diseases. TNF-α could be originated from both infiltrating monocytes and macrophages and resident glomerular cells. In vitro studies have shown that cultured rat and mouse mesangial cells produce TNF-α when exposed to adriamycin and puromycin Citation[[2]]. Therefore, recent interest has focused on the participation of TNF-α in the mechanisms responsible for initiation and progression of glomerular injury Citation[[6]].

Despite numerous experimental and clinical studies on the ischemic acute renal failure, the mechanisms underlying renal dysfunction following renal ischemia have not been clearly understood. One of the main functional changes in patients and experimental animals with ischemic acute renal failure is a decrease in glomerular filtration rate (GFR). The pathophysiologic mechanisms for the initiation and maintenance of the filtration failure have not been clearly elucidated Citation[[7]]. Cytokines such as TNF-α may play a critical role in ischemic acute renal failure, since TNF-α has been implicated in the pathogenesis of glomerular injury Citation[[6]].

Pentoxifylline (PTX) is a methylxanthine derivative with multiple hemorrheologic properties that has favorable effects on microcirculatory blood flow, but the precise mechanisms of its pharmacological action are not understood. Clinically, PTX has been used to treat intermittent claudication Citation[[8]]. In addition, PTX has been known to decrease serum TNF-α levels Citation[[5]], Citation[[9]] and TNF-α mRNA expression Citation[[10]]. This study was therefore undertaken to examine the effect of PTX on alterations in renal function in rabbits subjected to ischemia/reperfusion in order to evaluate the role of TNF-α in ischemic acute renal failure.

MATERIALS AND METHODS

Experimental Protocol

New Zealand White male rabbits weighing 1.5–2.5 kg were used. Anesthesia was induced by intramuscular injection of a mixture of ketamine (15 mg/kg) and xylazine (2.5 mg/kg) and maintained by sustaining doses (ketamine 5 mg/kg and xylazine 1 mg/kg) at intervals of approximately 30 min. A ventral midline incision was made and both renal arteries were clamped with nontraumatic vascular clamps for 60 min. The incision was closed and the animal was allowed to recover. Hartman solution was infused at a rate of 0.5 mL/min into an ear vein throughout the entire experimental period. After completion of surgery and recovery from anesthesia, the rabbits were returned to their metabolic cages. Urine was collected for 24 h prior to the induction of ischemia (the basal period) and for 24 h after ischemia (the postischemic period). On the same day, blood sample was collected from the ear artery. After 24 h of reflow, animals were sacrificed and the isolated kidneys were used for measurement of p-aminohippurate (PAH) uptake, Na+-K+-ATPase activity, and reduced glutathione (GSH). In experiments for PTX effect, the animals received PTX (30 mg/kg, i.v.) 10 min before the reflow of blood. The control animals untreated with the drug received an equal volume of saline.

Analysis and Calculation

Urine and blood samples were analyzed for Na+ (flame photometer, Beckman), creatinine, blood urea nitrogen (BUN), glucose, and phosphate (Sigma Chemical, St. Louis, MO). Glomerular filtration rate (GFR) was estimated from the endogenous creatinine clearance. Fractional Na+ excretion was calculated in the standard fashion. Alterations of renal function in postischemic kidney were estimated by comparing with the basal period of the same animal.

Renal blood flow was measured from a renal artery with a flowmeter (Transonic System Inc., NY) immediately before ischemia (basal) and 24 h after reflow.

Na+-K+-ATPase Activity Measurement

The renal cortical microsomal Na+-K+-ATPase activity was measured as described previously Citation[[11]]. The microsomal fraction was prepared from cortex of postischemic kidneys. The ATPase activity of the microsomal fraction was determined by measuring inorganic phosphate (Pi) released by ATP hydrolysis during incubation of microsome with an appropriate medium containing 3 mM ATP (Sigma) as the substrate. The total ATPase activity was determined in the presence of 100 mM Na+, 20 mM K+, 3 mM Mg2+, 2 mM EDTA, and 40 mM imidazole (pH 7.4). The Mg2+-ATPase activity was determined in the absence of K+ and in the presence of 1 mM ouabain. The difference between the total and the Mg2+-ATPase activity was taken as a measure of the Na+-K+-ATPase activity. After a 5-min preincubation at 37°C, the reaction was initiated by the addition of the microsomal fraction. At the end of a 10-min incubation, the reaction was terminated by the addition of ice-cold 6% perchloric acid. The mixture was then centrifuged at 3,500 g, and Pi in the supernatant fraction was determined by the method of Fiske and SubbaRow Citation[[12]].

Preparation of Renal Cortical Slices

New Zealand white male rabbits weighing 1.5 to 2 kg were sacrificed and the kidneys were removed rapidly. The kidneys were perfused immediately through the renal artery with an ice-cold isotonic saline solution containing 140 mM NaCl, 10 mM KCl and 1.5 mM CaCl2 to remove as much blood as possible. Thin slices (0.25–0.35 mm) of renal cortex were prepared using a Stadie-Riggs microtome and were stored in an ice-cold modified Cross-Taggart medium containing 130 mM NaCl, 10 mM KCl, 1.5 mM CaCl2, 5 mM glucose and 20 mM Tris/HCl (pH 7.4).

Uptake of PAH by Renal Cortical Slices

PAH uptake by renal cortical slices was measured in control and ischemic kidneys. After 24 h of reperfusion, both kidneys were removed and the renal artery was immediately perfused with an ice-cold isotonic saline solution containing 140 mM NaCl, 10 mM KCl and 1.5 mM CaCl2, to remove as much blood as possible. Renal cortical slices were prepared as described above and were stored in an ice-cold modified Cross-Taggart medium containing 130 mM NaCl, 10 mM KCl, 1.5 mM CaCl2, 5 mM Na acetate and 20 mM Tris/HCl (pH 7.8). Approximately 50 mg (wet wt.) of slices were then transferred into a 20 mL beaker containing 4 mL of the modified Cross-Taggart medium, and incubated with 75 μM 14C-PAH (Amersham, Alrington heights, IL). Incubation was carried out for 60 min in a Dubnoff metabolic shaker at 25°C under a 100% oxygen atmosphere. Immediately after incubation, the slices were quickly removed from the beaker, blotted, weighed and solubilized in 1 N NaOH. Aliquots of the incubation medium and the solubilized tissue were pipetted into a scintillation vial containing scintillation cocktail and the radioactivity was determined using a liquid scintillation counter (Packard Tricarb 300C). PAH uptake by renal slices was expressed as the slice to medium (S/M) ratio: the concentration of the compound in the tissue (mole/g wet tissue) divided by that in the medium (mole/mL medium).

Morphological Studies

Morphological evaluation was performed in kidneys subjected to 24 h reperfusion of blood following 60 min of ischemia. Kidneys were removed after intravascular perfusion with freshly prepared 1/2 Karnovsky solution. Following perfusion fixation, the samples were immersed in fixative for 4 h and processed for embedding in paraffin. Paraffin sections were stained with hematoxylin-eosin, examined, and photographed.

RNA Isolation and RT-PCR

Total cellular RNA was extracted from renal cortex using RNAzol B (Tel. Test. Inc., Friendswood, TX). RNA was quantified by spectrophotometry and formaldehyde gel electrophoresis, and stored at −80°C. Total RNA was reverse transcribed into first stand cDNA using oligo dT primer, and amplified by 35 cycles (94°C, 1 min; 52°C, 1 min; 72°C, 1 min) of polymerase chain reaction (PCR) using 20 pmole of specific primers. On completion of the PCR reaction, products were examined on 2% agarose gel. β-actin primers were used as an internal standard. The primers used for PCR are as follows:

Oxidant-Induced Injury in Renal Cortical Slices

Slices were incubated in a Cross-Taggart medium with 1.0 mM t-butylhydroperoxide (tBHP) for 60 min under a 100% O2 atmosphere. The cell injury was estimated by measuring the release of lactate dehydrogenase (LDH). After incubation, tissues were homogenized in 2 mL of distilled water and the homogenate was centrifuged at 2,000 g for 5 min. The pellet was discarded and the supernatant was saved. LDH activity was determined in the supernatant and incubation medium using a LDH assay kit (Iatron Lab., Japan).

Lipid peroxidation was estimated by measuring the renal cortical content of malondialdehyde (MDA) according to the method of Uchiyama and Mihara Citation[[13]]. Slices were homogenized in ice-cold 1.15% KCl (5% wt/vol). A 0.5 mL aliquot of homogenate was mixed with 3 mL of 1% phosphoric acid and 1 mL of 0.6% thiobarbituric acid. The mixture was heated for 45 min on a boiling water bath. After addition of 4 mL of n-butanol the contents were vigorously vortexed and centrifuged at 2,000 g for 20 min. The absorbency of the upper, organic layer was measured at 535 and 520 nm with a diode array spectrophotometer (Hewlett Packard, 8452A), and compared with freshly prepared malondialdehyde tetraethylacetal standards. MDA values were expressed as pmoles per mg protein. Protein was measured by the method of Bradford Citation[[14]].

Statistical Analysis

The data are expressed as mean ± SEM and the difference between two groups was evaluated using Student's t-test. A probability level of 0.05 was used to establish significance.

RESULTS

Effect of PTX Pretreatment on Renal Function in Rabbits with Ischemic Acute Renal Failure

summarizes changes in renal function in rabbits with ischemic acute renal failure treated and untreated with PTX. The serum creatinine level increased significantly after ischemia (3.20 ± 0.34 vs. 0.82 ± 0.12 mg/dL in the basal period). Similarly, the BUN level after renal ischemia was more than 4-fold greater than that observed in the basal period (146.84 ± 15.04 vs. 46.68 ± 7.38 mg/dL in the basal period). The PTX pretreatment significantly reduced the levels of serum creatinine and BUN after renal ischemia. PTX also attenuated the reduction of GFR in postischemic kidney. The average postischemic level of GFR in PTX-untreated animals was 0.09 ± 0.30 L/d/kg, which was less than 4% of the basal (2.34 ± 0.18 L/d/kg). The corresponding value in PTX-pretreated animals was 0.33 ± 0.06 L/d/kg, approximately 13.4% of the basal value (2.46 ± 0.03 L/d/kg). Renal ischemia induced an increase in fractional Na+ excretion (FENa) 5.5-fold in PTX-unpretreated animals (5.69 ± 0.18 vs. 1.03 ± 0.15% in the basal period), but it increased the FENa only 3.1-fold in PTX-pretreated animals (3.12 ± 0.02 vs. 1.00 ± 0.05% in the basal period).

Table 1. Effect of Pentoxifylline (PTX) on Renal Function in Rabbits with Ischemic Acute Renal Failure

Renal ischemia induced a marked increase in fractional excretion of glucose and inorganic phosphate, and the effect was significantly attenuated in PTX-pretreated animals (). PAH uptake by renal cortical slices and Na+-K+-ATPase activity in microsomal fraction prepared from cortical tissues were dramatically reduced by renal ischemia, and the reduction was significantly less in tissue preparation of PTX-pretreated animals ( and ). The depletion of GSH content in renal tissue by ischemia was remarkedly prevented by PTX pretreatment ().

Figure 1. Effect of pentoxifylline (PTX) on fractional excretion of glucose (FEGlucose, A) and inorganic phosphate (FEPi, B) in rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with the respective basal value; #p < 0.05 compared with ischemia alone.

Figure 1. Effect of pentoxifylline (PTX) on fractional excretion of glucose (FEGlucose, A) and inorganic phosphate (FEPi, B) in rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with the respective basal value; #p < 0.05 compared with ischemia alone.

Figure 2. Effect of pentoxifylline (PTX) on p-aminohippurate (PAH) uptake by renal cortical slices in rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with control; #p < 0.05 compared with ischemia alone.

Figure 2. Effect of pentoxifylline (PTX) on p-aminohippurate (PAH) uptake by renal cortical slices in rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with control; #p < 0.05 compared with ischemia alone.

Figure 3. Effect of pentoxifylline (PTX) on Na+-K+-ATPase activity in microsomal fraction from kidney cortex of rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with control; #p < 0.05 compared with ischemia alone.

Figure 3. Effect of pentoxifylline (PTX) on Na+-K+-ATPase activity in microsomal fraction from kidney cortex of rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with control; #p < 0.05 compared with ischemia alone.

Figure 4. Effect of pentoxifylline (PTX) on glutathione (GSH) content in kidney cortex of rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with control; #p < 0.05 compared with ischemia alone.

Figure 4. Effect of pentoxifylline (PTX) on glutathione (GSH) content in kidney cortex of rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with control; #p < 0.05 compared with ischemia alone.

Effect of PTX on Renal Blood Flow

shows changes of renal blood flow in postischemic kidneys. The renal blood flow 24 h after reperfusion was markedly reduced, and the value was identical in PTX-pretreated and unpretreated animals.

Figure 5. Effect of pentoxifylline (PTX) on renal blood flow in rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with the respective basal value.

Figure 5. Effect of pentoxifylline (PTX) on renal blood flow in rabbits with ischemic acute renal failure. Data are mean ± SEM of nine animals in each group. *p < 0.05 compared with the respective basal value.

Effect of PTX on Morphological Changes

Morphological evaluation was performed in postischemic kidneys. A shows proximal tubules of control kidneys with normal microvilli appearance in brush border (arrows). When kidneys were subjected to ischemia, most of brush borders in the proximal tubule (T) were destructed (B). However, most of the brush borders of proximal tubules (T) in PTX-pretreated animals were regularly arranged (arrows) as in control kidneys (Fig.z C).

Figure 6. Morphological studies of pentoxifylline (PTX) effect on ischemic acute renal failure in rabbits. Samples were stained with hematoxylin-eosin. In control kidney (A), the proximal tubule shows normal structural appearance with plentiful brush border villi (arrows). In kidneys subjected to ischemia/reperfusion injury (B), brush borders of proximal tubule (T) were destructed. However, when kidneys were subjected to ischemia/reperfusion after PTX pretreatment (C), brush borders (arrows) of proximal tubule were regularly arranged as in control. Magnitude × 132.

Figure 6. Morphological studies of pentoxifylline (PTX) effect on ischemic acute renal failure in rabbits. Samples were stained with hematoxylin-eosin. In control kidney (A), the proximal tubule shows normal structural appearance with plentiful brush border villi (arrows). In kidneys subjected to ischemia/reperfusion injury (B), brush borders of proximal tubule (T) were destructed. However, when kidneys were subjected to ischemia/reperfusion after PTX pretreatment (C), brush borders (arrows) of proximal tubule were regularly arranged as in control. Magnitude × 132.

Effect of PTX on Na+-glucose, Na+-Pi, and TNF-α mRNA

In previous study, we observed that ischemia causes marked reduction in reabsorption of glucose and phosphate in rabbits Citation[[15]]. However, it was not determined whether such changes are associated with alterations in the expression of transporters. As shown in (upper panel), the expression of mRNA for Na+-glucose and Na+-Pi transporters was suppressed after ischemia. However, the expression of mRNA was increased in PTX-pretreated animals.

Figure 7. Effect of pentoxifylline (PTX) on expression levels of Na+-glucose (SG), Na+-phosphate (SP), TNF-α, and β-actin. Total RNAs were isolated from the cortex of kidneys subjected to ischemia/reperfusion with or without PTX pretreatment. Lane 1, 4, 7: control kidneys; Lane 2, 5, 8: ischemia; Lane 3, 6, 9; ischemia + PTX.

Figure 7. Effect of pentoxifylline (PTX) on expression levels of Na+-glucose (SG), Na+-phosphate (SP), TNF-α, and β-actin. Total RNAs were isolated from the cortex of kidneys subjected to ischemia/reperfusion with or without PTX pretreatment. Lane 1, 4, 7: control kidneys; Lane 2, 5, 8: ischemia; Lane 3, 6, 9; ischemia + PTX.

In order to evaluate the effect of PTX on TNF-α expression in kidneys with ischemic acute renal failure, the expression of TNF-α mRNA was measured in kidneys subjected to ischemia with or without PTX pretreatment. The expression of TNF-α mRNA was increased by ischemia/reperfusion and this was prevented by PTX (, lower panel).

Effect of PTX In Vitro on Oxidant-Induced Lipid Peroxidation in Renal Cortical Slices

Since PTX has been reported to inhibit the generation of reactive oxygen species Citation[[16]] and scavenge hydroxyl radicals Citation[[17]], the protective effect of PTX against the ischemia/reperfusion injury may be attributed to an inhibition of lipid peroxidation. To test this possibility, we examined the effect of PTX on the lipid peroxidation induced by an oxidant tBHP. As shown in , the tBHP-induced lipid peroxidation was not altered by PTX, but it was completely inhibited by 5 μM N,N′-diphenyl-p-phenylenediamine (DPPD), an antioxidant.

Table 2. Effect of Pentoxifylline (PTX) on Lipid Peroxidation Induced by t-Butylhydroperoxide (tBHP) in Renal Cortical Slices

DISCUSSION

The present study demonstrated that the administration of PTX at a single dose of 30 mg/kg intravenously 10 min before the removal of arterial clamp provides a protective effect against the impairment of renal function in postischemic kidneys. The reduction in GFR and the increases in serum creatinine level and fractional Na+ excretion associated with the ischemia/reperfusion were significantly attenuated by PTX pretreatment (). PTX also prevented the proximal tubular dysfunction induced by ischemia as indicated by a significant protection of the impairments in renal glucose and phosphate reabsorption in vivo, PAH uptake by renal cortical slices, and Na+-K+-ATPase activity in microsomal fraction (. In addition, the GSH depletion in postischemic kidneys was significantly prevented by PTX pretreatment (). Such protective effects of PTX against proximal tubular dysfunction were supported by morphological studies ().

Renal blood flow usually remains reduced for a period of time beyond the initiation phase of ischemic acute renal failure in animal models Citation[[18]]. Persistent vasoconstriction and cellular swelling cause poor blood reperfusion in the deep cortex and outer medulla after removal of the renal artery clamp Citation[[19]]. These reports suggest that persistent medullary ischemia exacerbates the injury caused by the original insult. Indeed, an improvement of blood flow during the reperfusion period by any of several maneuvers reduces the severity of ischemic acute renal failure and accelerates recovery following ischemia Citation[19-21]. PTX has been reported to stimulate the production of vasodilatory prostaglandins Citation[[22]] and to interact with adenosine receptors Citation[[23]] which have been known to increase the renal vascular resistance Citation[[24]]. Thus, the protective effect of PTX against the ischemic acute renal failure could be due to a restoration of renal blood flow. However, the present study showed that renal blood flow in the postischemic period was less than 41% of that in the basal period and was not affected by PTX pretreatment (). These results may be consistent with the notion that restoration of renal blood flow in a postischemic kidney does not necessarily restore the GFR Citation[[25]].

TNF-α is a potent proinflammatory cytokine capable of upregulating its own expression as well as the expression of other genes pivotal to the inflammatory response Citation[[26]]. Furthermore, TNF-α causes a significant cellular damage and dysfunction via direct cytotoxicity and by recruitment of neutrophils and monocytes Citation[[27]]. Ischemia/reperfusion induces TNF-α production in various cell types including the kidney in vivo Citation[27-28] and in vitro Citation[[29]]. Similarly, we found that TNF-α mRNA levels are increased in kidneys of rabbits with ischemic acute renal failure (). These data suggest that TNF-α may be an important mediator of ischemia/reperfusion injury. Modulation of TNF-α synthesis and activity might have important therapeutic implications. PTX administration has consistently been found to inhibit the production of TNF-α in animals and humans Citation[[9]], Citation[[30]] as well as to suppress TNF-α mRNA levels in in vitro studies Citation[[10]]. In the present study, PTX decreased the expression of TNF-α mRNA and prevented the reduction of Na+-glucose and Na+-phosphate transporter mRNA expression in kidneys of rabbits with ischemic acute renal failure. In view of these findings, we suggest that PTX provides a protective effect against the ischemic acute renal failure in rabbits by inhibiting the TNF-α production.

Since PTX has been shown to inhibit the generation of leukocyte-derived reactive oxygen species in humans subjected to exercise Citation[[16]] and scavenge hydroxyl radicals in vitro Citation[[17]], the beneficial effect of PTX could be associated with its antioxidant action. In a previous study, however, we did not obtain any evidence supporting the involvement of reactive oxygen species in rabbits with ischemic acute renal failure Citation[[31]], similar to others Citation[32-33]. To determine if protection by PTX was the result of its antioxidant action, we examined the effect of PTX on the LDH release and lipid peroxidation induced by an oxidant tBHP in renal cortical slices. PTX protected partially against the LDH release induced by tBHP (Fig. 8). However, the tBHP-induced lipid peroxidation was not altered by PTX (). Taken together, it is unlikely that the protective effect of PTX against ischemic acute renal failure is associated with its antioxidant action.

Another important finding in the present study is the changes in the expression of Na+-dependent transporters in the proximal tubule. Renal ischemia has been demonstrated to cause a significant impairment in glucose reabsorption in rats Citation[34-35] and rabbits Citation[[15]]. Active reabsorption of glucose in the proximal tubule is driven by Na+-dependent transport systems Citation[[36]]. However, it was not determined if ischemia/reperfusion-induced impairment in glucose reabsorption was the result of alterations in the expression of Na+-dependent transporter system. In this study, the reabsorption of glucose and phosphate in ischemia/reperfused kidneys was significantly impaired (). Such changes were accompanied by a reduction in the expression of Na+-glucose and Na+-phosphate transporters as evidenced by a decreased expression of their mRNAs (, upper panel).

In conclusion, PTX attenuated the reduction of GFR and the impairment of tubular function in rabbits with ischemic acute renal failure. Renal blood flow was decreased after ischemia/reperfusion, which was not altered by PTX pretreatment. PTX did not affect lipid peroxidation in renal cortical slices. Expression of TNF-α mRNA was increased after ischemia/reperfusion, which was inhibited by PTX. The beneficial effect of PTX on ischemic acute renal failure may be due to a suppression of the TNF-α production.

REFERENCES

  • Diamond J R, Pesek I. Glomerular tumor necrosis factor and interleukin 1 during acute aminonucleoside nephrosis. An immunohistochemical study. Lab Invest 1991; 64: 21–28
  • Egido J, Gomez C M, Ortiz A, Bustos C, Alonso J, Gomez G C, Gomez G D, Lopez A M, Plaza J, Gonzalez E. Role of tumor necrosis factor-alpha in the pathogenesis of glomerular diseases. Kidney Int 1993; 39: S59–S64
  • Tipping P G, Leong T W, Holdsworth S R. Tumor necrosis factor production by glomerular macrophages in anti-glomerular basement membrane glomerulonephritis in rabbits. Lab Invest 1991; 65: 272–279
  • Mulligan M S, Johnson K J, Todd R F, Issekutz T B, Miyasaka M, Tamatani T, Smith C W, Anderson D C, Ward P A. Requirements for leukocyte adhesion molecules in nephrotoxic nephritis. J Clin Invest 1993; 91: 577–587
  • Navarro J F, Mora C, Rivero A, Gallego E, Chahin J, Macia M, Mendez M L, Garcia J. Urinary protein excretion and serum tumor necrosis factor in diabetic patients with advanced renal failure: effects of pentoxifylline administration. Am J Kidney Dis 1999; 33: 458–463
  • Baud L, Ardaillou R. Tumor necrosis factor alpha in glomerular injury. Kidney Int 1994; Suppl 45: S32–S36
  • Myers B D, Moran S M. Hemodynamically mediated acute renal failure. N Eng J Med 1986; 314: 97–105
  • Gillings D B. Pentoxifylline and intermittent claudication: review of clinical trials and cost-effectiveness analyses. J Cardiovasc Pharmacol 25 1995; Supp l2: 544–550
  • Staudinger T, Presterl E, Graninger W, Locker G J, Knapp S, Laczika K, Klappacher G, Stoiser B, Wagner A, Tesinsky P, Kordova H, Frass M. Influence of pentoxifylline on cytokine levels and inflammatory parameters in septic shock. Intensive Care Med 1996; 22: 888–893
  • Strieter R M, Remick D G, Ward P A, Spengler R N, Lynch J P, Larrick J, Kunkel S L. Cellular and molecular regulation of tumor necrosis factor-alpha production by pentoxifylline. Biochem Biophys Res Commun 1988; 155: 1230–1236
  • Kim Y K, Lee S H, Goldinger J M, Hong S K. Effect of ethanol on organic ion transport in rabbit kidney. Toxicol Appl Pharmacol 1986; 86: 411–420
  • Fiske C H, SubbaRow Y. The colorimetric determination of phosphorus. J Biol Chem 1925; 66: 375–400
  • Uchiyama M, Mihara M. Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Anal Biochem 1978; 6: 271–278
  • Bradford M M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976; 72: 248–254
  • Kim Y K, Woo J S, Kim Y H, Jung J S, Kim B S, Lee S H. Effect of renal ischaemia on organic compound transport in rabbit kidney proximal tubule. Pharmacol Toxicol 1995; 77: 121–129
  • Ciuffetti G, Mercuri M, Ott C, Lombardini R, Paltriccia R, Lupattelli G, Santambrogio L, Mannarino E. Use of pentoxifylline as an inhibitor of free radical generation in peripheral vascular disease. Results of a double-blind placebo-controlled study. Eur J Clin Pharmacol 1991; 41: 511–515
  • Franzini E, Sellak H, Hakim J, Pasquier C. Oxidative damage to lysozyme by the hydroxyl radical: comparative effects of scavengers. Biochim Biophys Acta 1993; 1203: 11–17
  • Hostetter T H, Brenner B M. Renal circulatory and nephron function in experimental acute renal failure. Acute Renal Failure, B M Brenner, J M Lazarus. Churchill Livingstone, New York 1988; 67–90
  • Franklin W A, Ganote C E, Jennings R B. Blood reflow after renal ischemia. Effects of hypertonic mannitol on reflow and tubular necrosis after transient ischemia in the rat. Arch Pathol 1974; 98: 106–111
  • Zager R A. Alterations of intravascular volume: influence on renal susceptibility to ischemic injury. J Lab Clin Med 1986; 108: 60–69
  • Mason J, Welsch J, Torhorst J. The contribution of vascular obstruction to the functional defect that follows renal ischemia. Kidney Int 1987; 31: 65–71
  • Sinzinger H. Pentoxifylline enhances formation of prostacyclin from rat vascular and renal tissue. Prostaglandins Leukot Med 1983; 12: 217–226
  • Vadiei K, Brunner L J, Luke D R. Effects of pentoxifylline in experimental acute renal failure. Kidney Int 1989; 36: 466–470
  • Jackson E K. Renal actions of purines. Purinergic approaches in experimental therapeutics, K A Jackson, M F Jarvis. John Wiley & Sons, New York 1997; 217–250
  • Lewis R M, Rice J H, Patton M K, Barnes J L, Nickel A E, Osgood RW, Fried T, Stein J H. Renal ischemic injury in the dog: characterization and effect of various pharmacologic agents. J Lab Clin Med 1984; 104: 470–479
  • Vilcek J, Lee T H. Tumor necrosis factor. New insights into the molecular mechanisms of its multiple actions. J Biol Chem 1991; 266: 7313–7316
  • Donnahoo K K, Shames B D, Harken A H, Meldrum D R. Review article: the role of tumor necrosis factor in renal ischemia-reperfusion injury. J Uro 1999; 162: 196–203
  • Colletti L M, Remick D G, Burtch G D, Kunkel S L, Strieter R M, Campbell-DA J. Role of tumor necrosis factor-alpha in the pathophysiologic alterations after hepatic ischemia/reperfusion injury in the rat. J Clin Invest 1990; 85: 1936–1943
  • Taylor C T, Fueki N, Agah A, Hershberg R M, Colgan S P. Critical role of cAMP response element binding protein expression in hypoxia-elicited induction of epithelial tumor necrosis factor-alpha. J Biol Chem 1999; 274: 19447–19454
  • Loftis L L, Meals E A, English B K. Differential effects of pentoxifylline and interleukin-10 on production of tumor necrosis factor and inducible nitric oxide synthase by murine macrophages. J Infect Dis 1997; 175: 1008–1011
  • Kim S Y, Kim C H, Yoo H J, Kim Y K. Effects of radical scavengers and antioxidant on ischemic acute renal failure in rabbits. Renal Failure 1999; 21: 1–11
  • Gamelin L M, Zager R A. Evidence against oxidant injury as a critical mediator of postischemic acute renal failure. Am J Physiol 1988; 255: F450–F460
  • Borkan S C, Schwartz J H. Role of oxygen free radical species in in vitro models of proximal tubular ischemia. Am J Physiol 1989; 57: F114–F125
  • Johnston P A, Rennke H, Levinsky N G. Recovery of proximal tubular function from ischemic injury. Am J Physiol 1984; 246: F159–F166
  • Molitoris B A, Kinne R. Ischemia induces surface membrane dysfunction. Mechanism of altered Na+-dependent glucose transport. J Clin Invest 1987; 80: 647–654
  • Ullrich K J. Driving forces for the transport of organic solutes. Renal transport of organic substances, R Greger, F Lang, S Silbernagl. Springer-Verlag, Berlin/Heidelberg/New York 1981; 17–29

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.