ArticlePDF Available

Abstract and Figures

Blockade of NO production is followed by an increase in leukocyte rolling and adhesion resulting in some deleterious effects of ischemia. Preischemic administration of NO protects vascular integrity after reperfusion. Exogenous NO causes a direct reduction in leukocyte adhesion. This work was performed to test the hypothesis that exogenous NO administered during the preischemic period to the kidney alone, without coming into contact with the leukocytes, could also reduce leukocyte-endothelium adhesion. Adult rats were subjected to in situ isolation of the left kidney. Solutions were infused through the renal artery and drained through an incision in the renal vein, thus avoiding the systemic circulation. Group IC rats served as an ischemic control, and received 0.9% saline. Group NP received Na nitroprusside. Group S was a nonischemic control. Groups IC and NP were subjected to 75 min of renal ischemia. After this period, vascular structures were repaired and reperfusion allowed. A right nephrectomy was performed. Serum urea and creatinine, myeloperoxidase activity, and histopathological studies were carried out at different intervals after reperfusion. Survival at 15 days was 46%, 80%, and 100% in groups IC, NP, and S, respectively. Differences between groups for serum urea and creatinine were significant only during the first seven days. Myeloperoxidase values were significantly higher in group IC. All rats from group IC and only 20% from group NP showed histological evidence of necrosis. Thus, exogenous NO is protective and acts selectively upon the kidney, modulating its interactions with polymorphonuclear cells after ischemia/reperfusion.
Content may be subject to copyright.
Journal of Investigative Surgery, 14:1–8, 2001
Copyright c2001 Taylor & Francis
0894-1939/01 $12.00 + .00
Original Research
Exogenous Nitric Oxide Protects Kidney
from Ischemia/Reperfusion
Enrique S ´anchez-P ´erez-Verıa,
MD,
Fernando L ´opez-Neblina,
MD, MSc,
Eliseo Portilla, DVM, MSc,
Genaro Ort´ız, MD, MSc,
Alejandro Gonz ´alez-Ojeda,
MD,
and Rigoberto Alvares, MD
Surgical Research Division, Centro
de Investigaci ´on Biom ´edica de
Occidente, Instituto Mexicano del
Seguro Social, and Colegio de
Especialistas en Cirug´ıa General
del Estado de Jalisco A.C.,
Guadalajara, Jalisco, Mexico
ABSTRACT Blockade of NO production is followed by an increase in
leukocyte rolling and adhesion resulting in some deleterious effects of
ischemia. Preischemic administration of NO protects vascular integrity
after reperfusion. Exogenous NO causes a direct reduction in leuko-
cyte adhesion. This work was performed to test the hypothesis that
exogenous NO administered during the preischemic period to the kid-
ney alone, without coming into contact with the leukocytes, could also
reduce leukocyte–endothelium adhesion. Adult rats were subjected to
in situ isolation of the left kidney. Solutions were infused through the
renal artery and drained through an incision in the renal vein, thus avoid-
ing the systemic circulation. Group IC rats served as an ischemic control,
and received 0.9% saline. Group NP received Na nitroprusside. Group S
was a nonischemic control. Groups IC and NP were subjected to 75 min
of renal ischemia. After this period, vascular structures were repaired and
reperfusion allowed. A right nephrectomy was performed. Serum urea
and creatinine, myeloperoxidase activity, and histopathological stud-
ies were carried out at different intervals after reperfusion. Survival at
15 days was 46%, 80%, and 100% in groups IC, NP, and S, respec-
tively. Differences between groups for serum urea and creatinine were
significant only during the first seven days. Myeloperoxidase values were
significantly higher in group IC. All rats from group IC and only 20%
from group NP showed histological evidence of necrosis. Thus, exoge-
nous NO is protective and acts selectively upon the kidney, modulating
its interactions with polymorphonuclear cells after ischemia/reperfusion.
KEYWORDS adhesion molecules, endothelium, ischemia/reperfusion, kidney,
neutrophil infiltration, nitric oxide, rat
The authors thank Adolfo C ´
ardenas,
Ernesto Duarte, David Garc´
ıa, Alberto
Ramos, Sergio Rodr´
ıguez, and
Gabriela Santill´
an for their kind
assistance in this study.
Address correspondence to Dr. Eliseo
Portilla, CIBO, Surgical Research
Division, AP 1-3838, Guadalajara, Jal.,
44340 Mexico. E-mail: eportilla@mail.
udg.mx
1
Original Research
Nitric oxide (NO) has been identied as an
important molecular effector in a variety of
physiological and pathological conditions.
The role of NO in ischemia and reperfusion (I/R) is
still controversial. Some authors state that NO may
be deleterious after I/R due to its interaction with
oxygen-based free radicals upon reperfusion [13].
At the same time, others have found that when en-
dogenous NO production is blocked by antagonists
such as nitroG-methyl ester-L-arginine (L-NAME) or
NG-monomethyl-L-arginine (L-NMA), the deleteri-
ous effects of I/R, including leukocyte rolling and
adhesion in the postcapillary venules, are increased
[46]. In the same sense, exogenous NO donors and
endogenous NO have been shown to exert a protec-
tive effect from I/R and endotoxemic damage, by
maintaining vascular integrity [7, 8]. Some attribute
the protective effects of NO to its potent action
as a free-radical scavenger [9] and its ability to main-
tain the microvascular integrity as an endothelium-
derived relaxing factor [10]. It has been demons-
trated that NO decreases platelet adhesion, and that
this action is related to the inactivation of superoxide
anions [4, 11].
Furthermore, the results of recent research have
proposed that the principal benecial effect of NO is
exerted by the alteration of inammatory responses
through the modulation of interactions between
leukocytes and endothelial cells (ECs). It has been
shown, by previous treatment with exogenous NO
at different intervals before reperfusion, that there
is a decreased neutrophil inltration into the reper-
fused zone and an alteration to the rolling, acti-
vation, and adhesion of polymorphonuclear cells
(PMNs) [12, 14]. The protective effect observed in
Q1
these studies could be due to an interaction between
NO and inammatory cells, because NO donors
were administered to the systemic circulation after
ligation of the renal pedicle. In this technique, the
treatment did not have any effect on the ischemic
endothelium and most probably the locus of action
was on the circulating leukocytes. However, it is not
clear if exogenous NO exerts the same effect when
administered selectively and exclusively to the en-
dothelium of the preischemic kidney, avoiding con-
tact with leukocytes from the general circulation.
Therefore, the aim of the present study was to ex-
amine specically the role of exogenous NO donors
on the inammatory response observed after reper-
fusion. Exogenous NO donors were administered
exclusively to the kidney, thereby avoiding contact
with the systemic circulation.
METHODS
All experiments were carried out following insti-
tutional guidelines for the use and care of labora-
tory animals. In total, 105 Sprague-Dawley male rats
(250300 g) from our local animal facility were
subjected to 75 min of warm ischemia by clamp-
ing the vessels of the left renal pedicle. Briey, un-
der intraperitoneal sodium pentobarbital anesthesia
(60 mg/kg body weight) and through a midline la-
parotomy, the abdominal viscera were retracted to
the right side. Under an operating microscope, the
left renal hilus was dissected with ligation and divi-
sion of the adrenal and gonadal arteries and veins.
The perirenal fat was preserved. The renal vascular
pedicle was occluded temporarily using a microvas-
cular bulldog clamp on the renal artery near the
aorta, and an occluding, untied 6-0 silk suture on
the renal vein near the vena cava.
Immediately after this, a longitudinal draining
venotomy was performed on the renal vein between
the kidney and the silk, and for renal perfusion the
renal artery was cannulated with a 27-gauge needle
connected by an extension to a syringe. Thus, an
isolated perfusion circuit to the organ was created
by draining the solution through the venotomy and
removing it from the abdominal cavity with a pneu-
matic aspirator, thereby avoiding access to the sys-
temic circulation (Figure 1). This technique has been
tested and standardized in a previous study [15]. The
animals were divided into three groups. Group IC,
an ischemic control group (n=35), received 2 mL
of 0.9% saline solution infused over 1 min. Group
NP, an experimental group (n=35) with Na nitro-
prusside (Na-NP) as an exogenous NO donor, re-
ceived an infusion of 2 mL of dilute Na-NP (1.5 mg
Na- NP in 10 mL of 0.9% saline solution mixed im-
mediately before administration with 0.2 mL hep-
arinized autologous fresh blood). Addition of the
2 E. S ´
ANCHEZ-P ´
EREZ-VERD´
IA ET AL.
Q1: Au: Please cite ref 13.
Original Research
FIGURE 1 Model of exclusive in situ renal perfusion and
study protocol. Immediately after induction of warm ischemia
in the left kidney, cannulation of the renal artery and veno-
tomy were performed in order to ush the kidney with a so-
lution containing the drug, and to avoid its passage to the
systemic circulation. Reperfusion to the left kidney took place
after 75 min with the renal vessels repaired, followed by a right
nephrectomy.
autologous blood promotes the liberation of NO
from the nitroprusside molecules when these come
into contact with sulfhydryl groups on the erythro-
cyte membrane [16]. Three minutes were allowed
after infusion of the drug (sufcient time for NO
liberation and diffusion to the endothelium) and
a new perfusion with pure saline solution was per-
formed to wash out Na-NP metabolites. Group S was
a sham group (n=35). These animals were subjected
to the same procedure of in situ isolated renal perfu-
sion with physiologic solution only, but without any
period of ischemia.
Immediately after the end of perfusion, the renal
vessels were repaired with an uninterrupted suture
of 10-0 nylon in the vein and a single stitch point in
the puncture hole of the artery. The abdominal con-
tents were repositioned in the abdominal cavity, the
incision was covered with a moistened polyethylene
sheet, and 75 min of total ischemic time was allowed.
At the end of the ischemic period, the abdominal
cavity was reopened and the silk in the vein and the
clamp in the artery were removed allowing reper-
fusion of the kidney. In group S, reperfusion took
place immediately after renal vessel reconstruction,
thereby causing a total ischemic time of approxi-
mately 15 min for this group.
After achieving hemostasis and conrming an ade-
quate reperfusion of the kidney, a right nephrectomy
was performed. The abdominal wall was closed with
2-0 silk suture.
Animals were fed a standard pellet diet (Ralston
Rations, Kansas City, KS) and had free access to wa-
ter while they were observed for survival period of
15 days. Laboratory studies (n=10 per group) in-
cluded serum urea (SUr) and serum creatinine (SCr),
which were assessed at 24, 48, and 72 h and at 7 and
15 days. For these tests, 1 mL central venous blood
was removed and replaced with the same volume
of lactated Ringers solution. SUr and SCr values
were determined with colorimetric tests (diacetyl- Q2
thiosemicarbazide technique for SUr; Jaffe method
for SCr) in a Coleman Jr. II spectrophotometer
(Perkin Elmer Co, Oak Brook, IL).
Neutrophil inltration was estimated in ve ani-
mals from each group by measuring the activity of
myeloperoxidase (MPO) in the renal tissue. MPO
is a heme-containing enzyme found primarily in
PMNs, and can be used as an indirect estimator of
neutrophil inltration into the tissue. This was mea-
sured in accordance with the method described by
Bradley and Prietbat [17]. After a 2-h period of reper-
fusion, the abdominal aorta was cannulated and the
kidney ushed with cold saline solution (4C) and
immediately frozen and stored in liquid nitrogen
until the assay was performed. Kidney tissue sam-
ples were weighed and homogenized in 10 mL of
an aqueous solution B containing 0.021% K2PO4,
0.663% KH2PO4, and 0.5% hexadecyltrimethyl am-
monium bromide (HTBA; all obtained from Sigma
Chemical Co., St. Louis, MO). The homogenates
were frozenthawed 3 times in order to induce cellu-
lar lysis and then centrifuged at 2000 rpm for 10 min
at 0C. After this, 0.1 mL supernatant was added to
2.9 mL freshly prepared solution C (described later)
and assayed for MPO spectrophotometrically at a
xed wavelength of 460 nm (Sequoia-Turner Co.
model 690). The change in absorbance was mon-
itored every minute for 10 min. Solution C con-
tained 0.0105 g K2HPO4and 0.3315 g KH2PO2in
40 mL distilled water, to which was added 5 mL of
EXOGENOUS NO AND I/R 3
Q2: Au: Ok acetyl?
Original Research
0.017% solution of dianisidine in methanol and
5 mL of 0.006% hydrogen peroxide in distilled water.
One unit of MPO was dened as the amount that
degraded 1 mmol peroxide/min at 25C. The unit
value per gram of tissue was calculated using the fol-
lowing formula: (highest absorbance/10 min)/g tis-
sue used/0.0113 =MPO/g tissue, where 0.0113 is a
constant [18].
Five additional animals per group were used for
histological studies. Using light microscopy, sam-
ples of the kidney obtained 24 h after ischemia/
reperfusion were evaluated for acute tubular necro-
sis, vesicular degeneration, and hyperchromasia in
the proximal convoluted tubules, according to pre-
viously described criteria [19]. The evaluation of his-
tological damage was carried out in a blind study.
Statistical analysis included analysis of variance
(ANOVA), with Fvalues considered signicant at
an alpha value of .05 for parametric variables (SUr,
SCr, and MPO activity). Survival was plotted by
KaplanMeier curves, and the nal survival at
15 days was analyzed using Fishers exact test with
Yatess correction.
Q3
RESULTS
Survival
Figure 2 shows differences in survival curves be-
tween the three groups. Final survival at 15 days was
FIGURE 2 Survival curves. There was a statistically signi-
cant difference in survival at 15 days only between groups IC
andS(p<.05).
46% in group IC, 80% in group NP, and 100% in
group S. There were statistically signicant differ-
ences in survival between the groups IC and S from
day8to15(p<.001). No statistically signicant
difference was found between groups NP and S ( p=
.22). Although group NP showed a better survival
than group IC, this did not attain statistical signi-
cance ( p=.10).
Renal Function Test
Figure 3 illustrates the changing levels of serum
urea (SUr) at 24, 48, and 72 h and at 7 and 15 days
for each group. It can be observed that during the
rst 24 h, no statistically signicant differences ex-
isted between groups IC and NP, but they did exist at
this stage when compared with group S. Group NP
showed signicantly lower levels than group IC at
48 h, 72 h, and at 7 days after the procedure. No sig-
nicant differences existed between groups NP and S
after 48 h. Measurements on group S remained uni-
form and without signicant differences from pre-
viously standardized levels of healthy, normal rats
from our animal house. The results in the serum crea-
tinine levels (SCr) were similar to those described for
SUr, with statistically signicant differences ( p<.01)
between groups IC and NP at 72 h and 7 days
postreperfusion, but no difference between groups
NP and S (Figure 4).
FIGURE 3 Serum urea levels (SUr). Signicant differences
were found comparing the groups as follows: group IC vs.
group S, p<.001; group IC vs. standard control, ∗∗p<.01;
group IC vs. group NP, p<.05; group NP vs. group S, •• p<
.01. Group S maintained equal levels of serum urea compared
to levels observed in the standard control animals.
4 E. S ´
ANCHEZ-P ´
EREZ-VERD´
IA ET AL.
Q3: Au: Is ok?
Original Research
FIGURE 4 Serum creatinine levels (SCr). Statistically signif-
icant differences were observed between the groups IC and S
at 24 h with p<.05, and between the groups IC and NP with
∗∗p<.05. No statistically signicant differences were observed
at any time between groups NP and S.
Neutrophil Inltration
The values for MPO activity in renal tissue, as an
indirect marker of neutrophil inltration, are shown
in Figure 5. These correlated well with survival and
renal function. The levels found in group IC were sig-
nicantly higher when compared with both groups
NP and S. No signicant differences existed between
these latter two groups. Furthermore, there was no
difference between these groups and previously stan-
dardized levels in similar rats that had not undergone
any kind of procedure (Figure 5).
FIGURE 5 Myeloperoxidase (MPO) activity in renal tissue
as a marker of neutrophil inltration. In group IC, the levels of
MPO were signicantly higher, p<.05, compared to all other
groups. No differences were found between groups NP and S,
or between these and previously standardized levels in similar
healthy normal rats from our laboratory (standard control).
Histology
No signicant changes were observed in group S,
and the percentage of animals with changes was
similar to that from kidneys of normal healthy rats
FIGURE 6 In the proximal convoluted tubules, (a) group IC
shows necrosis, nuclear hyperchromasia, and cloudy swelling
with vesicular degeneration of cells resulting in loss of the
cytoarchitecture at the luminal border of the tubule. The
glomeruli exhibit alterations resulting in a loss of integrity in
Bowmans capsule. (b) Group NP shows 3040% of protec-
tion, as revealed by reduced necrosis and vesicular degenera-
tion in the proximal convoluted tubule. (c) Neither group S nor
standard control samples (not shown) showed any signicant
alterations.
EXOGENOUS NO AND I/R 5
Q4: Au: Please explain art asterisks, in legend.
Original Research
TABLE 1 Histopathological effects on kidney following intra-arterial administration of exogenous nitric oxide donor during
warm renal ischemia (% of rats with morphologically signicant change)
Vesicular degeneration of convoluted
Group Hyperchromasia Necrosis proximal tubules
Standard control 0 0 0
Group IC 100 80 100
(ischemic control)
Group NP 60 40 60
(ischemia +Na-NP)
Group S 0 0 20
(sham)
from our laboratory. In group IC, necrosis was
present in all three segments of the proximal con-
voluted tubules in four of ve samples, and all ex-
hibited hyperchromasia and vesicular degeneration.
In group NP, necrosis was observed in only 20%
of animals, whereas 60% exhibited hyperchroma-
sia and vesicular degeneration (see Figure 6 and
Table 1).
DISCUSSION
Leukocyteendothelial cell interaction is a cru-
cial event in I/R injury [2025]. The decrease in
NO and prostacyclin release, and enhanced produc-
tion of pro-inammatory substances (oxygen free
radicals, endothelins, lipid mediators, complement
systems and cytokines) promote PMN recruitment
and adherence to the injured area during reperfusion
[2630].
Leukocyte recruitment is determined by a series
of mechanisms described as activation, rolling, adhe-
sion, and migration [31]. Each stage is characterized
by the expression of different kinds of molecules at
the surfaces of the leukocyte and EC, namely, inte-
grins in the former and the immunoglobulin super-
family in the latter [28, 32]. Many proinammatory
mediators have the ability to initiate and maintain
this process. However, much less is known about the
endogenous mediators that could reverse or prevent
this event. NO may be an antiadherent and/or anti-
inammatory molecule.
In previous studies, exogenous NO administered
systemically ameliorated neutrophil inltration and
showed a protective effect against I/R injury [12, 33].
Studies using intravital microscopy in a mesenteric
I/R model in the rat showed that the systemic
administration of NO reduced PMN rolling and ad-
herence in postcapillary venules [13].
The model used in this work allows us to differen-
tiate the systemic actions of NO from those on the
kidney alone. We changed the administration route
to an in situ isolated renal perfusion, and adminis-
tered an NO donor through the renal artery with the
aim of obtaining a selective effect on the ischemic
kidney and its endothelium. Our hypothesis was that
exogenous NO would modify leukocyteEC inter-
actions. In this model, circulating leukocytes did not
have contact with the exogenous NO. The signicant
reduction of PMN inltration into the reperfused
kidney, and the decreased mortality with improve-
ment in renal function, can be interpreted as a re-
duction in nephron damage. This was corroborated
by light microscopy.
The I/R protective effect observed in our model
cannot be due to the vasodilator activity of Na nitro-
prusside, because this action disappears after 5 min
of administration [17]. It was administered 75 min
before reperfusion took place, so it can be assumed
that any vasodilator effect was already absent. As
the endothelial cell is the major component for
neutrophilendothelium interaction in the organ,
we believe that the benecial action of exogenous
NO could be due to inhibitory signals left in the EC,
which can in some way block the expression of ad-
hesion molecules following the period of ischemia.
This is supported by the demonstration by Lefer that
NO blocks the expression of ICAM-1 in endothelial
cell cultures [34].
Although it can be speculated that exogenous
NO acts over the endothelium, modulating its inter-
actions with PMN after ischemia/reperfusion, fur-
ther molecular studies are necessary to determine
6 E. S ´
ANCHEZ-P ´
EREZ-VERD´
IA ET AL.
Original Research
precisely the level of action by which NO is able
to reduce EC activation and dysfunction.
NO could be used to pretreat organs upon is-
chemia, thereby reducing reperfusion injury. Further-
more, secondary and undesirable effects following
systemic administration of NO, as well as its hy-
potensive effect, can be avoided.
REFERENCES
1. Peplow PV, Mikhailidis DP. Platelet-activating factor (PAF)
and its relation to prostaglandin, leukotrienes and
other aspects of arachidonate metabolism. Prostaglandins
Leukocyte Essential Fatty Acids. 1990;41:7182.
2. Dahl ´en SE, Bj ´ork J. Leukotrienes promote plasma leakage
and leukocyte adhesion in postcapillar venules: In vivo ef-
fects with relevance to the acute inammatory response.
Proc Natl Acad Sci USA. 1981;78:38873891.
3. Padmaja S, Huie RE. The reaction of nitric oxide with or-
ganic peroxyl radicals. Biochem Biophys Res Commun.
1993;195:539544.
4. Kubes P, Kanawar S, Niu XF, Gaboury JP. NO synthesis
inhibition induced leukocyte adhesion via superoxide and
mast cells. FASEB J. 1993;7:1293.
5. Mueller AR, Platz KP, Langrehr JM, Hoffman RA,
Nussler AK, Nalesnik M, Billiar TR, Schraut WH. The effects
of administration of nitric oxide inhibitors during small
bowel preservation and reperfusion. Transplantation.
1994;58:13091316.
6. Aoki N, Johnson G, Lefer AM. Benecial effects of two
forms of NO administration in feline splanchnic artery oc-
clusion shock. Am J Physiol. 1990;258:G275.
7. Wright CE, Rees DD, Moncada S. Protective and patho-
logical roles of nitric oxide in endotoxic shock. Cardiovasc
Res. 1992;26:48.
8. Hutcheson JR, Whittle BJR, Boughton-Smith NK. Role of
nitric oxide in maintaining vascular integrity in endotoxin-
induced acute intestinal damage in the rat. Br J Pharmacol.
1990;101:815.
9. Gaobury J, Woodman RC, Granger DN. NO prevents
leukocyte adherence: Role of superoxide. Am J Physiol.
1993;265:H862.
10. Lefer AM, Lefer DJ. Pharmacology of the endothelium
in ischemiareperfusion and circulatory shock. Annu Rev
Pharmacol Toxicol. 1993;33:7190.
11. Radomski MW, Palmer RM, Moncada S. The role of nitric
oxide and cGMP in platelet adhesion to vascular endothe-
lium. Biochem Biophys Res Commun. 1987;148:1482
1489.
12. L ´opez-Neblina F, Paez-Rollys A, Toledo-Pereyra LH. Mod-
ulation of neutrophil inltration through nitric oxide in
the ischemic rat kidney. Transplant Proc. 1995;27:1883
1885.
13. L ´opez-Neblina F, Paez-Rollys A, Garc´ı a Criado FJ. El ´oxido
n´ı trico ex ´ogeno previene el rodamiento y la adhesion
leucocitaria en isquemia mesenterica en la rata. Cirujano
General. 1995;17:9093.
14. L ´opez-Neblina F, Toledo-Pereyra LH, Mirmiran R,
Paez-Rollys A. Time dependence of Na-nitroprusside
administration in the prevention of neutrophil inl-
tration in the rat ischemic kidney. Transplantation.
1996;61:179183.
15. S ´anchez-P ´erez-Verd´ı a E, Portilla E, L ´opez-Neblina F,
Gonz ´alez-Ojeda A. Tratamiento endotelial exclusivo
in vivo del ri ˜n´on de la rata. Cirujano General. 1997;19:47
48.
16. Gerber JG, Nies AS. Antihypertensive agents and the drug
therapy of hypertension. In: Goodman A, Rall TW, Nies A,
Taylor P, eds. The Pharmacological Basis of Therapeutics.
M´exico: Editorial M ´edica Panamericana; 1991:766793.
17. Bradley P, Prietbat D. Measurement of cutaneous inam-
mation: stimulation of neutrophil content with enzyme
marker. J Invest Dermatol. 1982;78:206.
18. Williams R, Paterson CA, Eakins KE, Nhattacherjee P.
Quantication of ocular inammation: evaluation of
polymorphonuclear leucocyte inltration by measuring
myeloperoxidase activity. Curr Eye Res. 1983;2:465.
19. Jablonski P, Howden B, Rae D, Birreli C, Marshall V,
Tange J. An experimental model for assessment of
renal recovery from warm ischemia. Transplantation.
1983;35:198.
20. Lee C, Kerrigan CL. Neutrophil localization following
reperfusion of an ischemic myocutaneous ap. Plast Re-
constr Surg. 1992;89:916923.
21. Romson JL, Hook BG. Reduction of the extent of ischemic
myocardial injury by neutrophil depletion in the dog.
Circulation. 1983;67:10161023.
22. Hernandez LA, Grisham MB. Role of neutrophils in
ischemia-reperfusion-induced microvascular injury. Am J
Physiol. 1987;253:H699H703.
23. Litt MR, Jeremy RW. Neutrophil depletion limited to reper-
fusion reduces myocardial infarct size after 90 minutes of
ischemia. Circulation. 1989;80:18161827.
24. Lee C, Kerrigan CL. Skin ap ischemia and altered neu-
trophil function. Surg Forum. 1990;40:593.
25. Eppinger MJ, Jones ML, Deeb GM, Bolling SF, Ward PA.
Pattern of injury and the role of neutrophils in reperfusion
injury of rat lung. J Surg Res. 1995;58:713718.
26. Bulkey GB. Mediators of splanchnic organ injury: overview
and perspective. In: Marston A, Bulkley G, Fiddian-Green
RG, Haglund UH, eds. Splanchnic Ischemia and Multiple
Organ Failure. London: Edward Arnold; 1989:191193.
27. Lefer AM, Tsao PS, Lefer DJ. Role of endothelial dysfunc-
tion in the pathogenesis of reperfusion injury after my-
ocardial ischemia. FASEB J. 1991;5:20292034.
28. Lorand DE, Patel KD, McIntyre TM. Coexpression
GMP-140 and PAF by endothelium stimulated by his-
tamine or thrombin: A justacrine system for adhesion and
activation of neutrophil. J Cell Biol. 1991;115:223234.
29. L ´opez-Neblina F, S ´anchez P ´erez-Verd´ı a E, Alvarez QR,
Perea SA. Role of nitric oxide in ischemia and reperfusion.
Cirujano General. 1997;19:309320.
30. Lindstrom P, Lerner R. Rapid adhesive responses of en-
dothelial cells and neutrophils induced by leukotriene B4
are mediated by leukocyte adhesion protein CD18. Scand
J Immunol. 1990;31:737744.
EXOGENOUS NO AND I/R 7
Original Research
31. L ´opez-Neblina F. Mol ´eculas de adhesi ´on, isquemia y
cirug´ı a. Cirujano General. 1996;18:183187.
32. Zimmerman GA, McIntyre TM. Endothelial cell-associated
platelet-activating factor: A novel mechanism for sig-
naling intercellular adhesion. J Cell Biol. 1990;110:529
540.
33. L ´opez-Neblina F, Paez AJ, Toledo LH. Role of nitric ox-
ide in renal ischemia/reperfusion in the rat. Circ Shock.
1995;44:9195.
34. Lefer DJ. The role of NO in the modulation of leukocyte
adhesion to the endothelium. IBC’s 3rd Symposium on
Nitric Oxide. Philadelphia, PA; 1994.
8 E. S ´
ANCHEZ-P ´
EREZ-VERD´
IA ET AL.
... Death of renal epithelial cells and delayed recovery of kidney function in the posttransplantation period can lead to interstitial fibrosis and may aggravate chronic kidney disease 3 . Renal I/R has been analyzed in experimental models using rodents 4,5 , in which many of allopathic and phytotherapeutic drugs have already been examined. Some of them were effective and others ineffective 6,7 . ...
Article
Full-text available
Purpose: To evaluate the effect of tadalafil in renal ischemia/reperfusion (I/R) injury in rats Methods: Group I/R saline rats (n=6) were subjected to 45 minutes of left renal ischemia and treated with saline; the I/R tadalafil rats (n=6) received oral 10mg/kg tadalafil microemulsion one hour before ischemia. In both groups, 8 hours after ischemia, laboratory analysis were performed Results: Better tissue perfusion was lower in ischemic left/kidney than in right/kidney in saline group, suggesting reduced kidney clearance. Fluorescence in left/kidneys of tadalafil treated rats was lower than in right/kidneys (difference not significant). The fluorescence signal intensity in kidneys of tadafil treated rats was higher than in saline rats. TNF-α levels were significantly lower in I/R tadalafil group rats compared to I/R saline group (154±10.3 vs 391.3±12.3), as well as IL-1β (163.4±13.2 vs 279±11.5pg/dL), and IL-6 (122.9±8.1 vs 173.7±6.3 respectively; p=0.0001). Urea, creatinine and C-reactive protein were significantly lower in tadafil treated rats then in saline group Conclusion: Tadalafil therapy decreased the expression of circulating pro-inflammatory cytokines in a renal I/R rodent model, while improving kidney function proofs.
... In cardiac surgery, SNP decreases cardiac cytokine release [141–143] and improves postischemic cardiac function [143]. In experimental models of ischemia-reperfusion injury, such as those in kidney [144] and lung [145], SNP attenuates the expression of proinflammatory cytokines and reduces leukocyte-endothelium adhesion, respectively. ...
Article
Full-text available
Cerebral ischemia initiates a cascade of detrimental events including glutamate-associated excitotoxicity, intracellular calcium accumulation, formation of Reactive oxygen species (ROS), membrane lipid degradation, and DNA damage, which lead to the disruption of cellular homeostasis and structural damage of ischemic brain tissue. Cerebral ischemia also triggers acute inflammation, which exacerbates primary brain damage. Therefore, reducing oxidative stress (OS) and downregulating the inflammatory response are options that merit consideration as potential therapeutic targets for ischemic stroke. Consequently, agents capable of modulating both elements will constitute promising therapeutic solutions because clinically effective neuroprotectants have not yet been discovered and no specific therapy for stroke is available to date. Because of their ability to modulate both oxidative stress and the inflammatory response, much attention has been focused on the role of nitric oxide donors (NOD) as neuroprotective agents in the pathophysiology of cerebral ischemia-reperfusion injury. Given their short therapeutic window, NOD appears to be appropriate for use during neurosurgical procedures involving transient arterial occlusions, or in very early treatment of acute ischemic stroke, and also possibly as complementary treatment for neurodegenerative diseases such as Parkinson or Alzheimer, where oxidative stress is an important promoter of damage. In the present paper, we focus on the role of NOD as possible neuroprotective therapeutic agents for ischemia/reperfusion treatment.
Article
Full-text available
Inflammation and oxidative stress upset memory. We explored influence of sodium nitroprusside (SNP) on memory deficits resulted from lipopolysaccharide (LPS).Groups include control, LPS, LPS + SNP 1 mg/kg, LPS + SNP 2 mg/kg, and LPS + SNP 3 mg/kg. Morris water maze and passive avoidance tests and biochemical measurements were carried out.In Morris water maze, LPS prolonged time and distance for finding the platform. In probe trial, it diminished time spent and traveled distance in the target zone. Injection of 2 and 3 mg/kg of SNP overturned the effect of LPS. In passive avoidance task, LPS postponed entrance into darkroom and reduced time spent in light room and incremented time spent in darkroom in 3, 24, and 72 h after electrical shock. All three doses of SNP restored the effects of LPS. Biochemical experiments confirmed that LPS elevated interleukin‐6 and malondialdehyde concentration and declined total thiol content and superoxide dismutase and catalase activity in the hippocampus and cortex tissues. SNP particularly at a 3 mg/kg dose ameliorated LPS effects on these parameters.SNP attenuated memory disabilities resulting from LPS through modifying inflammation and boosting antioxidant defense.
Article
Renal ischemia-reperfusion (I/R) injury is associated with delayed graft function and decreased long-term allograft function. It is also a major cause of acute renal failure. It is commonly seen in the field of renal surgery or transplantation. Production of reactive oxygen species (ROS) is a major patho-physiological component producing the injury after I/R. Inflammatory cytokines were accused to have a casual role of remote systemic effects of I/R. Ischemic preconditioning (IP) is the phenomenon that a prior ischemic stress renders the organ resistant to a subsequent ischemic insult. However the mechanism of IP protection is not completely known.
Article
The role of nitric oxide synthases (NOS) and the nitric oxide (NO) substrate l ‐arginine in renal ischaemia–reperfusion (I/R) has been studied extensively. However, the results reported are often controversial. In the present study, we examined the effects of the neuronal (n) NOS inhibitor 7‐nitroindazole (7‐NI) and l ‐arginine administration on renal I/R injury and the renal NO system in rats. Following 7 days pretreatment with 7‐NI (50 mg/kg per day), l ‐arginine (2 g/kg per day) or vehicle (dimethylsulphoxide : sesame oil, 1 : 9), the left renal vascular pedicles were clamped for 50 min in male Sprague‐Dawley rats and kidneys were removed 24 h after reperfusion ( n = 7/group). Neither 7‐NI nor l ‐arginine had any effect on parameters of renal function, the grade of tissue injury or the number of terminal deoxyribonucleotidyl transferase‐mediated dUTP–digoxigenin nick end‐labelling (TUNEL)‐positive tubular cells compared with vehicle‐treated rats. 7‐Nitroindazole decreased nNOS mRNA expression and inducible (i) NOS protein levels, but had no effect on endothelial NOS expression. l ‐Arginine supplementation increased mRNA expression of all NOS isoforms, but only increased protein expression of iNOS. The results of the present study demonstrate that selective inhibition of nNOS has no effect on renal injury, indicating that nNOS does not play a central role in the pathophysiology of renal I/R. In addition, although l ‐arginine has no effect on renal I/R injury in the model used in the present study, its administration increases the mRNA expression of NOS isoforms.
Article
We studied whether melatonin is able to reduce organ damage during renal ischemia/reperfusion via its effects on the oxidative response in early and late reperfusion. Renal ischemia/reperfusion injury (I/R) was induced in two groups of rats by 75 min occlusion of the left renal artery and vein and right nephrectomy, followed by reperfusion. The formation of reactive oxygen species was evaluated in the early reperfusion phase (60 min) by lipid peroxidation products and glutathione assay. In the late reperfusion phase (24 h) tissue neutrophil infiltration, inducible nitric oxide synthase (iNOS) gene expression, and histopathology were evaluated. Groups received either systemic melatonin (MEL) or normal saline (NS). There were two nonischemic sham control groups, one with and another without melatonin (S+MEL and S). Creatinine was higher in the NS group at all times. A reduction in glutathione and increases in lipid peroxidation products and myeloperoxidase activity induced by I/R indicated renal injury involving reactive oxygen formation. Melatonin reversed this oxidant response and reduced the rise in creatinine and iNOS expression. Seven-day group survivals were 5/10 for NS, 8/10 for MEL, and 10/10 for both Sham groups. Exogenous melatonin is able to preserve renal functional status following I/R-induced injury by increasing glutathione and reducing lipid peroxidation in the early reperfusion phase, without any apparent effect on neutrophil infiltration in the late reperfusion phase.
Article
To elucidate the role of nitric oxide (NO) in the pathogenesis of ischemic acute renal failure, we examined the effects of (+/-)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide (FK409) and N(G)-nitro-L-arginine methyl ester (L-NAME) as a NO donor and a non-selective NO synthase inhibitor on ischemia/reperfusion-induced renal injury and renal endothelin-1 content. Ischemic acute renal failure was induced by occlusion of the left renal artery and vein for 45 min followed by reperfusion, 2 weeks after contralateral nephrectomy. At 24 h after reperfusion, renal function in untreated acute renal failure rats markedly decreased and histological examination revealed severe renal damage. In addition, increases in renal endothelin-1 contents were evident in the acute renal failure rats at 2, 6, and 24 h after reperfusion, respectively. Pretreatment with FK409 (1 or 3 mg/kg, i.v.) attenuated ischemia/reperfusion-induced renal dysfunction, histological damage, and endothelin-1 overproduction after reperfusion. In contrast, pretreatment with L-NAME (1 or 10 mg/kg, i.v.) aggravated renal injuries of acute renal failure rats at 24 h after reperfusion, and the effect is accompanied by further increases in the renal endothelin-1 content at 2 and 6 h, but not at 24 h, after reperfusion. These results suggest that suppressive effects of NO on the renal endothelin-1 overproduction induced by ischemia/reperfusion in an early phase are probably responsible for the protective effect of NO against ischemic acute renal failure.
Article
Recent studies show that melatonin reduces the blood pressure (BP) and ischemia/reperfusion (I/R)-induced damage. This study was designed to investigate the effects of melatonin on the renal I/R injury in rats given the nitric oxide synthase (NOS) inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME). After right nephrectomy, I/R was induced by occlusion of the left renal vessels for 60 min, followed by 24h reperfusion. The administration of melatonin significantly attenuated BP in NOS-inhibited hypertensive rats. Malondialdehyde (MDA) levels, a stable metabolite of the free-radical-mediated lipid peroxidation cascade, were found to be significantly higher in the I/R group (3.48+/-0.2mg/l serum) than in the control group (2.69+/-0.2mg/l serum). L-NAME (40 mgkg(-1) for 15 days)+I/R significantly increased the MDA levels compared to I/R alone. Melatonin administration to L-NAME rats significantly reduced the MDA values resulting from I/R. We also demonstrated that I/R, and especially L-NAME+I/R, lead to structural changes in the kidney and that melatonin attenuates these changes. These results suggest that melatonin reduces BP and I/R injury in NOS inhibited rats by L-NAME.
Article
The current shortage of organ donors has led many centers to use marginal and nonheart-beating donors (NHBDs). Recent research has implicated the infiltration of lymphocytes as an important mediator of ischemia-reperfusion injury (IRI). FTY720 is an immunosuppressant that promotes lymphocyte sequestration into lymph nodes. The purpose of this study was to examine the potential for FTY720 to abrogate IRI when subjected to increasing ischemic times. Male Sprague-Dawley rats underwent bilateral flank incision with removal of the right kidney and clamping of the left hilum. Groups were divided into ischemia times of 45, 55, and 65min; each group was further divided into a control group (IRI only), IRI+FTY720 (1 mg/kg/d), and IRI+cyclosporine (15 mg/kg/d), n=4 per group. Thre days after 45 min of ischemia, serum creatinine in the ischemia only (477+/-37 micromol/L) and cyclosporine groups (698+/-32 micromol/L) was significantly increased compared with the FTY720-treated animals (194+/-66 micromol/L). The beneficial effect of FTY720 was also observed at 55 and 65 min; indeed, FTY720-treated animals demonstrated signs of recovery from 65 min of ischemia whereas control and cyclosporine-treated animals required sacrifice between days 3 and 5. Treatment with FTY720 reduced renal damage assessed histologically and also reduced apoptosis and increased cell proliferation. Treatment with FTY720 reduced IRI and prevented unrecoverable acute renal failure after significant ischemic injury. This study suggests that FTY720 may help improve the quality of grafts from NHBD and marginal donors by abrogating the IRI insult.
Article
Many studies indicate that the production of reactive oxygen species (ROS) after renal ischemia/reperfusion (I/R) may initiate the cascade of cellular injury. It has been demonstrated that ozone oxidative preconditioning (OzoneOP) may prevent the damage induced by ROS and attenuate renal I/R injury. On the basis of those results, we postulated that OzoneOP was similar to the ischemic preconditioning (IP). The aim of our present work was to assess whether the combination of OzoneOP and IP provided synergistic protection. Seven groups of rats were classified as follows: 1) sham-operated control; 2) I/R; 3) OzoneOP+I/R; 4) IP+I/R; 5) OzoneOP+IP+I/R; 6) O2+I/R; 7) sham-operated control+OzoneOP. Rats were sacrificed at 24 h after I/R injury. Serum and tissue were taken to determine urea nitrogen (BUN), creatinine (Cr), nitric oxide (NO), histological examination, and NO synthase (endothelial, eNOS and inducible, iNOS) expression. Malondialdehyde (MDA) content, glutathione (GSH) content, superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activity were determined in renal tissue. Renal dysfunction, histological damage, and renal oxidative stress were significantly improved by OzoneOP or IP alone. OzoneOP+IP could not further relieve severe renal damage. Either IP or OzoneOP treatment alone increased NO release and NO synthase (endothelial, eNOS and inducible, iNOS) expression. The combination of OzoneOP and IP could not further enhance NO levels and NOS expression. These findings indicate that both of the preconditioning settings shared similar mechanisms of protection in the parameters measured. However, OzoneOP combined with IP had no synergistic effect. IP and OzoneOP appeared to share a common mediator: NO. These findings suggested the potential role of OzoneOP against renal failure during surgery or transplantation.
Article
Full-text available
Endothelial dysfunction occurs after myo- cardial ischemia and reperfusion characterized by a marked reduction in endothelium-dependent relaxation (EDR) due to reduced release or action of endothelium- derived relaxing factor (EDRF). This reduced EDR oc- curs in coronary rings isolated from cats 2.5 mm after reperfusion and in isolated perfused cat hearts 2.5 nun af- ter reperfusion. No decrease in EDR occurs before reper- fusion in either preparation, suggesting that this impair- ment in EDR occurs during reperfusion. The decrease in EDR occurs soon after the generation of superoxide radi- cals by the reperfused coronary endothelium. Accumula- tion of neutrophils and myocardial cell injury does not occur until 3-4.5 h after reperfusion. Thus, endothelial
Article
Endothelial dysfunction occurs after myocardial ischemia and reperfusion characterized by a marked reduction in endothelium-dependent relaxation (EDR) due to reduced release or action of endothelium-derived relaxing factor (EDRF). This reduced EDR occurs in coronary rings isolated from cats 2.5 min after reperfusion and in isolated perfused cat hearts 2.5 min after reperfusion. No decrease in EDR occurs before reperfusion in either preparation, suggesting that this impairment in EDR occurs during reperfusion. The decrease in EDR occurs soon after the generation of superoxide radicals by the reperfused coronary endothelium. Accumulation of neutrophils and myocardial cell injury does not occur until 3-4.5 h after reperfusion. Thus, endothelial generation of superoxide radicals acts as a trigger mechanism for endothelial dysfunction which is then amplified by neutrophil adherence and diapedesis into the ischemic region enhancing post-reperfusion ischemic injury. Agents that preserve endothelial function or inhibit neutrophil activation (e.g., superoxide dismutase, prostacyclin analogs, TGF-beta, antibodies to adhesive proteins) can protect against endothelial dysfunction and myocardial injury, if administered before reperfusion.
Article
We examined the hypothesis that myeloperoxidase (MPO), a plentiful constituent of neutrophils, might serve as a marker for tissue neutrophil content. To completely extract MPO from either neutrophils or skin, hexadecyltrimethylammonium bromide (HTAB) was used to solubilize the enzyme. With this detergent treatment, 97.8 +/- 0.2% of total recoverable MPO was extracted from neutrophils with a single HTAB treatment; 93.1 +/- 1.0% was solubilized with a single treatment of skin. Neutrophil MPO was directly related to neutrophil number; with the dianisidine-H2O2 assay as few as 10(4) neutrophils could be detected. The background level of MPO within uninflamed tissue was 0.385 +/- 0.018 units per gram of tissue, equivalent to only 7.64 +/- 0.36 X 10(5) neutrophils. In experimental staphylococcal infection, skin specimens contained 34.8 +/- 3.8 units MPO per gram, equivalent to 8.55 +/- 0.93 X 10(7) neutrophils. These studies demonstrate that MPO can be used as a marker for skin neutrophil content: it is recoverable from skin in soluble form, and is directly related to neutrophil number. Further, normal skin possesses a low background of MPO compared to that of inflamed skin.
Article
The binding of neutrophils (polymorphonuclear leukocytes [PMNs]) to endothelial cells (ECs) presents special requirements in the regulation of intercellular adhesion. ECs that are stimulated by certain agonists, including thrombin and cytokines (tumor necrosis factor alpha, interleukin-1), generate molecular signals that induce the adhesion of PMNs (endothelial cell-dependent neutrophil adhesion). Our experiments demonstrate that the mechanism of binding induced by thrombin is distinct from that induced by the cytokines based on the time courses, the requirement for protein synthesis, and differential binding of HL60 promyelocytic leukemia cells to ECs activated by the two classes of agonists. The rapid EC-dependent PMN adhesion (initiated in minutes) that occurs when the ECs are stimulated by thrombin is temporally coupled with the accumulation of platelet-activating factor, a biologically active phosphoglyceride that remains associated with ECs and that activates PMNs by binding to a cell surface receptor. A portion of the newly synthesized platelet-activating factor (PAF) is on the EC surface, as demonstrated by experiments in which the rate of hydrolysis of PAF synthesized by activated ECs was accelerated by extracellular PAF acetylhydrolase. When ECs were treated with exogenous PAF they became adhesive for PMNs; the PMN binding was prevented by incubating the ECs with PAF acetylhydrolase or by treating the PMNs with competitive PAF receptor antagonists. Thus PAF associated with the EC plasma membrane induces PMN binding, an observation supported by experiments in which PAF in model membranes (liposomes) stimulated rapid PMN adhesion to ECs and to cell-free surfaces. In addition, competitive antagonists of the PAF receptor inhibited the binding of PMNs to ECs activated by thrombin and other rapidly acting agonists, but not to ECs activated by tumor necrosis factor alpha, indicating that PAF that is endogenously synthesized by ECs can mediate neutrophil adhesion. These experiments demonstrate a novel mechanism by which a cell-associated phospholipid, PAF, can serve as a signal for an intercellular adhesive event.
Article
The aim was to investigate the effects of NG-monomethyl-L-arginine (L-NMMA), an inhibitor of both the constitutive (Ca2+ dependent) and inducible (Ca2+ independent) nitric oxide (NO) synthases, or of pretreatment with the glucocorticoid dexamethasone, an inhibitor of the induction of the Ca2+ independent NO synthase, on lipopolysaccharide induced shock in the anaesthetised rabbit. Mean arterial blood pressure, and blood flow in the portal vein, hepatic artery, and hindquarter vascular beds were measured in 49 halothane anaesthetised New Zealand White rabbits given lipopolysaccharide (Salmonella minnesota, 500 micrograms.kg-1 intravenously). The effects of pre- or post-lipopolysaccharide treatment with L-NMMA (300 mg.kg-1 intravenously) and of pretreatment with dexamethasone (3 mg.kg-1 intravenously) were determined. The effect of the NO donor S-nitroso-N-acetyl-penicillamine (SNAP, 300 micrograms.kg-1.h-1 intravenously) in animals treated with lipopolysaccharide and L-NMMA was also studied. Lipopolysaccharide elicited an initial transient fall in mean arterial pressure and decreases in blood flow in the vascular beds, followed by a progressive fall in mean arterial pressure. L-NMMA when given either before or after lipopolysaccharide markedly exacerbated its effects and resulted in severe hypotension, intense vasoconstriction, and increased mortality. Pretreatment with dexamethasone had no effect on the initial haemodynamic changes following lipopolysaccharide, but prevented the subsequent fall in mean arterial pressure observed in animals treated with lipopolysaccharide alone. Dexamethasone failed, however, to protect animals also treated with L-NMMA before lipopolysaccharide. Animals pretreated with L-NMMA and SNAP showed reduced haemodynamic changes when compared with controls (lipopolysaccharide only) or lipopolysaccharide and L-NMMA treated animals. Inhibition of both constitutive and inducible NO synthases during endotoxaemia is deleterious. This can be overcome by replacing NO intravenously with a donor of NO. Selective inhibition of the inducible NO synthase may, however, be beneficial in shock.
Article
The neutrophil has been implicated as a source of oxygen free radicals provoking the reperfusion injury in various ischemic organs. This provided the motivation to explore the pathophysiologic role of the neutrophil in a swine model of postischemic latissimus dorsi myocutaneous flaps. Neutrophil function, neutrophil sequestration, and the anatomic distribution of muscle injury were estimated following a 6- to 8-hour global ischemic insult. Neutrophil function as measured by phorbol myristate acetate-stimulated superoxide production was found to be enhanced on reperfusion of ischemic flaps (n = 17). Neutrophil sequestration estimated from the arterial-venous difference of flap blood (n = 12) demonstrated that postischemic flaps more avidly sequester neutrophils than nonischemic flaps. The anatomic distribution of muscle injury (n = 7) was predominantly localized to the proximal portion of the ischemic flap. The enhanced functional response exhibited by neutrophils reperfusing an ischemic myocutaneous flap supports an active neutrophil role in the mediation of reperfusion injury.