α-melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

10
a-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils HSI CHIAO,YUKIMASA KOHDA,PAUL MCLEROY,LEONARD CRAIG,STUART LINAS, and ROBERT A. STAR Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, and University of Colorado, Denver, Colorado, USA a-Melanocyte-stimulating hormone inhibits neutrophil-indepen- dent renal injury. Background. We previously showed that a-melanocyte stimu- lating hormone (a-MSH) decreases ischemia/reperfusion injury even when started six hours after ischemia. a-MSH inhibits both neutrophil accumulation and nitric oxide production. To deter- mine the relative importance of a-MSH on the neutrophil path- way, we examined the effects of a-MSH in injury models where neutrophil effects are minimal or absent. Methods. We studied the effects of a-MSH in (1) intercellular adhesion molecule-1 (ICAM-1) knock-out and background mice that were subjected to 40 minutes of ischemia and 24 hours reperfusion, and (2) isolated kidneys that were subjected to in vivo ischemia for 20 minutes and then perfused ex vivo for one hour without neutrophils. To begin to search for direct tubule effects of a-MSH, we studied the effect of a-MSH on nitric oxide (NO) in endotoxin/interferon-g-treated mouse cortical tubule cells. Results. ICAM-1 knock-out mice had 75% less neutrophil infiltration than background mice after ischemia. Despite the relative lack of neutrophils, a-MSH inhibited renal injury in ICAM-1 knock-out mice. a-MSH also significantly preserved GFR and tubular sodium reabsorption in the isolated perfused ischemic kidney model. a-MSH and a nitric oxide inhibitor did not exhibit synergy. Finally, a-MSH inhibited nitrite production by 20% in the mouse cortical tubule cells (MCT), similar to parallel observations in a cultured mouse macrophage line (RAW cells). Conclusions. We conclude that a-MSH decreases renal injury when neutrophil effects are minimal or absent, indicating that a-MSH inhibits neutrophil-independent pathways of renal injury. The preservation of sodium absorption ex vivo and inhibition of nitrite production in cultured MCT cells suggests that a-MSH inhibits tubular injury by direct tubular effects. Organ ischemia is the primary cause of stroke, myocar- dial infarction, and acute renal failure, and increases the risk of rejection following organ transplantation. While prolonged ischemia causes anoxic cell death, additional waves of injury initiated following sublethal injury are caused in part by maladaptive inflammatory and cytotoxic injury cascades activated during the reperfusion period. Recent studies of renal ischemia-reperfusion injury have focused on the roles of neutrophils and cytotoxic nitric oxide. Following renal ischemia, neutrophils accumulate in the outer stripe of outer medulla beginning four hours after ischemia, which is coincident with histologic evidence of ischemia [1–3]. While infiltrating neutrophils can remove necrotic and apoptotic cells and hence is a passive response to injury, many studies indicate that the neutrophils play a pathogenic role. For example, neutrophils may plug the ascending vasa recta in the outer stripe of the outer medulla, and further impair oxygen supply to the proximal straight tubule, the major site of injury in the clamp model of renal injury [2]. Support for this pathogenic role comes from studies where ischemic injury is partially inhibited by neutrophil depletion in some [4, 5], but not all [6, 7], studies. Ischemic renal injury is potentiated by infusion of activated neutrophils [8, 9] and is decreased by blocking antibodies or antisense oligonucleotides to intercellular adhesion molecule-1 (ICAM-1) or P-selectin or mice lack- ing ICAM-1 [10 –14]. The amount of protection by neutro- phil depletion/inhibition is more striking with shorter ischemia times. However, cardiac and renal ischemia- reperfusion injury can occur in the absence of neutrophils [15, 16], suggesting other mechanisms of injury. Recent studies have focused on maladaptive cytotoxic cascades activated during reperfusion that liberate high concentra- tions of cytotoxic nitric oxide (NO). Hypoxic/reperfusion injury to isolated non-perfused proximal tubules can be inhibited by blocking nitric oxide production [17]. The inducible isoform of NO synthase (iNOS) is induced by ischemia in proximal tubules. Blocking iNOS with antisense oligonucleoides protects cultured proximal tubule cells from hypoxic damage in vitro and against ischemic damage in vivo [18]. In addition to being directly cytotoxic, the high concentrations may also inhibit endothelial NO, with re- sultant increased organ ischemia [19]. Taken together, the Key words: ischemia, reperfusion, a-MSH, neutrophils, nitric oxide, ICAM-1. Received for publication January 30, 1998 and in revised form April 8, 1998 Accepted for publication April 14, 1998 © 1998 by the International Society of Nephrology Kidney International, Vol. 54 (1998), pp. 765–774 765

Upload: robert-a

Post on 06-Jul-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

a-Melanocyte-stimulating hormone inhibits renal injury in theabsence of neutrophils

HSI CHIAO, YUKIMASA KOHDA, PAUL MCLEROY, LEONARD CRAIG, STUART LINAS, and ROBERT A. STAR

Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, and University of Colorado, Denver,Colorado, USA

a-Melanocyte-stimulating hormone inhibits neutrophil-indepen-dent renal injury.

Background. We previously showed that a-melanocyte stimu-lating hormone (a-MSH) decreases ischemia/reperfusion injuryeven when started six hours after ischemia. a-MSH inhibits bothneutrophil accumulation and nitric oxide production. To deter-mine the relative importance of a-MSH on the neutrophil path-way, we examined the effects of a-MSH in injury models whereneutrophil effects are minimal or absent.

Methods. We studied the effects of a-MSH in (1) intercellularadhesion molecule-1 (ICAM-1) knock-out and background micethat were subjected to 40 minutes of ischemia and 24 hoursreperfusion, and (2) isolated kidneys that were subjected to in vivoischemia for 20 minutes and then perfused ex vivo for one hourwithout neutrophils. To begin to search for direct tubule effects ofa-MSH, we studied the effect of a-MSH on nitric oxide (NO) inendotoxin/interferon-g-treated mouse cortical tubule cells.

Results. ICAM-1 knock-out mice had 75% less neutrophilinfiltration than background mice after ischemia. Despite therelative lack of neutrophils, a-MSH inhibited renal injury inICAM-1 knock-out mice. a-MSH also significantly preservedGFR and tubular sodium reabsorption in the isolated perfusedischemic kidney model. a-MSH and a nitric oxide inhibitor did notexhibit synergy. Finally, a-MSH inhibited nitrite production by20% in the mouse cortical tubule cells (MCT), similar to parallelobservations in a cultured mouse macrophage line (RAW cells).

Conclusions. We conclude that a-MSH decreases renal injurywhen neutrophil effects are minimal or absent, indicating thata-MSH inhibits neutrophil-independent pathways of renal injury.The preservation of sodium absorption ex vivo and inhibition ofnitrite production in cultured MCT cells suggests that a-MSHinhibits tubular injury by direct tubular effects.

Organ ischemia is the primary cause of stroke, myocar-dial infarction, and acute renal failure, and increases therisk of rejection following organ transplantation. While

prolonged ischemia causes anoxic cell death, additionalwaves of injury initiated following sublethal injury arecaused in part by maladaptive inflammatory and cytotoxicinjury cascades activated during the reperfusion period.Recent studies of renal ischemia-reperfusion injury havefocused on the roles of neutrophils and cytotoxic nitricoxide. Following renal ischemia, neutrophils accumulate inthe outer stripe of outer medulla beginning four hours afterischemia, which is coincident with histologic evidence ofischemia [1–3]. While infiltrating neutrophils can removenecrotic and apoptotic cells and hence is a passive responseto injury, many studies indicate that the neutrophils play apathogenic role. For example, neutrophils may plug theascending vasa recta in the outer stripe of the outermedulla, and further impair oxygen supply to the proximalstraight tubule, the major site of injury in the clamp modelof renal injury [2]. Support for this pathogenic role comesfrom studies where ischemic injury is partially inhibited byneutrophil depletion in some [4, 5], but not all [6, 7],studies. Ischemic renal injury is potentiated by infusion ofactivated neutrophils [8, 9] and is decreased by blockingantibodies or antisense oligonucleotides to intercellularadhesion molecule-1 (ICAM-1) or P-selectin or mice lack-ing ICAM-1 [10–14]. The amount of protection by neutro-phil depletion/inhibition is more striking with shorterischemia times. However, cardiac and renal ischemia-reperfusion injury can occur in the absence of neutrophils[15, 16], suggesting other mechanisms of injury. Recentstudies have focused on maladaptive cytotoxic cascadesactivated during reperfusion that liberate high concentra-tions of cytotoxic nitric oxide (NO). Hypoxic/reperfusioninjury to isolated non-perfused proximal tubules can beinhibited by blocking nitric oxide production [17]. Theinducible isoform of NO synthase (iNOS) is induced byischemia in proximal tubules. Blocking iNOS with antisenseoligonucleoides protects cultured proximal tubule cellsfrom hypoxic damage in vitro and against ischemic damagein vivo [18]. In addition to being directly cytotoxic, the highconcentrations may also inhibit endothelial NO, with re-sultant increased organ ischemia [19]. Taken together, the

Key words: ischemia, reperfusion, a-MSH, neutrophils, nitric oxide,ICAM-1.

Received for publication January 30, 1998and in revised form April 8, 1998Accepted for publication April 14, 1998

© 1998 by the International Society of Nephrology

Kidney International, Vol. 54 (1998), pp. 765–774

765

Page 2: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

experimental evidence suggests that cytotoxic NO alsoplays a pathogenic role in renal injury.

a-MSH is an anti-inflammatory cytokine that inhibitsboth neutrophil and nitric oxide pathways. a-MSH inhibitsneutrophil infiltration by both direct and indirect mecha-nisms: it inhibits the synthesis of neutrophil chemokineinterleukin (IL)-8 and neutrophil adhesion moleculeICAM-1 [3, 20], and directly inhibits IL-8 stimulated neu-trophil migration in vitro [20–22] via melanocortin a-MSHreceptors [23]. a-MSH also inhibits the induction of NO invivo and in vitro [20, 24, 25]. The ability of a-MSH to inhibitboth neutrophil and NO pathways suggested that a-MSHmight inhibit ischemia/reperfusion injury. Indeed, a-MSHinhibited renal ischemia-reperfusion injury even whenstarted six hours after reperfusion [3]. a-MSH inhibitedboth neutrophil infiltration, induction of mRNA for IL-8and ICAM-1, and induction of iNOS enzyme and activity.a-MSH was more effective in inhibiting renal damage afterischemia than other agents that inhibit only one pathway.The potential ability of a-MSH to inhibit a non-neutrophilpathway is particularly interesting. Studies employing iso-lated perfused hearts and kidneys have shown that cardiacand renal ischemia-reperfusion injury can occur in theabsence of neutrophils [15, 16]. Furthermore, the role ofneutrophils in human acute renal failure is controversial.Neutrophils are seen in human biopsy material, but not tothe extent as in the animal models [26]. However, renalbiopsies are generally not obtained early in the course ofacute renal failure, and special stains are required to detectneutrophils [27]. Monoclonal antibodies to ICAM-1 hadonly a small effect in post-transplant acute renal failure inhumans [28]. Thus, agents that inhibit both neutrophil-dependent and neutrophil-independent pathways may beuseful as therapeutic agents.

In this study, we examined the effects of a-MSH on renalischemia/reperfusion injury in two injury models whereneutrophil infiltration is either less prominent (ICAM-1knock-out mice) or absent (isolated kidney perfused in theabsence of neutrophils). Renal damage was assessed bycreatinine and by changes in renal histology in the in vivomodel, and by GFR and sodium absorption in the ex vivoisolated perfused kidney model. We also examined theeffects of a-MSH on NO production in cultured mousecortical tubule (MCT) cells.

METHODS

Animals

Female C57BL/6J or ICAM-1 knock-out [B/6J-ICAM(tm1BAY)] mice aged 7 to 8 weeks (20 to 22 g) werepurchased from Jackson Laboratories (Bar Harbor, ME,USA). All animals had free access to water and food (4%mouse-rat diet; Harlan Sprague-Dawley or low potassiumdiet containing 50% of usual dietary potassium [29]). Rats(275 to 300 g) were purchased from Sasco (Omaha, NE,

USA). Animal care followed the criteria of the Universityof Texas Southwestern Medical Center and the Universityof Colorado for the care and use of laboratory animals inresearch.

Chemicals

a-MSH was purchased from Phoenix Pharmaceuticals,Inc. (Mountain View, CA, USA). Sterile normal saline wasfrom Baxter Healthcare Corp. (Deerfield, IL, USA); Naph-thol AS-D Chloroacetate Esterase Stain Kit (Kit # 91-C;Sigma); Periodic Acid-Schiff (PAS) Kit (Kit # 395-8,Sigma), and 10% formalin (Fisher); Albumin (fractionV-Cohn bovine; Integren, Purchase, NY, USA); hydroxyl-[methyl-14C]-inulin (Amersham); and Lw-nitro-L-arginine(L-NNA, Sigma). All other chemicals were purchased fromSigma Chemical Co.

Surgery and experimental protocol

Surgery was performed as described previously [3]. The40 minute ischemia time was chosen to provide significantrenal injury that could be reversed by experimental agents.The mice were anesthetized with an i.m. injection of 100mg/kg ketamine, 10 mg/kg xylazine, and 1 mg/kg aceproma-zine. The animals were placed on 39°C heating table tomaintain constant body temperature. Blood (100 ml) wascollected from the tail vein. Both renal pedicles werecross-clamped for 40 minutes. a-MSH or vehicle was givenat the time of clamp release. After the clamps wereremoved, 1 ml of prewarmed (37°C) normal saline wasinstilled into the abdominal cavity. The abdomen wasclosed, and the animals were placed in a 29°C incubatorovernight. At the time of sacrifice, blood was collected formeasurement of plasma creatinine. Both kidneys wereharvested for histological study or RNA. The animals weredivided into three groups: sham, ischemia/vehicle, andischemia/a-MSH. Sham-treated animals went through thesame surgical procedure as the other animals, includingdissection of the renal pedicle, however, renal clamps werenot applied. The vehicle and a-MSH treated animalsreceived vehicle (normal saline) or 25 mg a-MSH intrave-nously during the ischemic period, and at 6 and 18 hourspost-perfusion. Creatinine was measured by an Beckmancreatinine autoanalyzer.

Histological examination

Formalin (10%)-fixed and paraffin-embedded kidneyspecimens were stained with hematoxylin and eosin (H &E; 2 mm), naphthol AS-D chloroacetate esterase (3 mm), orperiodic acid Schiff (PAS; 2 mm) stains. Histologicalchanges were evaluated by semiquantitative measurementsof neutrophil infiltration, tissue necrosis and erythrocytecongestion as described previously [3].

Chiao et al: a-MSH inhibits neutrophil-independent renal injury766

Page 3: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

RNA extraction and Northern blot hybridization

Renal total RNA was purified using guanidinium thiocy-anate-phenol-chloroform extraction [30]. Samples of totalRNA (20 mg) were fractionated via electrophoresis on0.9% agarose-formaldehyde gel and transferred onto anylon membrane. The equality of RNA samples aftertransfer to the membrane was substantiated by UV illumi-nation of ethidium bromide. Membranes were fixed bybaking at 80°C, then prehybridized at 42°C in 50% form-amide, 0.5% SDS and 53 Denhardt’s, 5 3 SSC, and 0.5mg/ml salmon sperm DNA. The membranes were hybrid-ized with [32P]dCTP-labeled cDNA clones. 0.5 to 2 kBportions of mouse KC/IL-8 and ICAM-1 were generated byPCR, and the products confirmed by automated sequenc-ing. The hybridized membranes were washed twice in 0.1 3SSC and 0.1% SDS at 50°C. Loading of RNA was normal-ized by rehybridizing with GAPDH.

Isolated perfused ischemic rat kidney experiments

Kidneys were made ischemic in vivo, then perfused exvivo as described previously [16]. The ischemia time waschosen to provide significant renal injury that could bereversed by experimental agents. Briefly, after anesthesia,the right renal artery was occluded for 20 minutes. Theright renal artery was cannulated and flushed with asolution containing vehicle or 100 nM a-MSH, and theentire kidney was transferred to a perfusion chamber forperfusion with a pulsatile pump at a constant pressure of100 mm Hg. After a 15-minute equilibrium period, threeurine collections and perfusion samples were obtained at 15minute intervals. Samples were frozen within 60 minutes ofcollection, and subsequently analyzed for [14C]-inulin andsodium. The perfusate solution consisted of 150 ml of aKrebs-Ringer bicarbonate buffer containing urea, [14C]-inulin, albumin and vehicle, 100 nM a-MSH, or 100 nM

a-MSH and 10 mM L-NNA. The final composition of theperfusion solution was (in mM) 140 Na, 5 K, 105 Cl, 25bicarbonate, 1.2 ionized Ca, 1 Mg, 1 sulfate, 1 phosphate, 7urea, 70 mg/ml albumin, and 5 glucose. Renal injury wasassessed by changes in glomerular filtration rate (GFR) andtubular sodium reabsorption (TNa). Sodium was measuredwith a model IL 343 flame photometer (InstrumentationLaboratory, Lexington, MA, USA). Radioactivity of [14C]-inulin was measured by a liquid scintillation counter.Urinary clearances of [14C]-inulin and sodium were calcu-lated from the urine and plasma concentration ratios.

Cell culture

Mouse cortical tubule (MCT) cells were grown in RPMI1640 medium supplemented with penicillin, streptomycin,and 10% heat-inactivated fetal bovine serum (AtlantaPremium Select; Atlanta Biologicals, Norcross, GA, USA).RAW 264.7 cells were maintained in Dulbecco’s modifiedEagle medium supplemented with 4.5 g/liter glucose, non-

essential amino acids, 4 mM L-glutamine, penicillin, strep-tomycin, and 10% heat-inactivated fetal bovine serum.MCT and RAW cells were seeded in 96 well microplates atconcentrations of 1.2 3 104 and 7.5 3 104 cells per 100 mlper well. The cells were incubated for 30 hours at 37°C in5% CO2/air. MCT cells were treated after 30 hours with0.35 U/ml recombinant mIFN-g with or without 0.35 nM

a-MSH in an additional 100 ml complete-RPMI/well. TheRAW cells were treated after 30 hours with 10 ng/ml LPS(E. coli 026:B6; Sigma) with or without 0.35 nM a-MSH inan additional 100 ml complete D-MEM/well. The treatedcells were incubated for an additional 16 to 20 hours at37°C. Nitrite concentrations were determined with Greissreagent using NaNO2 as a standard [30]. All assays wereperformed using three to four replicates, and repeated in atleast four independent experiments.

Statistical analysis

All data were described as mean 6 SEM. Differenttreatments were compared using ANOVA techniques(completely randomized design) followed by Tukey’s testfor individual comparisons between group means. The nullhypothesis was rejected when P , 0.05.

RESULTS

Functional protection by a-MSH in ICAM-1 knock-outmice

Forty minutes of renal ischemia caused severe injury; allof the background C57BL/6J animals in the placebo groupdied. All of the a-MSH treated background mice survivedwith a mean plasma creatinine at 24 hours of 3.3 6 0.3mg/dl. In contrast, all the placebo-treated ICAM-1 knock-out animals survived, with a mean creatinine of 3.3 6 0.3mg/dl. a-MSH significantly reduced the plasma creatininemeasured 24 hours after injury to 0.9 6 0.3 mg/dl. Thus, theICAM-1 knock-out mice survived a lethal amount of renalinjury, similar to the protection against a lessor amount ofischemic injury reported by Kelley et al [10]. a-MSHinhibited the decline in renal function in the ICAM-1knock-out mice, and allowed the background mice tosurvive.

The initial group of mice were fed normal rat/mousechow, which may have caused lethal hyperkalemia (notmeasured). Therefore, the experiment was repeated inanimals fed a low potassium diet that contained about 50%of the potassium content of the normal chow. Since thismodification allowed the placebo-treated background ani-mals to survive (see below), all subsequent experimentswere conducted using this diet. Figure 1 shows the effect ofa-MSH in background and ICAM-1 knock-out mice sub-jected to 40 minutes of ischemia with reperfusion for 24hours. Under these conditions, the difference between thevehicle-treated background and ICAM-1 knock-out ani-mals was not statistically significant. a-MSH significantly

Chiao et al: a-MSH inhibits neutrophil-independent renal injury 767

Page 4: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

inhibited renal injury in both the background (placebo,3.2 6 0.2 mg/dl; a-MSH, 1.5 6 0.2 mg/dl) and ICAM-1knock-out animals (placebo, 2.9 6 0.2 mg/dl; a-MSH, 0.7 60.2 mg/dl). a-MSH was significantly more effective in theICAM-1 knock-out animals (by ANOVA and Tukey mul-tiple comparison test), with 103% reduction in creatinine inICAM-1 knock-out mice versus 59% reduction in back-ground mice.

Histology

To determine if the functional changes were associatedwith anatomical evidence of necrosis, we investigated his-tologic changes at 24 hours post-ischemia. Figure 2 showstypical histologic patterns seen in the four groups ofanimals. Placebo-treated animals of either group showedwidespread necrosis of S3 proximal tubules in the outerstripe of the outer medulla (Fig. 2 A, B). The placebo-treated background animals showed extensive neutrophilaccumulation in the outer stripe, and erythrocyte conges-tion in the inner stripe (Fig. 2A). In contrast, the placebo-treated ICAM-1 knock-out animals had much less neutro-phil accumulation in the outer stripe (Fig. 2B). a-MSH hadonly slight effect in background mice (Fig. 2C). In contrast,a-MSH had a dramatic protective effect on renal histologyin the ICAM-1 knock-out mice (Fig. 2D). There were onlya few casts, patchy congestion, and minimal necrosis.

The histologic changes were measured using semiquan-titative scales (0 to 31) for outer stripe tissue necrosis andinner stripe erythrocyte congestion (Fig. 3), and quantita-tive counts of the number of neutrophils in the outer stripeof the outer medulla (Fig. 4). a-MSH decreased outerstripe tissue necrosis in both background and ICAM-1knock-out mice, but had a more impressive effect in theICAM-1 knock-out mice. a-MSH inhibited inner stripeerythrocyte congestion in both sets of animals. The

ICAM-1 knock-out mice had significantly less neutrophilaccumulation than the background mice (background,346 6 39 vs. ICAM-1 knock-out, 82 6 13 neutrophils permm2). a-MSH significantly reduced neutrophil infiltrationin both the background and ICAM-1 knock-out animals(background, 210 6 36; ICAM-1 knock-out, 12 6 10neutrophils per mm2), but had a more pronounced effect inthe ICAM-1 knock-out animals (85% reduction in ICAM-1knock-out mice vs. 39% reduction in background animals).

The ICAM-1 mice had detectable neutrophil accumula-tion in the kidney, perhaps in response to tissue injuryrather than causing it. Since fewer neutrophils accumulateat shorter ischemia times, we tried less ischemia time in theICAM-1 knock-out mice. While we did get less ischemicdamage, we still found neutrophil accumulation in theouter medulla (data not shown).

RNA blot analysis of ICAM-1 mRNA

To demonstrate that ICAM-1 was absent in the knock-out mice, we performed RNA blotting for ICAM-1 mRNA.Figure 5 shows that ICAM-1 mRNA was absent, as ex-pected, in the ICAM-1 knock-out animals. Ischemia in-creased ICAM-1 mRNA abundance in the backgroundanimals, and a-MSH inhibited induction of ICAM-1. Thesechanges are similar to what we found in Balb/c mice at 24hours [3].

Effects of a-MSH in the isolated perfused kidney

We performed additional experiments using an isolatedperfused kidney model where neutrophils could be com-pletely eliminated during the reperfusion period. Rat kid-neys were made ischemic in situ for 20 minutes, flushedwith buffer, and perfused ex vivo in an isolated kidneyperfusion system. The flush and perfusion solutions con-tained vehicle or a-MSH (100 nM). a-MSH caused a slightbut statistically nonsignificant decrease in perfusion flow tothe ischemic kidneys (control, 34 6 3 ml/min vs. a-MSH,29 6 2 ml/min). Figure 6 shows the effects of a-MSH on thefunction of the ischemic kidneys studied ex vivo. Theshaded areas correspond to glomerular filtration rate(GFR) and tubular sodium absorption (TNa,, a marker ofproximal tubule function) previously found in non-ischemickidneys studied ex vivo [9, 16]. As expected, the vehicle-treated ischemic kidneys (open circles) had significantlylower GFR and tubular sodium absorption than non-ischemic kidneys, with recovery of both GFR and sodiumabsorption over time. Compared with vehicle, a-MSHaddition significantly improved both glomerular (GFR) andtubular (TNa) function at 30, 45, and 60 minutes of reper-fusion (closed circles). In non-ischemic kidneys, a-MSHhad no effect on GFR or tubular sodium absorption (datanot shown).

To evaluate the contribution of NO in mediating theneutrophil independent protective effects of a-MSH, westudied the effect of 100 nM a-MSH and 10 mM L-NNA.

Fig. 1. Effect of a-MSH in background and ICAM-1 knock-out mice.Mice were subjected to 40 minutes ischemia, received vehicle or 25 mga-MSH just before clamp release, placed on a low potassium diet, andsacrificed after 24 hours. Data shown as mean 6 standard error. *P ,0.05.

Chiao et al: a-MSH inhibits neutrophil-independent renal injury768

Page 5: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

This concentration of L-NNA has been shown previously topartially inhibit ischemia/reperfusion damage in the ab-sence of neutrophils (Fig. 6; open triangles) [16]. We foundthat addition of L-NNA and a-MSH together (open dia-monds) had similar effects as a-MSH alone (closed circles).There was no evidence of additivity.

Effect of a-MSH on nitrite production

Since the lack of additivity between L-NNA and a-MSHin the isolated perfused kidney preparation suggested thata-MSH might inhibit renal NO, we tested this possibility inMCT cells. Incubation with 0.35 nM a-MSH for 16 hoursdecreased IFN-g-stimulated NO production in MCT cellsby 19.7 6 2.4% (Fig. 7). The degree of inhibition was notstatistically different than that observed in parallel experi-ments in cultured mouse macrophages (RAW cells).

DISCUSSION

a-MSH inhibits both inflammatory (neutrophil) andcytotoxic (nitric oxide) pathways in renal ischemia. Thepresent studies were designed to assess the role of neutro-

phil-independent pathways both in an in vivo model(ICAM-1 knock-out) and an ex vivo model (isolated isch-emic kidney perfused without neutrophils). The major newfindings in this paper are: (1) a-MSH is more effective inICAM-1 knock-out mice than in the background strain, (2)a-MSH inhibits renal injury in the isolated kidney perfusedwithout neutrophils, (3) a-MSH has direct tubule effects,and (4) a-MSH is less effective in C57BL/6J than Balb/cmice. Taken together, the evidence suggests that a-MSHcan inhibit renal injury without neutrophils present, per-haps via a direct tubular effect. These findings are discussedbelow.

a-MSH is more effective in ICAM-1 knock-out mice thanin the background strain

Previous studies by Kelley et al and Rabb et al haveshown that interruption of ICAM-1 either by geneticknock-out or blocking antibody made mice less sensitive torenal ischemia [10, 11, 13]. Since the degree of protectionwas more pronounced at the shorter ischemia times, it is

Fig. 2. Typical renal histology of outer stripe of outer medulla. Sections stained with Ledar stain for neutrophils (pink, arrowheads). (A) Backgroundmouse, vehicle. (B) ICAM-1 knock-out mouse, vehicle. (C) Background mouse, a-MSH. (D) ICAM-1 knock-out mouse, a-MSH.

Chiao et al: a-MSH inhibits neutrophil-independent renal injury 769

Page 6: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

suspected that ICAM-1 dependent and hence neutrophil-dependent injury pathways are more important at shorterischemia times [27]. We used a more severe insult (40 minclamp) to obtain both neutrophil-dependent and neutro-phil-independent mechanisms of injury. Even with thissevere injury, ICAM-1 knock-out animals were less sensi-tive to ischemia, as assessed by survival on a normal mousediet (100% survival in ICAM-1 knock-out mice vs. 100%death in background mice). A similar but smaller survivaladvantage in ICAM-1 knock-out mice has been noted using32 minutes of ischemia (50% of the background animalsdied within 72 hours, whereas all of the ICAM-1 knock-outmice survived [10]). On a low potassium diet, we found asmall numerical difference in serum creatinine at 24 hours

(Fig. 1), although it was not statistically significant. WhileKelly et al previously demonstrated a dramatic protectiveeffect in the ICAM-1 knock-out mice using 32 minutes ofischemia, their background animals had a creatinine at 1.4mg% at 24 hours versus the 2.9 mg% in the present studythat employed 40 minutes ischemia. Nevertheless, theICAM-1 knock-out mice had 76% less neutrophil infiltra-tion in response to ischemia than the background mice (82neutrophils per mm2 in ICAM-1 knock-out mice vs. 346 inbackground mice). Thus, the ICAM-1 knock-out mice weremodestly protected from ischemic damage, indicating renalischemic/reperfusion injury is mediated by both neutrophiland non-neutrophil injury cascades.

We previously found that a-MSH inhibited renal isch-emic injury in Balb/c mice [3]. In this study, a-MSHinhibited renal injury following 40 minutes of bilateralischemia in both background and ICAM-1 knock-out micein normal and low potassium diets (Fig. 1). However, thedegree of protection was larger in the ICAM-1 knock-outmice, where a-MSH inhibited the rise in plasma creatinineat 24 hours post-ischemia. The degree of protection wascollaborated by the renal histological changes (Fig. 2).Whereas a-MSH was not very effective in the backgroundanimals (see below), a-MSH reduced renal histologicaldamage to near baseline values in the ICAM-1 knock-outmice (Fig. 3). a-MSH also inhibited neutrophil infiltrationin the ICAM-1 knock-out animals, to levels similar to thesham animals. Taken together, the results demonstrate thata-MSH was more effective in preserving renal function andcellular viability in the ICAM-1 knock-out animals than thebackground animals. The degree of protection to moder-ately severe renal ischemia/reperfusion injury has beenseen previously by a-MSH in Balb/c mice and P-selectininhibitors. However, the P-selectin inhibitor had to be given

Fig. 3. Effect of a-MSH on semiquantitativehistologic changes (tissue necrosis anderythrocyte congestion). The Methods sectioncontains a description of the methodology.

Fig. 4. Effect of a-MSH on the number of neutrophils in the outer stripeof the outer medulla. Neutrophil counts expressed as mean 6 1 standarderror. Symbols are: (f) vehicle; (u) a-MSH. *P , 0.05 versus vehicle.**P , 0.05 versus background.

Chiao et al: a-MSH inhibits neutrophil-independent renal injury770

Page 7: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

before renal ischemia, whereas a-MSH could be given upto three to six hours after ischemia [3, 14]. The ICAM-1knock-out animals had 76% less neutrophil infiltration anda-MSH still greatly inhibited renal damage, suggesting thata-MSH must also work via neutrophil-independent path-ways. However, it is also possible that the 26% remainingneutrophils were sufficient to cause renal injury, and thata-MSH protected the kidney by inhibiting accumulation ofthe these neutrophils. To test if some effects of a-MSH areindependent of neutrophils required a different experimen-tal approach.

a-MSH inhibits renal injury in the isolated kidneyperfused without neutrophils

Preventing neutrophil infiltration is very difficult in ex-perimental animals. Neutrophil depletion with anti-neutro-

phil serum is protective, but only if neutrophils weredepleted to extremely low levels. Since neutrophil depen-dent injury is more prevalent at low ischemia times [27], wetried shorter ischemia times. As expected, we observed lessischemic damage, however, we were unable to eliminateneutrophil infiltration in the ICAM-1 knock-out mice.Therefore, additional studies were performed using a dif-ferent experimental system (isolated perfused kidney prep-aration) where neutrophils could be eliminated during thereperfusion period. Kidneys were made ischemic in vivothen transferred to an ex vivo perfusion system where theycould be perfused without neutrophils. Even under theseconditions, treatment with a-MSH resulted in markedprotection of both glomerular and tubular functions;a-MSH returned GFR and tubular function towards nor-mal more rapidly than in untreated kidneys. The free

Fig. 5. Outer medullary ICAM-1 mRNA by RNA blotting. RNA (20 mg/lane) was electrophoresed and blotted for ICAM-1. Each lane represents RNAfrom a single animal. Blot shown is representative of two similar experiments.

Fig. 6. Effect of a-MSH on kidney function.(A) Glomerular filtration rate (GFR). (B)Tubular sodium reabsorption (TNa) following 20minutes of ischemia in vivo and perfusionwithout neutrophils by the isolated kidneytechnique. The shaded area represents normalvalues for non-ischemic kidneys. Data from L-NNA were obtained from [16]. Symbols are:(E) vehicle; (F) a-MSH; (‚) L-NNA; (l) L-NNA 1 a-MSH. N 5 5 experiments for vehicleand a-MSH studies, and N 5 3 for a-MSH andL-NNA studies.

Chiao et al: a-MSH inhibits neutrophil-independent renal injury 771

Page 8: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

radical scavengers DMTU, DMSO, and xanthine oxidaseinhibitors have also proven to be more effective in protect-ing isolated perfused kidneys from ischemia/reperfusioninjury in the absence of neutrophils [31, 32]. a-MSH was aseffective as each of these agents in protecting kidneys fromischemia. Kainz et al have reported in abstract form asimilar study employing ex vivo ischemia, where kidneyswere perfused for one hour ex vivo, made ischemic for 20minutes, then reperfused without neutrophils. a-MSH alsoinhibited renal injury under these conditions [33]. Thus,a-MSH inhibits renal ischemia/reperfusion damage in theabsence of neutrophils, indicating that a-MSH must act onother injury cascades. Since a-MSH was only given to theischemic kidney and not infused systemically, the protectiveeffects of a-MSH do not require a systemic action (hemo-dynamic, anti-inflammatory, or otherwise), but are largelymediated by the direct action of a-MSH on the kidney.Whether a-MSH has additional beneficial systemic actionsduring renal ischemia is unknown.

We previously found that a-MSH inhibited induction ofinducible NO synthase (iNOS) in murine macrophages invitro, endotoxin-induced liver injury, and ischemia-inducedrenal injury [3, 20, 25]. a-MSH inhibited production of highconcentrations of NO in the outer medulla during ischemia[3]. High concentrations of NO produced by proximaltubules are thought to be a pathogenically important causeof renal injury, since inhibition of NO decreases injury invitro and in vivo [17, 18]. We evaluated the potential role ofNO in mediating the neutrophil-independent effects ofa-MSH using the NO inhibitor L-NNA. a-MSH was moreeffective than L-NNA alone, suggesting that a-MSH has

effects other than inhibition of NO. We also found thata-MSH and L-NNA together were about as effective asa-MSH alone. The lack of additivity between a-MSH and aNO inhibitor suggests that the two agents share commonmechanisms of action. The identity of the NO- and neutro-phil-independent pathway(s) are unknown, but worthy offurther study.

a-MSH has direct tubule effects

The isolated perfused kidney studies cannot conclusivelyprovide a mechanism. a-MSH did not increase the perfu-sion flow rate, suggesting that it does not act as a vasodi-lator. Therefore, the effect of a-MSH to preserve sodiumexcretion suggests that a-MSH might have a direct effecton the proximal tubule. a-MSH also inhibits injury to theS3 proximal straight tubule in the relative absence ofneutrophils (Fig. 2), consistent with a local effect of a-MSHin the vicinity of the outer medulla. We previously showedthat a-MSH inhibited production of high concentrations ofNO in the outer medulla during ischemia [3]. Since theisolated kidney preparation experiments suggested thata-MSH may act in part via inhibition of NO, we tested theeffects of a-MSH on cytokine-induced iNOS in MCT cells,a cell line derived from proximal tubules. a-MSH inhibitedcytokine-stimulated NO production (Fig. 7). This concen-tration of a-MSH is slightly below the reported EC50 for allmelanocortin receptors (1 nM) (discussed in [25]), indicat-ing that the MCT cells express a functional melanocortinreceptor. We have recently produced a polyclonal antibodyto the MC-1 melanocortin receptor that stains human andmouse neutrophils in a plasma membrane pattern [23].This antibody also detects MC1 in MCT cells by proteinblotting, suggesting that a-MSH may work in part viastimulation of MC1 a-MSH receptors. It is not known ifthis receptor is expressed on the apical or basolateralmembrane.

While the signals responsible for increasing iNOS in vivoare unknown, ischemia increases the renal production ofpro-inflammatory cytokines and chemokines (includingIL-1, TNF-a, IL-8, MCP, etc.) [3, 10, 34], which maysubsequently induce expression of iNOS. Taken together,the data indicate that a-MSH has at least one direct tubularaction that is independent of its actions on neutrophils.Whether a-MSH acts at other tubular or vascular sites inthe kidney is unknown. We have detected expression ofMC-1 on the apical membrane of collecting ducts fromcortex to inner medulla [23]; however, the physiologicalfunction of this receptor is unknown.

a-MSH is not effective in the C57BL/6J background mice

We previously found that a-MSH dramatically reducedrenal reperfusion injury and neutrophil infiltration inBalb/c mice. a-MSH restored both to levels detected insham-operated animals. In those mice, a-MSH inhibitedthe rise in renal RNA abundance for the neutrophil

Fig. 7. Effect of a-MSH on LPS and IFN-g stimulated nitrite productionin mouse cortical tubule (MCT) cells and RAW 264.7 cells. Symbols are:(M) control; (f) a-MSH. *P , 0.05 versus control (no a-MSH).

Chiao et al: a-MSH inhibits neutrophil-independent renal injury772

Page 9: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

chemokine IL-8 and ICAM-1 [3]. In contrast, the C57BL/6Jmice are more sensitive to renal ischemia, and a-MSH onlyinhibited renal injury by 53% (as judged by changes increatinine at 24 hr) and neutrophil infiltration by 39%.a-MSH inhibited the ischemia-induced increase in ICAM-1as in Balb/c mice. Mice strains vary in their degree ofsusceptibility to cerebral ischemia. Yang et al found thatthe C57BL/6 strain was the most susceptible to cerebralischemia following bilateral common carotid occlusionamong seven host strains [35]. Survival correlated with thedegree of anatomical anastomosis between carotid andbasilar arteries. It is not known if there are anatomicaldifferences in renal vasculature between C57BL/6 andBalb/c mice. However, the C57BL/6 and Balb/c mice strainshave immunological differences that cause susceptibility tocertain diseases. For example, most clones from Leishma-nia major resistant C57BL/6 mice produce Th1 cytokines(IFN-g), whereas T-cells from L. major susceptible Balb/cmice produce Th2-dominant responses (that is, IL-4) [36,37]. In contrast, C57BL/6 mice are more susceptible to C.parvum/LPS-induced liver injury than Balb/c mice [38].Blockade of IL-12 or IFN-g abrogates injury in C57BL/6mice, while IL-12 induces C. parvum/LPS-induced injury inresistant Balb/c mice [38]. Thus, subtle differences inanatomical arrangement or cytokine responsiveness be-tween the strains might account for differences in suscep-tibility to ischemia and effectiveness of a-MSH. Elucida-tion of these differences may allow further insight into themechanisms of ischemia and actions of a-MSH.

Physiologic significance

Animal models of ischemic injury suggest the involve-ment of multiple injury cascades involving neutrophils,inflammation, nitric oxide, superoxide, calcium, etc. Incontrast, the mechanisms of human acute renal failure arepoorly understood largely because human acute renalfailure is multi-factorial, there are only a few reportedseries of human biopsy material, and biopsies are generallynot obtained at the time of initiation of acute renal failure.The role of neutrophils in human acute renal failure isparticularly controversial. The reported series do showsome neutrophils, but not to the extent seen in the rat andmouse models of renal ischemia/reperfusion injury. Never-theless, our studies show that a-MSH decreases renal injuryin the relative and complete absence of neutrophils, indi-cating that a-MSH works on neutrophil-independent path-ways. Given the uncertainty of the mechanisms of humanacute renal failure, it is likely that an agent that inhibits onseveral injury cascades is more likely to be therapeuticallyimportant than one that only inhibits a single cascade. Sincea-MSH works on both neutrophil-dependent and neutro-phil-independent pathways, it may be more effective thanagents that only inhibit neutrophil events. The preservationof sodium absorption ex vivo and inhibition of NO produc-

tion in cultured MCT cells suggests that a-MSH may work,in part, via direct tubular effects.

ACKNOWLEDGMENTS

This study was supported by the National Institutes of Health Grant DK45923 and NHLBI 2P50-HL-40784. This work as done during the tenureof an Established Investigatorship from the American Heart Association(RAS). H. Chiao was supported by funds from a NIH training grant, andY. Kohda was supported by a fellowship award from the National KidneyFoundation. Portions of this paper were presented at the annual meetingof the American Society of Nephrology, November 1997, and published inabstract form (J Am Soc Nephrol 8:590, 1997).

Reprint requests to Robert A. Star, M.D., Department of Internal Medicine,University of Texas-Southwestern Medical Center, 5323 Harry Hines, Dallas,Texas 75235-8856, USA.E-mail: [email protected]

APPENDIX

Abbreviations used in this article are: a-MSH, a-melanocyte-stimulat-ing hormone; GFR, glomerular filtration rate; H & E, hematoxylin andeosin; ICAM-1, intercellular adhesion molecule-1; IFN-g, interferon-g;IL, interleukin; iNOS, inducible nitric oxide synthase; L-NNA, Lv-nitro-L-arginine; LPS, lipopolysaccharide; MCT, mouse corticle tubule; NO,nitric oxide; PBS, phosphate buffered saline; TNa, tubular sodium reab-sorption; TNF-a, tumor necrosis factor-a.

REFERENCES

1. WILLIAMS P, LOPEZ H, BRITT D, CHAN C, EZRIN A, HOTTENDORF R:Characterization of renal ischemia-reperfusion injury in rats. J Phar-macol Toxicol Methods 37:1–7, 1997

2. WILLINGER CC, SCHRAMEK H, PFALLER K, PFALLER W: Tissuedistribution of neutrophils in postischemic acute renal failure. Vir-chows Archiv B-Cell Pathol 62:237–243, 1992

3. CHIAO H, KOHDA Y, MCLEROY P, CRAIG L, HOUSINI I, STAR RA:a-Melanocyte-stimulating hormone protects against renal injury afterischemia in mice and rats. J Clin Invest 99:1165–1172, 1997

4. KLAUSNER JM, PATERSON IS, GOLDMAN G, KOBZIK L, RODZEN C,LAWRENCE R, VALERI CR, SHEPRO D, HECHTMAN HB: Postischemicrenal injury is mediated by neutrophils and leukotrienes. Am J Physiol256:F794–F802, 1989

5. HELLBERG POA, KALLSKOG TOK: Neutrophil-mediated post-isch-emic tubular leakage in the rat kidney. Kidney Int 36:555–561, 1989

6. PALLER MS: Effect of neutrophil depletion on ischemic renal injury inthe rat. J Lab Clin Med 113:379–386, 1989

7. THORNTON MA, WINN R, ALPERS CE, ZAGER RA: An evaluation ofthe neutrophil as a mediator of. in vivo renal ischemic-reperfusioninjury. Am J Pathol 135:509–515, 1989

8. LINAS SL, WHITTENBURG D, PARSONS PE, REPINE JE: Mild renalischemia activates primed neutrophils to cause acute renal failure.Kidney Int 42:610–616, 1992

9. LINAS SL, WHITTENBURG D, PARSONS PE, REPINE JE: Ischemiaincreases neutrophil retention and worsens acute renal failure: Roleof oxygen metabolites and ICAM-1. Kidney Int 48:1584–1591, 1995

10. KELLY KJ, WILLIAMS WW JR, COLVIN RB, MEEHAN SM, SPRINGERTA, GUTIERREZ-RAMOS J-C, BONVENTRE JV: Intercellular adhesionmolecule-1-deficient mice are protected against ischemic renal injury.J Clin Invest 97:1056–1063, 1996

11. RABB H, MENDIOLA CC, SABA SR, DIETZ JR, SMITH CW, BONVENTREJV, RAMIREZ G: Antibodies to ICAM-1 protect kidneys in severeischemic reperfusion injury. Biochem Biophys Res Commun 211:67–73,1995

12. HALLER H, DRAGUN D, MIETHKE A, PARK JK, WEIS A, LIPPOLDT A,GROSS V, LUFT FC: Antisense oligonucleotides for ICAM-1 attenuatereperfusion injury and renal failure in the rat. Kidney Int 50:473–480,1996

13. KELLY KJ, WILLIAMS WW JR, COLVIN RB, BONVENTRE JV: Antibodyto intercellular adhesion molecule-1 protects the kidney againstischemic injury. PNAS USA 91:812–816, 1994

Chiao et al: a-MSH inhibits neutrophil-independent renal injury 773

Page 10: α-Melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

14. TAKADA M, NADEAU KC, SHAW GD, MARQUETTE KA, TILNEY NL:The cytokine-adhesion molecule cascade in ischemia/reperfusion in-jury of the rat kidney–Inhibition by a soluble P-selectin ligand. J ClinInvest 99:2682–2690, 1997

15. LIU GS, STANLEY AW, DOWNEY JM: Ischaemic preconditioning is notdependent on neutrophils or glycolytic substrate at reperfusion inrabbit heart. Cardiovasc Res 26:1195–1198, 1992

16. LINAS S, WHITTENBURG D, REPINE JE: Nitric oxide prevents neutro-phil-mediated acute renal failure. Am J Physiol Renal Physiol 272:F48–F54, 1996

17. YU L, GENGARO PE, NIEDERBERGER M, BURKE TJ, SCHRIER RW:Nitric oxide: A mediator in rat tubular hypoxia/reoxygenation injury.Proc Natl Acad Sci USA 91:1691–1695, 1994

18. NOIRI E, PERESLENI T, MILLER F, GOLIGORSKY MS: In vivo targetingof inducible NO synthase with oligodeoxynucleotides protects ratkidney against ischemia. J Clin Invest 97:2377–2383, 1996

19. SCHWARTZ D, MENDONCA M, SCHWARTZ I, XIA YY, SATRIANO J,WILSON CB, BLANTZ RC: Inhibition of constitutive nitric oxidesynthase (NOS) by nitric oxide generated by inducible NOS afterlipopolysaccharide administration provokes renal dysfunction in rats.J Clin Invest 100:439–448, 1997

20. CHIAO H, FOSTER S, THOMAS R, LIPTON J, STAR RA: a-Melanocyte-stimulating hormone reduces endotoxin-induced liver inflammation.J Clin Invest 97:2038–2044, 1996

21. CATANIA A, LIPTON JM: The neuropeptide alpha-melanocyte-stimu-lating hormone: A key component of neuroimmunomodulation. Neu-roimmunomodulation 1:93–99, 1994

22. RAJORA N, CERIANI G, CATANIA A, STAR RA, MURPHY MT, LIPTONJM: a-MSH production, receptors, and influence on neopterin, in ahuman monocyte/macrophage cell line. J Leukoc Biol 59:248–253,1996

23. KOHDA Y, CRAIG L, CHIAO H, STAR R: Distribution of renalmelanocortin-1 a-MSH receptors. (abstract) J Am Soc Nephrol8:590A, 1997

24. CATANIA A, RAJORA N, CAPSONI F, MINONZIO F, STAR RA, LIPTONJL: The neuropeptide a-MSH has specific receptors on neutrophilsand reduces chemotaxis in vivo. Peptides 17:675–669, 1996

25. STAR RA, RAJORA N, HUANG J, CHAVEZ R, CATANIA A, LIPTON JM:Evidence of autocrine modulation of macrophage nitric oxide syn-thase by a-melanocyte-stimulating hormone. Proc Natl Acad Sci USA92:8016–8020, 1995

26. SOLEZ K, MOREL-MAROGER L, SRAER JD: The morphology of “acute

tubular necrosis” in man: Analysis of 57 renal biopsies and a compar-ison with the glycerol model. Medicine (Baltimore) 58:362–376, 1979

27. RABB H, O’MEARA YM, MADERNA P, COLEMAN P, BRADY HR:Leukocytes, cell adhesion molecules and ischemic acute renal failure.Kidney Int 51:1463–1468, 1997

28. HAUG CE, COLVIN RB, DELMONICO FL, AUCHINCLOSS H, TOLKOFF-RUBIN N, PREFFER FI, ROTHLEIN R, NORRIS S, SCHARSCHMIDT L,COSIMI AB: A phase I trial of immunosupression with anti-ICAM-1(CD54) mAb in renal allograft recipients. Transplantation 55:766–773,1993

29. LEVI M, MCDONALD LA, PREISIG PA, ALPERN RJ: Chronic Kdepletion stimulates rat renal brush-border membrane Na-citratecotransporter. Am J Physiol 261:F767–F773, 1991

30. CHOMCZYNSKI P, SACCHI N: Single-step method of RNA isolation byacid guanidinium thiocyanate-phenol-chloroform extraction. AnalBiochem 162:156–159, 1987

31. LINAS SL, WHITTENBURG D, REPINE JE: O2 metabolites cause reper-fusion injury after short but not prolonged renal ischemia. Am JPhysiol 253:F685–F691, 1987

32. LINAS SL, WHITTENBURG D, REPINE JE: Role of xanthine oxidase inischemia/reperfusion injury. Am J Physiol 258:F711–F716, 1990

33. KAINZ S, BERNARD J, SCHRAMEK H, PFALLER W: a-MSH amelioratesischemic acute renal failure in the blood free perfused kidney.(abstract) J Am Soc Nephrol 8:2743–2744, 1997

34. SAFIRSTEIN R, MEGYESI J, SAGGI SJ, PRICE PM, POON M, ROLLINS BJ,TAUBMAN MB: Expression of cytokine-like genes JE and KC isincreased during renal ischemia. Am J Physiol 261:F1095–F1101, 1991

35. YANG GM, KITAGAWA K, MATSUSHITA K, MABUCHI T, YAGITA Y,YANAGIHARA T, MATSUMOTO M: C57BL/6 strain is most susceptibleto cerebral ischemia following bilateral common carotid occlusionamong seven mouse strains: Selective neuronal death in the murinetransient forebrain ischemia. Brain Res 752:209–218, 1997

36. KELSO A, TROUTT AB, MARASKOVSKY E, GOUGH NM, MORRIS L,PECH MH, THOMSON JA: Heterogeneity in lymphokine profiles ofCD41 and CD81 T cells and clones activated in vivo and in vitro.Immunol Rev 123:85–114, 1991

37. SCHARTON-KERSTEN T, SCOTT P: The role of the innate immuneresponse in Th1 cell development following Leishmania major infec-tion. J Leukoc Biol 57:515–522, 1995

38. TANAKA Y, TAKAHASHI A, WATANABE K, TAKAYAMA K, YAHATA T,HABU S, NISHIMURA T: A pivotal role of IL-12 in Th1-dependentmouse liver injury. Int Immunol 8:569–576, 1996

Chiao et al: a-MSH inhibits neutrophil-independent renal injury774