Vous êtes sur la page 1sur 23

Pathophysiology

of Ischemic Acute
Renal Failure
Michael S. Goligorsky
Wilfred Lieberthal

A
cute renal failure (ARF) is a syndrome characterized by an
abrupt and reversible kidney dysfunction. The spectrum of
inciting factors is broad: from ischemic and nephrotoxic agents
to a variety of endotoxemic states and syndrome of multiple organ
failure. The pathophysiology of ARF includes vascular, glomerular
and tubular dysfunction which, depending on the actual offending
stimulus, vary in the severity and time of appearance. Hemodynamic
compromise prevails in cases when noxious stimuli are related to
hypotension and septicemia, leading to renal hypoperfusion with sec-
ondary tubular changes (described in Chapter 13). Nephrotoxic
offenders usually result in primary tubular epithelial cell injury,
though endothelial cell dysfunction can also occur, leading to the
eventual cessation of glomerular filtration. This latter effect is a con-
sequence of the combined action of tubular obstruction and activation
of tubuloglomerular feedback mechanism. In the following pages we
shall review the existing concepts on the phenomenology of ARF
including the mechanisms of decreased renal perfusion and failure of
glomerular filtration, vasoconstriction of renal arterioles, how formed
elements gain access to the renal parenchyma, and what the sequelae
are of such an invasion by primed leukocytes.
CHAPTER

14
14.2 Acute Renal Failure

Vasoactive Hormones

Ischemic or toxic insult

Tubular injury and


Hemodynamic changes dysfunction

Afferent arteriolar Mesangial Reduced tubular Backleak of Tubular obstruction


vasoconstriction contraction reabsorption of NaCl glomerular filtrate

Reduced GPF and P Reduced glomerular Increased delivery of Backleak of urea, Compromises patency
filtration surface area NaCl to distal nephron creatinine, of renal tubules and
available for filtration (macula densa) and and reduction in prevents the recovery
and a fall in Kf activation of TG feedback "effective GFR" of renal function

FIGURE 14-1
Pathophysiology of ischemic and toxic acute renal which directly reduce GFR. Tubular injury reduces
failure (ARF). The severe reduction in glomerular GFR by causing tubular obstruction and by allow-
filtration rate (GFR) associated with established ing backleak of glomerular filtrate. Abnormalities in
ischemic or toxic renal injury is due to the com- tubular reabsorption of solute may contribute to
bined effects of alterations in intrarenal hemody- intrarenal vasoconstriction by activating the tubu-
namics and tubular injury. The hemodynamic alter- loglomerular (TG) feedback system. GPF—glomeru-
ations associated with ARF include afferent arterio- lar plasmaflow; P—glomerular pressure; Kf—
lar constriction and mesangial contraction, both of glomerular ultrafiltration coefficient.

FIGURE 14-2
Ischemic or toxic injury Vasoactive hormones that may be responsible for the hemodynamic abnormalities in acute
to the kidney tubule necrosis (ATN). A persistent reduction in renal blood flow has been demonstrated
in both animal models of acute renal failure (ARF) and in humans with ATN. The mecha-
Increase in Deficiency of nisms responsible for the hemodynamic alterations in ARF involve an increase in the
vasoconstrictors vasodilators intrarenal activity of vasoconstrictors and a deficiency of important vasodilators. A num-
ber of vasoconstrictors have been implicated in the reduction in renal blood flow in ARF.
Angiotensin II The importance of individual vasoconstrictor hormones in ARF probably varies to some
Endothelin extent with the cause of the renal injury. A deficiency of vasodilators such as endothelium-
Thromboxane derived nitric oxide (EDNO) and/or prostaglandin I2 (PGI2) also contributes to the renal
Adenosine PGI2
EDNO hypoperfusion associated with ARF. This imbalance in intrarenal vasoactive hormones
Leukotrienes
Platelet-activating favoring vasoconstriction causes persistent intrarenal hypoxia, thereby exacerbating tubu-
factor lar injury and protracting the course of ARF.

Imbalance in vasoactive hormones


causing persistent intrarenal
vasoconstriction

Persistent medullary hypoxia


Pathophysiology of Ischemic Acute Renal Failure 14.3

FIGURE 14-3
Glomerular basement The mesangium regulates single-nephron glomerular filtration rate
membrane (SNGFR) by altering the glomerular ultrafiltration coefficient (Kf).
This schematic diagram demonstrates the anatomic relationship
Glomerular capillary
between glomerular capillary loops and the mesangium. The
endothelial cells mesangium is surrounded by capillary loops. Mesangial cells (M)
M are specialized pericytes with contractile elements that can respond
to vasoactive hormones. Contraction of mesangium can close and
Glomerular epithelial prevent perfusion of anatomically associated glomerular capillary
M cells loops. This decreases the surface area available for glomerular fil-
tration and reduces the glomerular ultrafiltration coefficient.
Mesangial cell contraction
Angiotensin II
Endothelin–1
Thromboxane Mesangial cell relaxation
Sympathetic nerves Prostacyclin
EDNO

FIGURE 14-4
A, The topography of juxtaglomerular apparatus (JGA), including
macula densa cells (MD), extraglomerular mesangial cells (EMC),
Afferent arteriole
and afferent arteriolar smooth muscle cells (SMC). Insets schemati-
Periportal cally illustrate, B, the structure of JGA; C, the flow of information
cell within the JGA; and D, the putative messengers of tubuloglomeru-
lar feedback responses. AA—afferent arteriole; PPC—peripolar cell;
Extraglomerular EA—efferent arteriole; GMC—glomerular mesangial cells.
mesangial cells (Modified from Goligorsky et al. [1]; with permission.)

Macula densa
cells Glomerus

AA
MD
AA AA
MD MD SMC+GC PPC
PPC G
PPC EMC GMC
G G
EMC GMC EMC GMC
Chloride
Adenosine EA
PGE2
EA EA Angiotensin
Nitric oxide
Osmolarity
B C D Unknown?
14.4 Acute Renal Failure

FIGURE 14-5
The normal tubuloglomerular (TG) feedback mechanism The tubuloglomerular (TG) feedback mech-
anism. A, Normal TG feedback. In the nor-
4. Afferent arteriolar and mesangial 3. Renin is released from specialized
contraction reduce SNGFR back toward cells of JGA and the intrarenal renin mal kidney, the TG feedback mechanism is
control levels. angiotensin system generates release a sensitive device for the regulation of the
of angiotensin II locally. single nephron glomerular filtration rate
(SNGFR). Step 1: An increase in SNGFR
increases the amount of sodium chloride
(NaCl) delivered to the juxtaglomerular
apparatus (JGA) of the nephron. Step 2:
The resultant change in the composition of
the filtrate is sensed by the macula densa
cells and initiates activation of the JGA.
Step 3: The JGA releases renin, which
results in the local and systemic generation
of angiotensin II. Step 4: Angiotensin II
2. The composition of filtrate induces vasocontriction of the glomerular
1. SNGFR increases passing the macula densa is arterioles and contraction of the mesangial
causing increase altered and stimulates the JGA. cells. These events return SNGFR back
in delivery of solute
to the distal nephron. toward basal levels. B, TG feedback in
ARF. Step 1: Ischemic or toxic injury to
renal tubules leads to impaired reabsorption
of NaCl by injured tubular segments proxi-
mal to the JGA. Step 2: The composition of
the filtrate passing the macula densa is
altered and activates the JGA. Step 3:
Angiotensin II is released locally. Step 4:
A
SNGFR is reduced below normal levels. It
is likely that vasoconstrictors other than
angiotensin II, as well as vasodilator hor-
Role of TG feedback in ARF mones (such as PGI2 and nitric oxide) are
also involved in modulating TG feedback.
4. Afferent arteriolar and mesangial 3. Local release of Abnormalities in these vasoactive hormones
contraction reduce SNGFR below angiotensin II in ARF may contribute to alterations in TG
normal levels. is stimulated.
feedback in ARF.

1. Renal epithelial cell injury 2. The composition of filtrate


reduces reabsorption passing the macula densa is
of NaCl by proximal tubules. altered and stimulates the JGA.

B
Pathophysiology of Ischemic Acute Renal Failure 14.5

FIGURE 14-6
Osswald's Hypothesis
Metabolic basis for the adenosine hypothesis. A, Osswald’s
Increased ATP hydrolysis (increased distal Na+ load) hypothesis on the role of adenosine in tubuloglomerular feedback.
B, Adenosine metabolism: production and disposal via the salvage
and degradation pathways. (A, Modified from Osswald et al. [2];
Increased generation of adenosine
with permission.)

Activation of JGA

Afferent arteriolar vasoconstriction

Nerve endings

[Na+] ATP Na+


Adenosine
Renin- ↓ Renin
Adenosine containing
cells secretion

ANG II
Vascular

[Cl ] smooth ↓ GFR
muscle
ANG I

Signal Transmission Mediator(s) Effects


A

Adenosine nucleotide metabolism

ATP ADP AMP Adenosine

A1 A2

Receptors
e

se

se
as

Transporter
P-

P-

id
AT

AD

ot
cle
nu
5'-

Phosphorylation
or
degradation

ATP ADP AMP Adenosine Inosine Hypo-


xanthine
Salvage Degradation
pathway pathway

Uric acid Xanthine


B
14.6 Acute Renal Failure

FIGURE 14-7
20 Elevated concentration of adenosine, inosine,
15 and hypoxanthine in the dog kidney and
Adenosine,
nmoles/mL

10 urine after renal artery occlusion. (Modified


5 from Miller et al. [3]; with permission.)
0
25
20
nmoles/mL
Inosine,

15
10
5
0
30
25
Hypoxanthine,

20
nmoles/mL

15
10
5
0
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18
Volume collected, mL

Post Ischemia

Glomerul I
SNGFR: 17.4±1.7 nL/min
PFR: 66.6±5.6 nL/min

Glomeruli II
SNGFR: 27.0±3.1 nL/min
B Anti-endothelin PFR: 128.7±14.4 nL/min

FIGURE 14-8
Endothelin (ET) is a potent renal vasoconstrictor. Endothelin (ET)
is a 21 amino acid peptide of which three isoforms—ET-1, ET-2
and ET-3—have been described, all of which have been shown to
be present in renal tissue. However, only the effects of ET-1 on the
kidney have been clearly elucidated. ET-1 is the most potent vaso-
constrictor known. Infusion of ET-1 into the kidney induces pro-
found and long lasting vasoconstriction of the renal circulation. A,
The appearance of the rat kidney during the infusion of ET-1 into
the inferior branch of the main renal artery. The lower pole of the
A kidney perfused by this vessel is profoundly vasoconstricted and
hypoperfused. B, Schematic illustration of function in separate
populations of glomeruli within the same kidney. The entire kidney
underwent 25 minutes of ischemia 48 hours before micropuncture.
Glomeruli I are nephrons not exposed to endothelin antibody;
Glomeruli II are nephrons that received infusion with antibody
through the inferior branch of the main renal artery. SNGFR—sin-
gle nephron glomerular filtration rate; PFR—glomerular renal plas-
ma flow rate. (From Kon et al. [4]; with permission.)
Pathophysiology of Ischemic Acute Renal Failure 14.7

FIGURE 14-9
Pre–proendothelin–1
Biosynthesis of mature endothelin-1 (ET-1). The mature ET-1
NH2 COOH peptide is produced by a series of biochemical steps. The precur-
sor of active ET is pre-pro ET, which is cleaved by dibasic pair-
53 74 92 203
specific endopeptidases and carboxypeptidases to yield a
39–amino acid intermediate termed big ET-1. Big ET-1, which
Lys–Arg Arg–Arg has little vasoconstrictor activity, is then converted to the mature
21–amino acid ET by a specific endopeptidase, the endothelin-
Dibasic pair–specific converting enzyme (ECE). ECE is localized to the plasma mem-
endopeptidase(s) brane of endothelial cells. The arrows indicate sites of cleavage
of pre-pro ET and big ET.

NH3 Big endothelin


COOH

Trp–Val Endothelin converting


enzyme (ECE)

Leu Ser Ser


Met Cys Ser Cys NH3
Mature endothelin
Asp
Lys
Glu
Cys Val Tyr Phe Cys His Leu Asp Ile Ile Trp COOH

FIGURE 14-10
Plasma ET Regulation of endothelin (ET) action; the
Mature ET role of the ET receptors. Pre-pro ET is pro-
duced and converted to big ET. Big ET is
converted to mature, active ET by endothe-
lin-converting enzyme (ECE) present on the
E ETB receptor endothelial cell membrane. Mature ET
EC
secreted onto the basolateral aspect of the
Endothelium NO PGI2 endothelial cell binds to two ET receptors
(ETA and ETB); both are present on vascu-
lar smooth muscle (VSM) cells. Interaction
of ET with predominantly expressed ETA
receptors on VSM cells induces vasocon-
striction. ETB receptors are predominantly
ECE located on the plasma membrane of
endothelial cells. Interaction of ET-1 with
these endothelial ETB receptors stimulates
Mature ET production of nitric oxide (NO) and prosta-
cyclin by endothelial cells. The production
Cyclic Cyclic of these two vasodilators serves to counter-
ETA receptor ETB receptor
GMP AMP balance the intense vasoconstrictor activity
Vascular
smooth of ET-1. PGI2—prostaglandin I2.
muscle
Vasoconstriction Vasodilation
14.8 Acute Renal Failure

FIGURE 14-11
Ischemia Vehicle
Endothelin-1 (ET-1) receptor blockade ameliorates severe ischemic
10 BQ123(0.1mg/kg • min, for 3h)
BQ123 acute renal failure (ARF) in rats. The effect of an ETA receptor
Number of rats

8 antagonist (BQ123) on the course of severe postischemic ARF was


6 examined in rats. BQ123 (light bars) or its vehicle (dark bars) was
administered 24 hours after the ischemic insult and the rats were
4
followed for 14 days. A, Survival. All rats that received the vehicle
2 were dead by the 3rd day after ischemic injury. In contrast, all rats
0 that received BQ123 post-ischemia survived for 4 days and 75%
A Basal 24h 1 2 3 4 5 6 14 recovered fully. B, Glomerular filtration rate (GFR). In both groups
control of rats GFR was extremely low (2% of basal levels) 24 hours after
ischemia. In BQ123-treated rats there was a gradual increase in
150
Ischemia GFR that reached control levels by the 14th day after ischemia.
120 C, Serum potassium. Serum potassium increased in both groups but
GFR, mL/h

BQ123(0.1mg/kg • min, for 3h)


90 reached significantly higher levels in vehicle-treated compared to the
60 BQ123-treated rats by the second day. The severe hyperkalemia
30 likely contributed to the subsequent death of the vehicle treated
0
rats. In BQ123-treated animals the potassium fell progressively after
B Basal 24h 1 2 3 4 5 6 14 the second day and reached normal levels by the fourth day after
control ischemia. (Adapted from Gellai et al. [5]; with permission.)

10 BQ123(0.1mg/kg • min, for 3h)


Plasma K+, mEq/L

Ischemia
8
6
4
2
0
Basal 24h 1 2 3 4 5 6 14
control
C Posttreatment days

FIGURE 14-12
Production of prostaglandins. Arachidonic acid is released from the
Lipid Membrane plasma membrane by phospholipase A2. The enzyme cycloxygenase
catalyses the conversion of arachidonate to two prostanoid interme-
diates (PGH2 and PGG2). These are converted by specific enzymes
into a number of different prostanoids as well as thromboxane
(TXA2). The predominant prostaglandin produced varies with the
Phospholipase A2
cell type. In endothelial cells prostacyclin (PGI2) (in the circle) is the
Arachidonic acid major metabolite of cycloxygenase activity. Prostacyclin, a potent
vasodilator, is involved in the regulation of vascular tone. TXA2 is
NSAID Cycloxygenase not produced in endothelial cells of normal kidneys but may be pro-
duced in increased amounts and contribute to the pathophysiology
PGG2 of some forms of acute renal failure (eg, cyclosporine A–induced
Prostaglandin nephrotoxicity). The production of all prostanoids and TXA2 is
intermediates blocked by nonsteroidal anti-inflammatory agents (NSAIDs), which
inhibit cycloxygenase activity.
Thromboxane
PGH2
TxA2

PGF2 PGI2 PGE2


Prostacyclin
Pathophysiology of Ischemic Acute Renal Failure 14.9

FIGURE 14-13
Aorta
Endothelin (ET) receptor blockade ameliorates acute cyclosporine-
induced nephrotoxicity. Cyclosporine A (CSA) was administered
Cyclosporine A intravenously to rats. Then, an ET receptor anatgonist was infused
in circulation directly into the right renal artery. Glomerular filtration rate (GFR)
Intra–arterial
infusion of ETA and renal plasma flow (RPF) were reduced by the CSA in the left
receptor antagonist
CSA
kidney. The ET receptor antagonist protected GFR and RPF from
the effects of CSA on the right side. Thus, ET contributes to the
intrarenal vasoconstriction and reduction in GFR associated with
acute CSA nephrotoxicity. (From Fogo et al. [6]; with permission.)

Right renal Left renal


artery artery

GFR and RPF: GFR and RPF:


near normal Reduced 20-25%
below normal
Right kidney Left kidney

FIGURE 14-14
Normal basal state
Prostacyclin is important in maintaining
Circulating levels of vasoconstrictors: Low renal blood flow (RBF) and glomerular fil-
tration rate (GFR) in “prerenal” states.
Afferent arteriolar tone
normal A, When intravascular volume is normal,
prostacyclin production in the endothelial
Intrarenal levels of prostacyclin: Low cells of the kidney is low and prostacyclin
Intraglomerular  P plays little or no role in control of vascular
normal
tone. B, The reduction in absolute or
“effective” arterial blood volume associated
with all prerenal states leads to an increase
A GFR normal in the circulating levels of a number of of
vasoconstrictors, including angiotensin II,
Intravascular volume depletion
catecholamines, and vasopressin. The
Circulating levels of vasoconstrictors: High increase in vasoconstrictors stimulates
phospholipase A2 and prostacyclin produc-
Afferent arteriolar tone
tion in renal endothelial cells. This increase
normal or mildly reduced
in prostacyclin production partially coun-
Intrarenal levels of prostacyclin: High teracts the effects of the circulating vaso-
constrictors and plays a critical role in
Intraglomerular  P
normal or mildly reduced
maintaining normal or nearly normal RBF
and GFR in prerenal states. C, The effect of
cycloxygenase inhibition with nonsteroidal
B GFR anti-inflammatory drugs (NSAIDs) in pre-
normal or mildly reduced renal states. Inhibition of prostacyclin
production in the presence of intravascular
Intravascular volume depletion volume depletion results in unopposed
and NSAID administration action of prevailing vasoconstrictors and
Circulating levels of vasoconstrictors: High results in severe intrarenal vascasoconstric-
tion. NSAIDs can precipitate severe acute
Afferent arteriolar tone
renal failure in these situations.
severely increased

Intrarenal levels of prostacyclin: Low


Intraglomerular  P
severely reduced

GFR
C severely reduced
14.10 Acute Renal Failure

A. VASODILATORS USED IN EXPERIMENTAL ACUTE RENAL FAILURE (ARF)

Time Given in
Vasodilator ARF Disorder Relation to Induction Observed Effect
Propranolol Ischemic Before, during, after ↓Scr, BUN if given before,
during; no effect if given after
Phenoxybenzamine Toxic Before, during, after Prevented fall in RBF
Clonidine Ischemic After ↓Scr, BUN
Bradykinin Ischemic Before, during ↑RBF, GFR
Acetylcholine Ischemic Before, after ↑RBF; no change in GFR
Prostaglandin E1 Ischemic After ↑RBF; no change in GFR
Prostaglandin E2 Ischemic, toxic Before, during ↑GFR
Prostaglandin I2 Ischemic Before, during, after ↑GFR
Saralasin Toxic, ischemic Before ↑RBF; no change in Scr, BUN
Captopril Toxic, ischemic Before ↑RBF; no change in Scr, BUN
Verapamil Ischemic, toxic Before, during, after ↑RBF, GFR in most studies
Nifedipine Ischemic Before ↑GFR
Nitrendipine Toxic Before, during ↑GFR
Diliazem Toxic Before, during, after ↑GFR; ↓recovery time
Chlorpromazine Toxic Before ↑GFR; ↓recovery time
Atrial natriuretic Ischemic, toxic After ↑RBF, GFR
peptide

BUN—blood urea nitrogen; GFR—glomerular filtration rate; RBF—renal blood flow; Scr–serum creatinine.

B. VASODILATORS USED TO ALTER COURSE


OF CLINICAL ACUTE RENAL FAILURE (ARF)

Vasodilator ARF Disorder Observed Effect Remarks


Dopamine Ischemic, toxic Improved V, Scr if used early Combined with furosemide
Phenoxybenzamine Ischemic, toxic No change in V, RBF
Phentolamine Ischemic, toxic No change in V, RBF
Prostaglandin A1 Ischemic No change in V, Scr Used with dopamine
Prostaglandin E1 Ischemic ↑RBF, no change v, Ccr Used with NE
Dihydralazine Ischemic, toxic ↑RBF, no change V, Scr
Verapamil Ischemic ↑Ccr or no effect
Diltiazem Transplant, toxic ↑Ccr or no effect Prophylactic use
Nifedipine Radiocontrast No effect
Atrial natriuretic Ischemic ↑Ccr
peptide

Ccr—creatinine clearance; NE—norepinephrine; RBF—renal blood flow; Scr—serum creatinine; V—urine flow rate.

FIGURE 14-15
Vasodilators used in acute renal failure (ARF). A, Vasodilators used in experimental
acute ARF. B, Vasodilators used to alter the course of clinical ARF. (From Conger [7];
with permission.)
Pathophysiology of Ischemic Acute Renal Failure 14.11

NH2 + NH2 NH2 + NOH NH2 + O Modular structure of nitric oxide synthases
H BH4 ARG CaM FMN FAD NADPH
NADPH NADP+ 1/
2 NADPH /2 NADP
1 +
Target domain Oxygenase domain Reductase domain
O2 H 2O O2 H 2O
NH NH NH Dimerization site(s) 13–14 18–20
+ • NO nNOS
2–3 4–5 6 7–9 10–12 1516–17 21–23 24–29
BH4 BH4 nitric oxide 11–12 16–18
eNOS
+
NH3 COO– +
NH3 COO– +
NH3 COO– 1 2–3 4 5–7 8–10 1314–15 19–21 22–26
M 12–13
G
A L-arginine N -hydroxy-L-arginine L-citrulline iNOS
2–3 4–5 6 7–9 10–11 13 14–18 19–21 22–26
2–3 4–8 9–12 13–16
FIGURE 14-16 Mammalian P450 Reductases
Chemical reactions leading to the generation of nitric oxide (NO), Bacterial Flavodoxins
Plant Ferredoxin NADPH Reductases
A, and enzymes that catalize them, B. (Modified from Gross [8]; B. mega P450
with permission.) DHF Reductases
B Mammalian Syntrophins (GLGF Motif)

L-arginine
M
DNA damage Cell death
NOS L-citrulline NO concentration Activation of Apoptosis
mM apoptotic signal
Nitric oxide Thiols Induction of stress proteins
GTP
Heme- & iron- Inactivation of enzymes
Smooth muscle GC Vasodilatation µM containing proteins
ROIs Antioxidant
cGMP Guanylate cyclase cGMP (cellular signal)
nM
Inhibition of B Time Consequences
Target cell
Immune cells iron-containing
death
enzymes
FIGURE 14-17
CNS and PNS Neurotransmission
Hemoglobin Major organ, A, and cellular, B, targets of nitric oxide (NO).
A, Synthesis and function of NO. B, Intracellular targets for NO
and pathophysiological consequences of its action. C, Endothelium-
NO3– + NO2– dependent vasodilators, such as acetylcholine and the calcium
ionophore A23187, act by stimulating eNOS activity thereby
cGMP increasing endothelium-derived nitric oxide (EDNO) production.
In contrast, other vasodilators act independently of the endotheli-
A Urine excretion um. Some endothelium-independent vasodilators such as nitroprus-
side and nitroglycerin induce vasodilation by directly releasing nitric
oxide in vascular smooth muscle cells. NO released by these agents,
Leukocyte like EDNO, induces vasodilation by stimulating the production of
– migration cyclic guanosine monophosphate (cGMP) in vascular smooth mus-
Endothelium-dependent
vasodilators cle (VSM) cells. Atrial natriuretic peptide (ANP) is also an endothe-
Platelet
– lium-independent vasodilator but acts differently from NO. ANP
+ aggregation
NO• directly stimulates an isoform of guanylyl cyclase (GC) distinct from
+
Shear stress soluble GC (called particulate GC) in VSM. CNS—central nervous
L-Arginine + NOS system; GTP—guanosine triphosphate; NOS—nitric oxide synthase;
PGC—particulate guanylyl cyclase; PNS—peripheral nervous sys-
NO• tem; ROI—reduced oxygen intermediates; SGC—soluble guanylyl
cyclase. (A, From Reyes et al. [9], with permission; B, from Kim
Nitroglycerin ANP et al. [10], with permission.)
+ +
sGC + pGC

GTP cGMP

Relaxation
C
14.12 Acute Renal Failure

FIGURE 14-18
Ischemia (I) Impaired production of endothelium-dependent nitric oxide (EDNO) contributes to the
alone vasoconstriction associated with established acute renal failure (ARF). Ischemia-reperfu-
sion injury in the isolated erythrocyte-perfused kidney induced persistant intarenal vaso-
I + ANP constriction. The endothelium-independent vasodilators (atrial natriuretic peptide [ANP]
and nitroprusside) administered during the reflow period caused vasodilation and restored
I+ the elevated intrarenal vascular resistance (RVR) to normal. In marked contrast, two
nitroprusside endothelium-dependent vasodilators (acetylcholine and A23187) had no effect on renal
I+ vascular resistance after ischemia-reflow. These data suggest that EDNO production is
Acetylcholine impaired following ischemic injury and that this loss of EDNO activity contributes to the
vasoconstriction associated with ARF. (Adapted from Lieberthal [11]; with permission.)
I + A23187

0 20 40 60 80
Increase in RVR above control, %

60 150
O2 BUN
50 *
Hypoxia 100

mg/dL
*
40 *

30 P<.001
50
Hypoxia + L-NAME
20 P<.001
0
Percent LDH release

Control 3.0
10
0 Cr
2.5
1.5
60 O2 Hypoxia + L-Arg
mg/dL

P<.05
50 *
1.0 *
P<.01 Hypoxia *
40
P<.001
30 0.5
P<.001
20
Control 0
10 Ischemia
Control

0
S

SCR
AS
Vehicle

0 10 20 30 40 50
A Time, min B

FIGURE 14-19
P<.001 Deleterious effects of nitric oxide (NO) on the viability of renal
50 tubular epithelia. A, Hypoxia and reoxygenation lead to injury of
tubular cells (filled circles); inhibition of NO production improves
40 the viability of tubular cells subjected to hypoxia and reoxygena-
tion (triangles in upper graph), whereas addition of L-arginine
LDH release, %

enhances the injury (triangles in lower graph). B, Amelioration of


30 ischemic injury in vivo with antisense oligonucleotides to the
NS iNOS: blood urea nitrogen (BUN), and creatinine (CR) in rats sub-
20 jected to 45 minutes of renal ischemia after pretreatment with anti-
sense phosphorothioate oligonucleotides (AS) directed to iNOS or
with sense (S) and scrambled (SCR) constructs. C, Resistance of
10
proximal tubule cells isolated from iNOS knockout mice to hypox-
ia-induced injury. LDH—lactic dehydrogenase. (A, From Yu et al.
0 [12], with permission; B, from Noiri et al. [13], with permission;
Normoxia Hypoxia Normoxia Hypoxia C, from Ling et al. [14], with permission.)
C Wild type mice iNOS knockout mice
Pathophysiology of Ischemic Acute Renal Failure 14.13

Iothalamate Radiocontrast

100 100
Chronic renal
Normal kidneys
Cortex

50 50 insufficiency
Percent of baseline

0 0
Iothalamate Compensatory increase in Increased Reduced or absent
200
PGI2 and EDNO release endothelin increase in PGI2 or EDNO
150
Medulla

100 100

50 50
Iothalamate Mild vasoconstriction Severe vasoconstriction
0 0
0 20 40 60 0 20 40 60
Minutes Minutes
No pretreatment Pretreatment with
No loss of GFR Acute renal failure
A (n = 6) L-NAME (n = 6) B

FIGURE 14-20
Proposed role of nitric oxide (NO) in radiocontrast-induced acute the normal kidney. The vasodilators counteract the effects of the
renal failure (ARF). A, Administration of iothalamate, a radiocon- vasoconstrictors so that intrarenal vasoconstriction in response to
trast dye, to rats increases medullary blood flow. Inhibitors of radiocontrast is usually modest and is associated with little or no
either prostaglandin production (such as the NSAID, loss of renal function. However, in situations when there is pre-
indomethacin) or inhibitors of NO synthesis (such as L-NAME) existing chronic renal insufficiency (CRF) the vasodilator response
abolish the compensatory increase in medullary blood flow that to radiocontrast is impaired, whereas production of endothelin and
occurs in response to radiocontrast administration. Thus, the stim- other vasoconstrictors is not affected or even increased. As a result,
ulation of prostaglandin and NO production after radiocontrast radiocontrast administration causes profound intrarenal vasocon-
administration is important in maintaining medullary perfusion striction and can cause ARF in patients with CRF. This hypothesis
and oxygenation after administration of contrast agents. B, would explain the predisposition of patients with chronic renal
Radiocontrast stimulates the production of vasodilators (such as dysfunction, and especially diabetic nephropathy, to contrast-
prostaglandin [PGI2] and endothelium-dependent nitric oxide induced ARF. (A, Adapted from Agmon and Brezis [15], with per-
[EDNO]) as well as endothelin and other vasoconstrictors within mission; B, from Agmon et al. [16], with permission.)

FIGURE 14-21
Cellular calcium metabolism and potential targets of the elevated
cytosolic calcium. A, Pathways of calcium mobilization. B, Patho-
physiologic mechanisms ignited by the elevation of cytosolic calci-
um concentration. (A, Adapted from Goligorsky [17], with permis-
sion; B, from Edelstein and Schrier [18], with permission.)
14.14 Acute Renal Failure

Pre NE Post NE
* 60 NS Verapamil before NE
400 100 P<.001
40
* * P<.05
Estimated [Ca2+]i , nM

80

Pl stained nuclei, %
*
20
* * *

CIn, mL/min
* 60
300 * 0
40 60 Verapamil after NE
NS
200 * Significant 20
vs. time 0 40
P<.02
150 0 P<.001
Hypoxia 20

0 10 20 30 0
A Time, min B Control 1h 24 h

FIGURE 14-22
Pathophysiologic sequelae of the elevated cytosolic calcium (C2+). verapamil before injection of norepinephrine (cross-hatched bars) sig-
A, The increase in cytosolic calcium concentration in hypoxic rat nificantly attenuated the drop in inulin clearance induced by norepi-
proximal tubules precedes the tubular damage as assessed by propidi- nephrine alone (open bars). (A, Adapted from Kribben et al. [19], with
um iodide (PI) staining. B, Administration of calcium channel inhibitor permission; B, adapted from Burke et al. [20], with permission.)

FIGURE 14-23
Dynamics of heat shock
proteins (HSP) in stressed
cells. Mechanisms of acti-
vation and feedback con-
trol of the inducible heat
shock gene. In the nor-
mal unstressed cell, heat
shock factor (HSF) is
rendered inactive by
association with the con-
stitutively expressed
HSP70. After hypoxia or
ATP depletion, partially
denatured proteins (DP)
become preferentially
associated with HSC73,
releasing HSF and allow-
ing trimerization and
binding to the heat shock
element (HSE) to initiate
the transcription of the
heat shock gene. After
translation, excess
inducible HSP (HSP72)
interacts with the trimer-
ized HSF to convert it
back to its monomeric
state and release it from
the HSE, thus turning off
the response. (Adapted
from Kashgarian [21];
with permission.)
Pathophysiology of Ischemic Acute Renal Failure 14.15

Free Radical Pathways in the Mitochondrion


Catalase/GPx complex?
Hydrogen H2O 2
peroxide
Hydrogen
H 2O 2 Outer Hydroperoxyl peroxide
membrane radical
Inner HO2
membrane

O2 HO2
Superoxide Hepatocyte
(and other cells) Plasma EC–SOD
anion
(From glycolysis/ Proteinase?
TCA cycle) Golgi
– e–
Mn-SOD 2O2 complex
O2
(tetramer) Tissue EC–SOD
+ Endoplasmic
Matrix 2H+ reticulum Secretory vesicle
Heparin +
Mitochondrion sulfate 2H+
Chromosome proteoglycans
(chrom) 4
Manganese H 2O 2
superoxide
dismatase
(Mn-SOD) mRNA Extracellular GPx +
superoxide subunit O2
dismutase
(EC-SOD) Se GPx
chrom 6 mRNA (tetramer)
Catalase H2O+O2
mRNA Glutathione
chrom 3 +GSSG (dimer)
Glutathione
peroxidase
chrom 21 (GPx) mRNA
chrom 11
Cu,Zn–SOD +2GSH Glutathione
(dimer) (monomer)
Catalase
subunit
Peroxisome Plasma
+O2 membrane
Copper–zinc 2O2– +2H+ damaged
superoxide (enlarged below)
dismutase
(Cu,Zn–SOD) mRNA
Perxisome reactions Lipid peroxidation of plasma membrane
Oxidative enzyme Phospholipid 2GSH's + LOOH LOH+
(eg, urate oxidase) GSSG+ Free
hydroperoxide
RH2 glutathione radical
LO

+ Catalase peroxidase
OH

O2 (tetramer) (PHGPx) R
Lipid
peroxide
LH
LO

Inside O RH
H

Heme cell L Lipid


Lipid
LH LOO radical
Hydrogen 2H2O+O2 LH
peroxide LH
LOOH O
LH

L LOO Outside
LOOH H cell
LH
e– Lipid
Vitamin E (a-Tocopherol–)
inhibits lipid peroxidation chain collpases
chain reaction (now hydrophilic)

FIGURE 14-24
Cellular sources of reactive oxygen species (ROS) defense systems from free radicals. Superoxide and hydro-
gen peroxide are produced during normal cellular metabolism. ROS are constantly being produced by the
normal cell during a number of physiologic reactions. Mitochondrial respiration is an important source of
superoxide production under normal conditions and can be increased during ischemia-reflow or gentamycin-
induced renal injury. A number of enzymes generate superoxide and hydrogen peroxide during their catalytic
cycling. These include cycloxygenases and lipoxygenes that catalyze prostanoid and leukotriene synthesis.
Some cells (such as leukocytes, endothelial cells, and vascular smooth muscle cells) have NADH/ or NADPH
oxidase enzymes in the plasma membrane that are capable of generating superoxide. Xanthine oxidase, which
converts hypoxathine to xanthine, has been implicated as an important source of ROS after ischemia-reperfu-
sion injury. Cytochrome p450, which is bound to the membrane of the endoplasmic reticulum, can be
increased by the presence of high concentrations of metabolites that are oxidized by this cytochrome or by
injurious events that uncouple the activity of the p450. Finally, the oxidation of small molecules including free
heme, thiols, hydroquinines, catecholamines, flavins, and tetrahydropterins, also contribute to intracellular
superoxide production. (Adapted from [22]; with permission.)
14.16 Acute Renal Failure

FIGURE 14-25
EVIDENCE SUGGESTING A ROLE FOR Evidence suggesting a role for reactive oxygen metabolites in acute
REACTIVE OXYGEN METABOLITES IN renal failure. The increased ROS production results from two
ISCHEMIC ACUTE RENAL FAILURE major sources: the conversion of hypoxanthine to xanthine by xan-
thine dehydrogenase and the oxidation of NADH by NADH oxi-
dase(s). During the period of ischemia, oxygen deprivation results
Enhanced generation of reactive oxygen metabolites and xanthine oxidase and in the massive dephosphorylation of adenine nucleotides to hypox-
increased conversion of xanthine dehydrogenase to oxidase occur in in vitro and in anthine. Normally, hypoxanthine is metabolized by xanthine dehy-
vivo models of injury. drogenase which uses NAD+ rather than oxygen as the acceptor of
Lipid peroxidation occurs in in vitro and in vivo models of injury, and this can be pre- electrons and does not generate free radicals. However, during
vented by scavengers of reactive oxygen metabolites, xanthine oxidase inhibitors, or ischemia, xanthine dehydrogenase is converted to xanthine oxi-
iron chelators. dase. When oxygen becomes available during reperfusion, the
Glutathione redox ratio, a parameter of “oxidant stress” decreases during ischemia and metabolism of hypoxanthine by xanthine oxidase generates super-
markedly increases on reperfusion. oxide. Conversion of NAD+ to its reduced form, NADH, and the
Scavengers of reative oxygen metabolites, antioxidants, xanthine oxidase inhibitors, accumulation of NADH occurs during ischemia. During the reper-
and iron chelators protect against injury. fusion period, the conversion of NADH back to NAD+ by NADH
A diet deficient in selenium and vitamin E increases susceptibility to injury. oxidase also results in a burst of superoxide production. (From
Inhibition of catalase exacerbates injury, and transgenic mice with increased superoxide Ueda et al. [23]; with permission.)
dismutase activity are less susceptible to injury.

250 3.0
FIGURE 14-26
24 Effect of different scavengers of reactive
16
2.5 oxygen metabolites and iron chelators on,
200
Plasma urea nitrogen, mg/dL

A, blood urea nitrogen (BUN) and, B, crea-


*P < 0.001 2.0 *P < 0.001 tinine in gentamicin-induced acute renal
Creatinine, mg/dL

150 failure. The numbers shown above the error


1.5 bars indicate the number of animals in each
100 group. Benz—sodium benzoate; Cont—con-
1.0 trol group; DFO—deferoxamine; DHB—
16* 8* 6* 5* 4* 2,3 dihydroxybenzoic acid; DMSO—
50 8*
8* dimethyl sulfoxide; DMTU—dimethylth-
6* 13* 4* 0.5
26 18 iourea; Gent—gentamicin group. (From
0 0.0 Ueda et al. [23]; with permission.)
O

HB

HB
FO

FO
nz

nz
U

U
t

t
t

t
Con

Con
Gen

Gen
MT

MT
MS

MS
+Be

+Be
+D

+D
+D

+D
+D

+D
+D

+D

A B

FIGURE 14-27
Superoxide Production of the hydroxyl radical: the Haber-Weiss reaction. Superoxide is converted to
O2– +Fe3+ hydrogen peroxide by superoxide dismutase. Superoxide and hydrogen peroxide per se
are not highly reactive and cytotoxic. However, hydrogen peroxide can be converted to
Iron stores the highly reactive and injurious hydroxyl radical by an iron-catalyzed reaction that
(Ferritin) requires the presence of free reduced iron. The availability of free “catalytic iron” is a
Release of Hydrogen critical determinant of hydroxyl radical production. In addition to providing a source of
free iron Peroxide hydroxyl radical, superoxide potentiates hydroxyl radical production in two ways: by
(H2O2) releasing free iron from iron stores such as ferritin and by reducing ferric iron and recy-
Fe2+ cling the available free iron back to the ferrous form. The heme moiety of hemoglobin,
myoglobin, or cytochrome present in normal cells can be oxidized to metheme (Fe3+).
Fe3+ The further oxidation of metheme results in the production of an oxyferryl moiety
OH Hydroxyl (Fe4+=O), which is a long-lived, strong oxidant which likely plays a role in the cellular
Radical injury associated with hemoglobinuria and myoglobinuria.
(OH–) Activated leukocytes produce superoxide and hydrogen peroxide via the activity of a
membrane-bound enzyme NADPH oxidase. This superoxide and hydrogen peroxide can
be converted to hydroxyl radical via the Haber-Weiss reaction. Also, the enzyme myeloper-
oxidase, which is specific to leukocytes, converts hydrogen peroxide to another highly
reactive and injurious oxidant, hypochlorous acid.
Pathophysiology of Ischemic Acute Renal Failure 14.17

FIGURE 14-28
:O–O• + •N–O :O–O–N–O 22 kcal/mol Initiation LH + OH• H2O + L• Cell injury: point of convergence between
...Large Gibbs energy
the reduced oxygen intermediates–generat-
ONOO– Propagation L• + O2 LOO• ing and reduced nitrogen intermediates–
:O2•– 6.7 x 109 M–1•s–1 [NO] ...Faster than SOD generating pathways, A, and mechanisms
O 2 + H 2O 2 of lipid peroxidation, B.
1 x 109 M–1•s–1 [SOD] LOO• + LH LOOH + L•
H
O O O O Termination L• + L• L–L
N O N O N O•
OH
...Peroxynitrous LOO• + NO• LOONO
A OH• acid in trans B

ONOO–
Cortex Medulla
X X: SOD, Cu2+, Fe3+

XO

NO2•
Tyr
116 KD 116 KD
NO2

OH R Nitrotyrosine
A

66 KD 66 KD

C CI LN C CI LN

Free
R R' radical
Unsaturated fatty acid R R'
• O2
O O
Free OO•
Control Control Ischemia L-Nil + Ischemia R • R' radical
R R'
B O2 Lipid based
peroxyradical (LOO•)
OO• O O
FIGURE 14-29 OH
O
Detection of peroxynitrite production and lipid peroxidation in R R'
ischemic acute renal failure. A, Formation of nitrotyrosine as an HNE
indicator of ONOO- production. Interactions between reactive
oxygen species such as the hydroxyl radical results in injury to Ab
the ribose-phosphate backbone of DNA. This results in single- HNE OH
O OH
O
and double-strand breaks. ROS can also cause modification and
deletion of individual bases within the DNA molecule. Interaction
between reactive oxygen and nitrogen species results in injury to X X
the ribose-phosphate backbone of DNA, nuclear DNA fragmenta- Protein
tion (single- and double-strand breaks) and activation of poly- (X: Cys, His, Lys) Formation of stable
D hemiacetal adducts
(ADP)-ribose synthase. B, Immunohistochemical staining of kid-
neys with antibodies to nitrotyrosine. C, Western blot analysis of
nitrotyrosine. D, Reactions describing lipid peroxidation and for- process is called lipid peroxidation. In addition to impairing the
mation of hemiacetal products. The interaction of oxygen radi- structural and functional integrity of cell membranes, lipid perox-
cals with lipid bilayers leads to the removal of hydrogen atoms idation can lead to a self-perpetuating chain reaction in which
from the unsaturated fatty acids bound to phospholipid. This additional ROS are generated.
(Continued on next page)
14.18 Acute Renal Failure

Cortex Medulla

Control Control Ischemia L-Nil + Ischemia


E
FIGURE 14-29 (Continued)
E, Immunohistochemical staining of kidneys with antibodies to C CI LN C CI LN
HNE–modified proteins. F, Western blot analysis of HNE expres-
sion. C—control; CI—central ischemia; LN—ischemia with L-Nil
pretreatment (Courtesy of E. Noiri, MD.) F

Leukocytes in Acute Renal Failure


FIGURE 14-30
Leukocyte adhesion molecules Role of adhesion molecules in mediating
Inactive leukocyte
β2 integrins (LFA1 or Mac1) leukocyte attachment to endothelium.
Selections
A, The normal inflammatory response is
Endothelial adhesion molecules mediated by the release of cytokines that
Activated leukocyte ICAM induce leukocyte chemotaxis and activation.
Ligand for leukocyte selections The initial interaction of leukocytes with
endothelium is mediated by the selectins and
their ligands both of which are present on
leukocytes and endothelial cells,
(Continued on next page)

Firm adhesion of
Selection–mediated
leukocytes
rolling of leukocytes
(integrin–mediated)
Diapedesis

Release of
oxidants
Tissue injury proteases
A elastases
Pathophysiology of Ischemic Acute Renal Failure 14.19

FIGURE 14-29 (Continued)


B. LEUKOCYTE ADHESION MOLECULES B. Selectin-mediated leukocyte-endothelial interaction results in
AND THEIR LIGANDS POTENTIALLY the rolling of leukocytes along the endothelium and facilitates the
IMPORTANT IN ACUTE RENAL FAILURE firm adhesion and immobilization of leukocytes. Immobilization of
leukocytes to endothelium is mediated by the 2-integrin adhesion
molecules on leukocytes and their ICAM ligands on endothelial
Major Families Cell Distribution cells. Immobilization of leukocytes is necessary for diapedesis of
leukocytes between endothelial cells into parenchymal tissue.
Selectins Leukocytes release proteases, elastases, and reactive oxygen radi-
L-selectin Leukocytes cals that induce tissue injury. Activated leukocytes also elaborate
P-selectin Endothelial cells cytokines such as interleukin 1 and tumor necrosis factor which
E-selectin Endothelial cells attract additional leukocytes to the site, causing further injury.
Carbohydrate ligands for selectins
Sulphated polysacharides Endothelium
Oligosaccharides Leukocytes
Integrins
CD11a/CD18 Leukocytes
CD11b/CD18 Leukocytes
Immunoglobulin G–like ligands
for integrins
Intracellular adhesion molecules (ICAM) Endothelial cells

FIGURE 14-31
125
Anti-ICAM Anti-ICAM Neutralizing anti–ICAM antibody amelio-
antibody 2 antibody rates the course of ischemic renal failure
100 Vehicle Vehicle
with blood urea nitrogen, A, and plasma
Blood urea nitrogen

creatinine, B. Rats subjected to 30 minutes


Plasma creatinine

1.5
75 of bilateral renal ischemia or a sham-opera-
tion were divided into three groups that
1
50 received either anti-ICAM antibody or its
vehicle. Plasma creatinine levels are shown
25 0.5 at 24, 48, and 72 hours. ICAM antibody
ameliorates the severity of renal failure at
0 0 all three time points. (Adapted from Kelly
0 24 48 72 0 24 48 72 96 et al. [24]; with permission.)
A Time following ischemia-reperfusion, d B Time following ischemia-reperfusion, d

1250 FIGURE 14-32


Vehicle Neutralizing anti-ICAM-1 antibody reduces myeloperoxidase activity
Anti-ICAM in rat kidneys exposed to 30 minutes of ischemia. Myeloperoxidase
1000
antibody is an enzyme specific to leukocytes. Anti-ICAM antibody reduced
Myeloperoxidase activity

myeloperoxidase activity (and by inference the number of leuko-


750 cytes) in renal tissue after 30 minutes of ischemia. (Adapted from
Kelly et al. [24]; with permission.)
500

250

0
0 4 24 48 72
Time after reperfusion, hrs
14.20 Acute Renal Failure

Mechanisms of Cell Death: Necrosis and Apoptosis


FIGURE 14-33
Apoptosis and necrosis: two distinct morphologic forms of cell death. A, Necrosis. Cells
undergoing necrosis become swollen and enlarged. The mitochondria become markedly
abnormal. The main morphoplogic features of mitochondrial injury include swelling and
flattening of the folds of the inner mitochondrial membrane (the christae). The cell plasma
membrane loses its integrity and allows the escape of cytosolic contents including lyzoso-
mal proteases that cause injury and inflammation of the surrounding tissues. B, Apoptosis.
In contrast to necrosis, apoptosis is associated with a progressive decrease in cell size and
maintenance of a functionally and structurally intact plasma membrane. The decrease in
cell size is due to both a loss of cytosolic volume and a decrease in the size of the nucleus.
The most characteristic and specific morphologic feature of apoptosis is condensation of
nuclear chromatin. Initially the chromatin condenses against the nuclear membrane. Then
the nuclear membrane disappears, and the condensed chromatin fragments into many
pieces. The plasma membrane undergoes a process of “budding,” which progresses to
fragmentation of the cell itself. Multiple plasma membrane–bound fragments of condensed
DNA called apoptotic bodies are formed as a result of cell fragmentation. The apoptotic
cells and apoptotic bodies are rapidly phagocytosed by neighboring epithelial cells as well
as professional phagocytes such as macrophages. The rapid phagocytosis of apoptotic bod-
ies with intact plasma membranes ensures that apoptosis does not cause any surrounding
inflammatory reaction.

A B

FIGURE 14-34
Hypothetical schema of cellular events trig-
Induction phase

gering apoptotic cell death. (From Kroemer


et al. [25]; with permission.)
[Ca2+]i ? ? ?
Signal transduction pathways

Mitochondrion Regulation by
Hcl-2 and
Mitochondrial permeability transition its relatives

? Activation of
Positive feedback loop ICE/ced-3-like
Effector phase

proteases ?
Consequences of permeability transition:
Disruption of ∆ψm and mitochondrial biogenesis
Breakdown of energy metabolism
Uncoupling of respiratory chain
Calcium release frommitochondrial matrix
Hyperproduction of superoxide anion
Depletion of glutathione
?

ROS Increase in ATP NAD/NADH Tyrosin kinases


effects [Ca2+]i depletion depletion G-proteins ?
Degradation phase

Cytoplasmic effects Nucleus


Disruption of anabolic reactions Activation of endonucleases
Dilatation of ER Activation of repair enzymes
Activation of proteases (ATP depletion)
Disruption of intracellular calcium Activation of poly(ADP) ribosly
compartimentalization transferase (NAD depletion)
Disorganization of cytoskeleton Chromatinolysis, nucleolysis
Pathophysiology of Ischemic Acute Renal Failure 14.21

FIGURE 14-35
Phagocytosis of an apoptotic body by a renal tubular epithelial cell.
Epithelial cells dying by apoptosis are not only phagocytosed by
macrophages and leukocytes but by neighbouring epithelial cells as
well. This electron micrograph shows a normal-looking epithelial cell
containing an apoptotic body within a lyzosome. The nucleus of an
epithelial cell that has ingested the apoptotic body is normal (white
arrow). The wall of the lyzosome containing the apoptotic body (black
arrow) is clearly visible. The apoptotic body consists of condensed
chromatin surrounded by plasma membrane (black arrowheads).

FIGURE 14-36
DNA fragmentation in apoptosis vs necrosis. DNA is made up of nucleosomal units. Each
Nucleosome nucleosome of DNA is about 200 base pairs in size and is surrounded by histones. Between
~200 bp nucleosomes are small stretches of DNA that are not surrounded by histones and are called
linker regions. During apoptosis, early activation of endonuclease(s) causes double-strand
Internucleosome breaks in DNA between nucleosomes. No fragmentation occurs in nucleosomes because the
"Linker" regions
DNA is “protected” by the histones. Because of the size of nucleosomes, the DNA is frag-
mented during apoptosis into multiples of 200 base pair pieces (eg, 200, 400, 600, 800).
When the DNA of apoptotic cells is electrophoresed, a characteristic ladder pattern is found.
DNA fragmentation In contrast, necrosis is associated with the early release of lyzosomal proteases, which
cause proteolysis of nuclear histones, leaving “naked” stretches of DNA not protected by
histones. Activation of endonucleases during necrosis therefore cause DNA cleavage at
multiple sites into double- and single-stranded DNA fragments of varying size.
Apoptosis Necrosis Electrophoresis of DNA from necrotic cells results in a smear pattern.

Loss
of
histones

800 bp 600 bp 400 bp 200 bp

DNA electrophoresis

Apoptic Necrotic
"ladder" "smear"
pattern pattern
14.22 Acute Renal Failure

FIGURE 14-37
POTENTIAL CAUSES OF APOPTOSIS Potential causes of apoptosis in acute renal failure (ARF). The
IN ACUTE RENAL FAILURE same cytotoxic stimuli that induce necrosis cause apoptosis. The
mechanism of cell death induced by a specific injury depends in
large part on the severity of the injury. Because most cells require
Loss of survival factors constant external signals, called survival signals, to remain viable,
Deficiency of renal growth factors (eg, IGF-1, EGF, HGF) the loss of these survival signals can trigger apoptosis. In ARF, a
Loss of cell-cell and cell-matrix interactions
deficiency of growth factors and loss of cell-substrate adhesion are
potential causes of apoptosis. The death pathways induced by
Receptor-mediated activators of apoptosis
engagement of tumour necrosis factor (TNF) with the TNF recep-
Tumor necrosis factor
tor or Fas with its receptor (Fas ligand) are well known causes of
Fas/Fas ligand
apoptosis in immune cells. TNF and Fas can also induce apoptosis
Cytotoxic events
in epithelial cells and may contribute to cell death in ARF.
Ischemia; hypoxia; anoxia
Oxidant injury
Nitric oxide
Cisplati

FIGURE 14-38
Apoptotic Trigger Apoptosis is mediated by a highly coordinated and genetically pro-
grammed pathway. The response to an apoptotic stimulus can be
divided into a commitment and execution phases. During the com-
mitment phase the balance between a number of proapoptotic and
Commitment phase antiapoptotic mechanisms determine whether the cell survives or
dies by apoptosis. The BCL-2 family of proteins consists of at least
Anti-apoptic factors Pro-apoptic factors 12 isoforms, which play important roles in this commitment phase.
Some of the BCL-2 family of proteins (eg, BCL-2 and BCL-XL) pro-
BclXL BAD tect cells from apoptosis whereas other members of the same family
Bcl–2 Bax (eg, BAD and Bax) serve proapoptotic functions. Apoptosis is exe-
cuted by a final common pathway mediated by a class of cysteine
proteases-caspases. Caspases are proteolytic enzymes present in cells
Execution phase
in an inactive form. Once cells are commited to undergo apoptosis,
Crma
these caspases are activated. Some caspases activate other caspases
Caspase activation in a hierarchical fashion resulting in a cascade of caspase activation.
p35
Eventually, caspases that target specific substrates within the cell are
? Point of no return?
activated. Some substrates for caspases that have been identified
Proteolysis of multiple include nuclear membrane components (such as lamin), cytoskeletal
intracellular substrates elements (such as actin and fodrin) and DNA repair enzymes and
transcription elements. The proteolysis of this diverse array of sub-
strates in the cell occurs in a predestined fashion and is responsible
for the characteristic morphologic features of apoptosis.
Apoptosis
Pathophysiology of Ischemic Acute Renal Failure 14.23

FIGURE 14-39
Stress Therapeutic approaches, both experimental
and in clinical use, to prevent and manage
acute renal failure based on its pathogenetic
Loss of tubular mechanisms. ETR—ET receptor; GFR—
• Restoration of Hemodynamic integrity and • Avoidance and
fluid and compromise glomerular filtration rate; HGF—hepatocyte
function discontinuation
electrolyte of nephrotoxins growth factor 1; IGF-1—insulin-like growth
balance • Survival factors factor 1; Kf—glomerular ultrafiltration coef-
• ETR antagonists PMN Back (HGF, IGF-1) ficient; NOS—nitric oxide synthase; PMN—
Kf RBF Obstruction
• Ca channel infiltration leak • ATP-Mg polymorphonuclear leukocytes; RBF—renal
inhibitors • T4
• ATP-Mg
blood flow; T4—thyroxine.
• ETR • Dopamine • ICAM-1 • Mannitol • IGF-1l • NOS inhibitors
antagonists • ANP antibody • Lasix • T4
• Ca channel • IGF-1 • RGD • ANP • HGF
inhibitors • RGD

GFR and
maintenance
phase
Restoration Reparation of
of renal tubular integrity
hemodynamics and function
Recovery

References
1. Goligorsky M, Iijima K, Krivenko Y, et al.: Role of mesangial cells in 14. Ling H, Gengaro P, Edelstein C, et al.: Injurious isoform of NOS in
macula densa-to-afferent arteriole information transfer. Clin Exp mouse proximal tubular injury. Kidney Int, 1998, 53:1642
Pharm Physiol 1997, 24:527–531. 15. Agmon Y, et al.: Nitric oxide and prostanoids protect the renal outer
2. Osswald H, Hermes H, Nabakowski G: Role of adenosine in signal medulla from radiocontrast toxicity in the rat. J Clin Invest 1994,
transmission of TGF. Kidney Int 1982, 22(Suppl. 12):S136–S142. 94:1069–1075.
3. Miller W, Thomas R, Berne R, Rubio R: Adenosine production in the 16. Agmon Y, Brezis M: NO and the medullary circulation. In: Nitric
ischemic kidney. Circ Res 1978, 43(3):390–397. Oxide and the Kidney. Edited by Goligorsky M, Gross S. New
4. Kon V, et al.: Glomerular actions of endothelin in vivo. J Clin Invest York:Chapman and Hall, 1997.
1989, 83:1762–1767. 17. Goligorsky MS: Cell biology of signal transduction. In: Hormones,
5. Gellai M, Jugus M, Fletcher T, et al.: Reversal of postischemic acute autacoids, and the kidney. Edited by Goldfarb S, Ziyadeh F. New
renal failure with a selective endothelin A receptor antagonist in the York:Churchill Livingstone, 1991.
rat. J Clin Invest 1994, 93:900–906. 18. Edelstein C, Schrier RW: The role of calcium in cell injury. In: Acute
6. Fogo, et al.: Endothelin receptor antagonism is protective in vivo in Renal Failure: New Concepts and Therapeutic Strategies. Edited by
acute cyclosporine toxicity. Kidney Int 1992, 42:770–774. Goligorsky MS, Stein JH. New York:Churchill Livingstone, 1995.
7. Conger J: NO in acute renal failure. In: Nitric Oxide and the Kidney. 19. Kribben A, Wetzels J, Wieder E, et al.:Evidence for a role of cytosolic
Edited by Goligorsky M, Gross S. New York:Chapman and Hall, free calcium in hypoxia-induced proximal tubule injury. J Clin Invest
1997. 1994, 93:1922.
8. Gross S: Nitric oxide synthases and their cofactors. In: Nitric Oxide 20. Burke T, Arnold P, Gordon J, Schrier RW: Protective effect of
and the Kidney. Edited by Goligorsky M, Gross S. New intrarenal calcium channel blockers before or after renal ischemia.
York:Chapman and Hall, 1997. J Clin Invest 1984, 74:1830.
9. Reyes A, Karl I, Klahr S: Role of arginine in health and in renal dis- 21. Kashgarian M: Stress proteins induced by injury to epithelial cells. In:
ease. Am J Physiol 1994, 267:F331–F346. Acute Renal Failure: New Concepts and therapeutic strategies. Edited
10. Kim Y-M, Tseng E, Billiar TR: Role of NO and nitrogen intermediates by Goligorsky MS, Stein JH. New York:Churchill Livingstone, 1995.
in regulation of cell functions. In: Nitric Oxide and the Kidney. Edited 22. J NIH Research
by Goligorsky M, Gross S. New York:Chapman and Hall, 1997. 23. Ueda N, Walker P, Shah SV: Oxidant stress in acute renal failure.
11. Lieberthal W:Renal ischemia and reperfusion impair endothelium- In: Acute Renal Failure: New Concepts and Therapeutic Strategies.
dependent vascular relaxation. Am J Physiol 1989, 256:F894–F900. Edited by Goligorsky MS, Stein JH. New York:Churchill
12. Yu L, Gengaro P, Niederberger M, et al.: Nitric oxide: a mediator in Livingstone, 1995.
rat tubular hypoxia/reoxygenation injury. Proc Natl Acad Sci USA 24. Kelly KJ, et al.: Antibody to anyi-cellular adhesion molecule-1 pro-
1994, 91:1691–1695. tects the kidney against ischemic injury. Proc Natl Acad Sci USA
13. Noiri E, Peresleni T, Miller F, Goligorsky MS: In vivo targeting of 1994, 91:812–816.
iNOS with oligodeoxynucleotides protects rat kidney against 25. Kroemer G, Petit P, Zamzami N, et al.: The biochemistry of pro-
ischemia. J Clin Invest 1996, 97:2377–2383. grammed cell death. FASEB J 1995, 9:1277–1287.

Vous aimerez peut-être aussi