Você está na página 1de 10

mini review http://www.kidney-international.

org
& 2012 International Society of Nephrology

Acute kidney injury and chronic kidney disease:


an integrated clinical syndrome
Lakhmir S. Chawla1,2 and Paul L. Kimmel2,3
1
Department of Anesthesiology and Critical Care Medicine, George Washington University Medical Center, Washington,
District of Columbia, USA; 2Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University
Medical Center, Washington, District of Columbia, USA and 3National Institute of Diabetes, Digestive, and Kidney Disorders,
National Institutes of Health, Bethesda, Maryland, USA

The previous conventional wisdom that survivors of acute Acute kidney injury (AKI) is responsible for approximately
kidney injury (AKI) tend to do well and fully recover renal 2 million deaths annually worldwide.13 AKI is increasingly
function appears to be flawed. AKI can cause end-stage renal common in critically ill patients, and those patients with the
disease (ESRD) directly, and increase the risk of developing most severe form of AKI, requiring renal replacement
incident chronic kidney disease (CKD) and worsening of therapy, have a mortality rate of 5080%.3 Over the past 10
underlying CKD. In addition, severity, duration, and years, there has been substantial progress in the field of AKI.
frequency of AKI appear to be important predictors of poor In particular, work on consensus definitions, epidemiologic
patient outcomes. CKD is an important risk factor for the and database studies, AKI biomarkers, and the appropriate
development and ascertainment of AKI. Experimental data dosing of renal replacement therapy has continued at a brisk
support the clinical observations and the bidirectional nature pace. The classic teaching regarding patients who survive an
of the relationships between AKI and CKD. Reductions in episode of AKI, in particular acute tubular necrosis (ATN),
renal mass and nephron number, vascular insufficiency, cell was that those patients achieved full or nearly full recovery.4
cycle disruption, and maladaptive repair mechanisms appear This notion was based on studies that followed survivors of
to be important modulators of progression in patients with AKI after hospital discharge.4,5 The most recent comprehen-
and without coexistent CKD. Distinction between AKI and sive study of AKI survivors followed 187 patients with ATN
CKD may be artificial. Consideration should be given to the for 10 years.5 The authors concluded that for those who
integrated clinical syndrome of diminished GFR, with acute survived renal function is adequate in most patients.5
and chronic stages, where spectrum of disease state and However, the notion that most patients with AKI return to
outcome is determined by host factors, including the balance pre-morbid renal function is contradicted by a series of small
of adaptive and maladaptive repair mechanisms over time. but careful clinical studies conducted over 50 years ago.6,7
Physicians must provide long-term follow-up to patients with Good clinical recovery, which is sustained, is the rule was
first episodes of AKI, even if they presented with normal renal reported by Lowe6 in his study of 14 selected patients who
function. survived an episode of oliguria or anuria associated with
Kidney International (2012) 82, 516524; doi:10.1038/ki.2012.208; ATN. Three patients (of the eight who had the assessment) in
published online 6 June 2012 this series, however, had creatinine clearance of o80 ml/min.
KEYWORDS: acute kidney injury; acute on chronic; acute renal failure; Lowe attributed any renal dysfunction to scarring or vascular
chronic kidney disease; progression; risk factor damage. Finkenstaedt and Merrill7 reported inulin clearances
of o70 ml/min in 6 of the 16 patients with no evidence of
renal disease before occurrence of acute renal failure (ARF)
1376 months after the episode. They concluded that the
findings demonstrated that an episode of ARF resulted in
more chronic renal damage than would be expected
and that subnormal renal function late in the follow-up
period occurred in the majority of cases studied. They
attributed chronic dysfunction to rupture of basement
membranes with abnormal epithelial regeneration, resulting
Correspondence: Lakhmir S. Chawla, Department of Anesthesiology and
Critical Care Medicine, George Washington University Medical Center, 900
in nephron loss.
23rd Street, NW, RILF, G-105, Washington, District of Columbia, 20037, USA. Up to 2007, however, when the epidemiology of AKI
E-mail: minkchawla@gmail.com survivorship was assessed, there was a lack of large longitudinal
Received 20 October 2011; revised 13 January 2012; accepted 21 studies to assess the effects of AKI on long-term renal function.8
February 2012; published online 6 June 2012 More recently, from 2008 through 2012, multiple observational

516 Kidney International (2012) 82, 516524


LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease mini review

studies assessing unique cohorts of patients, often using large excluded. Over the 5-year follow-up period, renal function
administrative databases, demonstrated that patients who survive deteriorated over time in all groups, but with significantly
an episode of AKI have a significant risk for progression to greater severity in those who had ATN and ARF compared
advanced-stage chronic kidney disease (CKD).914 with controls. Patients with AKI, especially those with ATN,
We review data suggesting that AKI is a cause of CKD and with pre-existing normal renal function were more likely
that prevalent CKD is a cause of incident AKI, and focus on than controls to enter stage 4 or 5 CKD. We found that
the interrelationships between these two entities. We review patients who had an episode of AKI were at high risk for the
the potential mechanistic factors underlying links between development of stage 4 CKD and had reduced survival time
AKI and CKD, and offer clinical considerations based on compared with control patients.12
these observations. Our studies in AKI survivors identified advanced age, the
presence of diabetes mellitus, and decreased baseline eGFR as
AKI AS A RISK FACTOR FOR INCIDENT CKD risk factors for the progression to advanced-stage CKD.12,15 In
Ishani et al.10 assessed a random sample of Medicare addition to these risk factors, along with our colleagues, we
beneficiaries and found that patients diagnosed with an recently showed that low serum albumin concentration (SAlb)
episode of AKI were more likely to develop end-stage renal is a strong predictor of poor long-term renal outcome.9 The
disease (ESRD) compared with patients without a history of value of the SAlb as a predictor of CKD progression is not
either AKI or CKD. In their analysis, the risk of developing surprising, because this parameter has been associated with
ESRD was eightfold higher in those with AKI compared with poor outcomes in both the general population and in a variety
patients without a history of AKI or CKD. As this study used of diseases including ESRD, surgical illness, and acute stroke.1618
ICD-9 diagnostic codes, it is difficult to determine the Low SAlb levels can be a result of nutrition-related factors
proportion of patients who suffered AKI and then progressed and/or high levels of inflammation.19,20 As several recent
directly to ESRD compared with patients who suffered AKI, studies have shown that markers of inflammation predict
recovered renal function, and then progressed to ESRD.10 AKI, it is likely that the relationship between SAlb level and
Lo et al.,11 by using a Kaiser Permanente database in CKD progression is based on a complex set of factors,
Northern California, retrospectively studied inpatient admis- including those related to diet, nutrition, and catabolism, as
sions. Patients with an episode of AKI treated with dialysis were well as increased inflammation.15,21
compared with patients without an episode of AKI. Patients Collectively, these studies underscore the strong associa-
with a history of ESRD or a preadmission estimated glomerular tion between AKI and the future development of CKD.
filtration rate (eGFR) o45 ml/min per 1.73 m2 were excluded.
In this study, the investigators found that an episode of SEVERITY, DURATION, AND FREQUENCY OF AKI AND CKD
dialysis-requiring AKI was associated with a 28-fold increased PROGRESSION
risk of developing advanced CKD, and a 2-fold increase in More recently, severity of AKI has been linked to CKD
mortality. As patients who developed ESRD within 30 days progression in survivors of AKI. Ishani et al.22 assessed patients
of discharge were excluded from the long-term follow-up of who underwent cardiac surgery, and found that the magnitude
advanced CKD, these data are consistent with a severe form of serum creatinine concentration during the postoperative
of AKI followed by renal recovery and then progression to hospital course was directly linked to progression. This effect
advanced-stage CKD.11 It should be noted that some patients was seen in patients with AKI with previously normal renal
assessed had pre-existing CKD. function (de novo AKI), as well as in patients with an episode
Wald et al.13 conducted a population-based cohort study of AKI superimposed on CKD (acute on chronic renal
in Ontario, Canada of AKI patients who required in-hospital failure).22 The magnitude of increase in serum creatinine after
dialysis and survived free of dialysis for at least 30 days after cardiac surgery was associated in a graded manner with
discharge. These patients were matched with patients without increased risk of CKD progression and mortality.
AKI or dialysis therapy during their index hospitalization. This same trend has been demonstrated in patients who
Patients with AKI requiring dialysis were over three times had percutaneous coronary revascularization. James et al.23
more likely to develop ESRD compared with control-matched showed that patients undergoing percutaneous coronary
patients. revascularization who had AKI were at risk for future develop-
We and colleagues at the Veterans Affairs Medical Center ment of ESRD. Patients with AKIN stage I AKI were over 4
in Washington, DC assessed a United States Department of times more likely to develop ESRD, whereas patients with AKIN
Veterans Affairs database to ascertain long-term renal function stage II/III were over 11 times more likely to develop ESRD.23
in over 79,000 hospitalized patients with and without AKI.12 We have similarly shown that the severity of AKI is linked
A particular focus was on the long-term outcome in patients to CKD progression.9 We assessed a cohort of 11,589 patients
with a diagnosis of ATN. Patients hospitalized for acute with a spectrum of AKI from RIFLE stage R through F.9
myocardial infarction or pneumonia without AKI were desig- Severity of AKI, assessed by peak serum creatinine, was
nated as controls. The remaining 5404 hospitalized patients associated with progression to advanced CKD (Figure 1). In
who comprised the AKI group had diagnostic codes indicat- particular, patients who required dialysis were at much
ing ARF or ATN. Patients with pre-existing CKD were higher risk for progression to CKD than patients with less

Kidney International (2012) 82, 516524 517


mini review LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease

a b
3.5 Mean eGFR 15 years post tertile 100
Tertile 1 (n = 767) 90
3.0
Tertile 2 (n = 766) Tertile 1: mean

Mean Scr (mg/dI)


80 eGFR>79.2

Mean eGFR
2.5 Tertile 3 (n = 767)
70
2.0 Tertile 2: mean
60 eGFR 61-79.2
1.5
50
1.0 Tertile 3: mean
40 eGFR<61
0.5 30

re

st

st

st
e

st

st

st

ita
pr

in

po

po

po
po

po

po

rp

sp
ur
ar

Ho

rs
s

rs
D

ye

th

th
ye

th

th

a
a

on

on

ye
on

on

ye

1
1

m
m

5
5

1
1

12
3

12

1
1

3
3
Time Time
9
Figure 1 | Effect of severity of acute kidney injury (AKI) on outcomes. AKI patients who survived for 1 year. (a) Mean eGFR over time
(tertiles). (b) AKI patients who survived for 1 year. Mean serum creatinine (Scr) over time (tertiles). Tertiles were defined based on Scr at 15
years post admission. Error bars show the 95% confidence interval at each time point.

severe AKI.9 Although it is seemingly intuitive that the 1

severity of AKI would be associated with progression to


advanced CKD, these three studies are the first to show this 0.8

link.9,22,23 These findings suggest that when the severity of Proportion surviving

AKI reaches a certain threshold the course of AKI is altered, 0.6


No AKI
(P < 0.0001)
initiating a chronic, progressive disease.
1 AKI
In addition to severity, the duration of AKI has been 0.4 2 AKIs
linked to mortality but not to CKD progression. Coca et al.24
3+AKIs
assessed a large cohort of patients with diabetes mellitus 0.2
undergoing cardiac surgery in the Veterans Administration
system. Both severity and duration of AKI were linked to
0
long-term mortality. However, the duration of AKI was not 0 20 40 60 80 100
linked to CKD progression.24 Consistent with these findings, Months of follow-up from baseline

Brown et al.25 assessed a separate cohort of US Veterans after Figure 2 | Effect of acute kidney injury (AKI) frequency on
cardiac surgery and confirmed that the duration of AKI was outcomes.26 Survival to stage 4 chronic kidney disease (CKD) in
linked to worse long-term survival. no AKI vs. multiple AKI episode groups.
Not surprisingly, patients who sustain multiple episodes of
AKI as compared with a single episode of AKI have higher
likelihoods of CKD progression. Thakar et al.26 have shown, authors hypothesized that physician-related decisions
in a cohort of US Veterans with diabetes, that those who to start chronic dialysis earlier might account for this
experienced two or more episodes of AKI were much more discrepancy. We believe that the more likely explanation is
likely to progress to stage 4 CKD than patients who the marked effect of AKI on the development and progres-
experienced only one episode of AKI (Figure 2).26 sion of CKD.
These data are consistent with the hypothesis that for some CKD has consistently been shown to be a significant risk
patients a single episode of AKI has biologic ramifications factor for the development of AKI.28,29 Probable explanations
beyond the acute event, engendering an ongoing state that include the hemodynamic instability and failure of auto-
predisposes to the development of further injury, manifested regulation30 in CKD patients, the ease of detection of small
differentially in time as worsened AKI (short-term) or the changes in GFR when renal function is impaired, and a
development or worsening of CKD over longer periods predisposition to further injury in patients with diminished
(Figure 3). Some patients can fully recover from their initial renal function.31,32 These consist of at least susceptibility to
AKI, but subsets of AKI survivors appear to go on to nephrotoxic agents, and the effects of ongoing humoral and
experience vicious cycles of intertwined AKI and CKD.26 It is renal pathologic mechanisms in the setting of CKD.
likely that the severity of renal injury along with other clinical, Unfortunately, this risk appears to be bidirectional. Ishani
treatment, and host risk factors mediate such processes. et al.10 showed that patients with CKD who experienced an
episode of AKI were 41 times more likely to develop ESRD
AKI AS AN ACCELERANT OF CKD than patients without kidney disease, whereas patients with
Hsu et al.27 showed that the incident growth of the ESRD CKD and no episodes of AKI had an 8.4-fold higher risk
population exceeded the prevalent CKD population. The compared with patients without kidney disease. The risk of

518 Kidney International (2012) 82, 516524


LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease mini review

100%
AKI Repair Chronic 34% had either reduced kidney function or were dialysis
dependent upon discharge.36 The most compelling data
regarding AKI and the risk of CKD in children came from a
long-term follow-up of participants in the aforementioned
study conducted by Askenazi et al.37 They followed up a
cohort of children for 3 to 5 years after an episode of AKI.
GFR Mortality was 79.9%, with the majority of the deaths
occurring within 2 years of the AKI episode. In all, 17 of
29 patients followed up in an outpatient clinic demonstrated
evidence of CKD, manifested by hyperfiltration, reduced
kidney function, hypertension, or microalbuminuria.37
Although this represents a small sample, it is consistent with
animal and adult human studies showing that subsets of
patients who develop AKI are at high risk for developing
CKD and experiencing progression.
0 Time
These data suggest that for some patients AKI and CKD
Figure 3 | Theoretical range of outcomes after acute kidney likely coexist in an intimate but vicious cycle. This concept is
injury (AKI). The range of possible long-term outcomes after an supported by the fact that CKD was thought to progress in a
episode of AKI comprises the entire spectrum of the glomerular
filtration rate (GFR). The final GFR is determined by the level of linear manner,38 but this is not always the case. Shah and
GFR at the time of injury, the severity and duration of the injury, Levey39 showed that as many as one-third of patients deviated
and responses across several phases over time. It is envisioned from their reciprocal creatinine slope. The African American
that repair and chronic phases have different and variable time Study of Kidney Disease and Hypertension study has delinea-
frames, and different magnitudes of responses. Responses during
repair and chronic phases are determined by the balance of ted several different courses in patients with CKD (personal
processes that result in amelioration and worsening of GFR. communication, Tom Greene). Some of these courses are
consistent with the notion of intermittent episodes of AKI
complicating CKD, with varying levels of repair occurring.
developing ESRD was enhanced almost fourfold by the An episode of AKI that results in a decrement in GFR will
superimposition of AKI in CKD patients. AKI enhanced the necessarily affect the trajectory of GFR change; the course will
risk almost 10-fold compared with patients without either ultimately be determined by whether the injury is reversible
CKD or AKI, and astonishingly was associated with greater (volume-related changes in GFR and filtration fraction) and
risk of developing ESRD than those identified with CKD in by the balance between effective and maladaptive repair
the Medicare population. Similarly, Hsu et al.33 showed that mechanisms (Figure 4). The exact mechanism(s) by which
patients with an eGFR o45 ml/min per 1.73 m2 who AKI accelerates/initiates CKD in humans are unknown.
experienced an episode of dialysis-requiring AKI were at Preclinical data, however, suggest that a variety of mechanisms
very high risk for impaired recovery of renal function. In a may be operative.
population-based cohort in Scotland, Ali et al.1 showed that
only 64% of patients with an episode of acute on chronic MECHANISMS UNDERLYING PROGRESSION OF AKI TO CKD
renal failure had full recovery of kidney function.1 An important concept for understanding the course of CKD is
In these studies, CKD was defined by an eGFR of that the progression to ESRD occurs via processes that may
460 ml/min per 1.73 m2. In addition to low baseline eGFR, be independent of the original pathology of CKD.32,40 Such
patients with CKD as assessed by proteinuria are at increased concepts were originally proposed for a variety of glomerular
risk for AKI. James et al.29 showed in a cohort of over 920,000 diseases,40 after follow-up studies in patients with post-
patients that the level of proteinuria and diminished baseline streptococcal glomerulonephritis delineated long-term sequelae41
eGFR were independent risk factors for developing AKI.29 linked to current definitions of CKD, including decrements
The risk of progression to CKD in patients who develop in GFR and the persistence of proteinuria, as well as the
AKI is not limited to adults but affects children as well.34,35 development of hypertension. Data from animal models
Large pediatric cohort studies suggest that certain subsets of suggest that kidneys after episodes of injury may be sus-
children (such as bone marrow transplant patients) are at ceptible to progression to CKD, which is similarly indepen-
high risk for CKD progression. In contrast to adults, children dent of the initial cause of AKI. Key constructs regarding CKD
tend to be less burdened with chronic disease. Thus, the progression are based on findings from the remnant kidney
evidence that AKI in children puts those patients at risk for model in which nephron loss leads to glomerular hypertrophy
CKD suggests processes subsequent to AKI that are integral in the remaining nephrons.42 Hypothesized mechanisms
to the acute episode, as opposed to simply changes that occur underlying CKD progression include effects of systemic and
in an acute on chronic disease paradigm. intrarenal hypertension and hyperfiltration, tubular hyper-
Hui-Stickle et al.36 assessed pediatric intensive care unit trophy, and hypertension resulting in arteriosclerosis, tubu-
patients upon discharge from a tertiary care center. In all, lointerstitial fibrosis, and glomerulosclerosis,4345 as well as

Kidney International (2012) 82, 516524 519


mini review LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease

c
Normal repair
and regeneration

a b
ATN
Neutrophils and
monocytes
Interstitium
Endothelial
injury d Maladaptive
response
Zones of
hypoxia
Interstitial
fibrosis

Peri-tubular
blood vessels
Cells
and casts
Depleted
blood supply
Cell cycle arrest
Epigenetic changes

Figure 4 | Tubule cross-section. (a) Cross section of normal renal tubule with intact epithelial cells, renal interstitium, and peri-tubular
blood vessels. (b) Cross section of renal tubule with acute tubular necrosis (ATN) with epithelial cell necrosis, intra-tubular cast formation,
endothelial injury of peri-tubular blood vessels, and migration of monocytes and macrophages into renal interstitium. (c) Cross section of
renal tubule after normal repair and regeneration showing restoration of normal renal architecture. (d) Cross section of renal tubule after
severe episode of AKI, resulting in maladaptive repair. Epithelial cells have evidence of cell cycle arrest and epigenetic changes that favor a
fibrosis phenotype. Renal interstitium shows evidence of fibrosis. Post-injury vascular supply is less dense than baseline. The combination
of decreased blood supply and fibrosis leads to zones of hypoxia wherein the combination of decreased vascular supply and fibrosis can
initiate a vicious cycle leading to ongoing fibrosis.

the effects of derangements of the endocrine response and hypertrophy and hyperfiltration via hypertension control and
abnormalities in circulating mediators associated with decre- reninangiotensinaldosterone system blockade.
ments in renal function. The latter may affect cell structure and
function, as well as physiologic responses. In the case of normal Interstitial inflammation and fibrosis
kidneys after an episode of injury, multiple injury pathways and In human CKD and in the remnant kidney model,
maladaptive processes have been identified, which may tubulointerstitial fibrosis predominates over glomerulo-
contribute to determining the course of renal function after sclerosis.32 As a consequence, human CKD often progresses
an initial insult. without the need for continued glomerular injury.45,4951
Further support of a pro-fibrotic effect in the setting of
Nephron loss and glomerular hypertrophy reduced renal mass is provided by experiments that show that
The pathophysiology of loss of nephron mass followed by the kidneys of animals exposed to ischemiareperfusion (IR)
glomerular hypertrophy has been well described in animal injury are more predisposed to developing fibrosis if nephron
models.43 After an episode of AKI, it is possible that signi- loss (e.g., unilateral nephrectomy) occurs before injury.52,53
ficant permanent loss of nephron mass results in a clinical In addition, inflammation after AKI can lead to interstitial
state comparable to that of the remnant kidney model. After immune cell infiltration followed by interstitial fibrosis.
subtotal nephrectomy, single-nephron GFR increases via Inflammation has been shown to be an important feature of
hyperfiltration and glomerular hypertrophy. Cellular prolif- ischemic and septic AKI.15,5456 Animal models of ATN
eration ensues, ultimately resulting in tubulointerstitial fibrosis consistently show an interstitial neutrophil infiltrate during
and progressive nephron loss.32 These processes are exacer- the acute phase of ATN, often followed by monocytic
bated by high-salt and high-protein diets.46,47 Increased lymphocytic infiltration in later stages.32,55,57,58 Monocytes
tubular workload predisposes nephron segments to fibrosis can exacerbate injury and promote fibrosis.59 The effects of
and senescence, which can be mitigated by protein and calorie tubulointerstitial fibrosis are critical, as tubulointerstitial
restriction.47,48 fibrosis injury scores predict the decline of renal function
The lessons from animal studies have been successfully better than glomerular injury scores.60,61 Reduced renal
translated into patient-care paradigms. As an example, a mass impairs recovery of experimental AKI and may
cornerstone of CKD therapy is mitigation of glomerular predispose to the development of fibrosis.32 In most models

520 Kidney International (2012) 82, 516524


LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease mini review

of experimental AKI, renal tubular regeneration occurs, but involving the cell cycle of reparative cells have shed light on
depending on the severity of the initial damage the recovery such processes.72,73
is often incomplete. Areas that do not complete full recovery When AKI is severe, the corresponding tubular injury and
may develop into focal areas of fibrosis.32,55,62 interstitial inflammation are associated with proliferation of
Both human and animal models link severity and tubular epithelial cells. Yang et al.63 showed that when kidney
frequency of AKI with worsening of long-term renal damage was accompanied by fibrosis there were large
function.9,22,63 Multiple episodes of IR-induced AKI have numbers of proximal tubule epithelial cells arrested in the
been shown to cause CKD and tubulointerstitial fibrosis in G2/M phase of the cell cycle. In contradistinction to prolif-
animal models. Thus, it is conceivable that subclinical erating epithelial cells, the arrested cells produced greater
episodes of AKI could both cause and worsen CKD.32 These levels of transforming growth factor-b1 and connective tissue
data are consistent with the previously cited human data growth factor. These findings suggest that in response to
showing the effect of multiple episodes of AKI to enhance injury a maladaptive repair process may ensue, causing
CKD progression.26 fibrosis instead of repair of tubular epithelia. Cell cycle arrest
causes tubular epithelial cells to convert to a phenotype that
Endothelial injury and vascular rarefaction promotes the growth and activation of fibroblasts. Interest-
Experimental models of AKI demonstrate endothelial ingly, in this same study, moderate IR injury did not
involvement and vascular injury. Tubular repair after injury produce this maladaptive process. Only severe injury with IR
tends to be robust, but the vascular restorative capacity after or aristolochic acid activated pro-fibrotic processes. Cell arrest
AKI is more blunted.64 Various different injury models (e.g., was mitigated by agents, such as p53 inhibitors, that affect the
IR, folate, nitric oxide synthase inhibition, and ureteral cell cycle, and was exacerbated by agents that prolong cell
obstruction) all demonstrate diminished vascular density cycle arrest.63
after injury.6568 The loss of vascular density varies between Animal studies support the notion that fibrosis is ongoing
30 and 50%.69 The loss of vascular reserve may be one after AKI. Bechtel et al.69 investigated mechanisms of fibrosis
of the key components of the development of fibrosis that were temporally remote from the initiation of kidney
after injury. Injured tissue that has insufficient vascular injury. They showed that fibrosis continues owing to a failure
supply becomes hypoxic, potentially setting into motion a of fibroblasts to return to their resting state. Epigenetic
self-propagating injury cascade. Rarefaction of capillaries factors including hypermethylation of RASAL1 gene loci were
initiates activation of hypoxia-inducible pathways that can involved. Both studies by Yang et al.63 and Bechtel et al.69
promote inflammation and downstream fibrosis.32 Thus, implicate the pathologic role of transforming growth factor-b
capillary rarefaction in fibrotic foci can be both a cause and in promoting fibrosis.63,69
an effect of tissue hypoxia, in turn leading to activation of When tubular epithelial cells arrest in the G2/M phase of
hypoxic signaling, inflammation, and activation of factors the cell cycle, they produce transforming growth factor-b1.
enhancing fibrosis. If transforming growth factor-b1 secretion and activation
In experimental models, treatment with angiogenic factors is sustained, epigenetic changes in fibroblasts may ensue,
such as vascular endothelial growth factor-121 ameliorates which can transform them into tumor-like myofibroblasts
vascular dropout and endothelial damage.54 However, these that proliferate in a growth factorindependent manner.74
compounds are only useful in the immediate post-injury Altogether, cell cycle arrest and epigenetic changes represent
period, and do not improve vascular dropout after AKI when at least two maladaptive repair mechanisms that appear to
administered 43 weeks after the insult.32 have major roles in determining the post-AKI fibrotic
phenotype. As cell cycle arrest and epigenetic changes appear
Cell cycle and maladaptive repair to be sequential in nature, they may form pathways amenable
Normal repair processes involve a host of growth factors and to interruption.63,73,75,76
proliferative and other signaling cascades, which must be
deployed, and function in coordinated, integrated temporal, Identification of patients and risk factors for CKD progression
and spatial manners. These mechanisms are critical for To help clinicians identify AKI survivors at increased risk for
appropriate repair and regeneration.70 Cell cycle abnormalities CKD development and progression, along with our collea-
affect the outcome of AKI. Preclinical studies demonstrate gues, we developed three multivariable models. The most
that p21, a cyclin-dependent kinase, is a critical check- accessible (and therefore clinically useful) model is based on
point effector for induction of the cell cycle.71 In preclinical sentinel clinical events (i.e., RIFLE stage, need for dialysis,
models of AKI, p21 antagonism worsens AKI, whereas p21 baseline eGFR, and sAlb).9 This model performed well,
induction improves outcomes in AKI models.72,73 Cell cycle explaining 11% of the variance in the development of CKD
dysfunction can have effects at the point of injury and further (area under ROC curve 0.77). We validated this equation in
downstream during repair. Pathways that affect the cell cycle a separate large cohort of hospitalized Veterans (Po0.001,
can also activate fibroblasts and inflammation. Selected anti- c-statisticX0.81). As these end points are accessible and
inflammatory therapies have been shown to decrease the clinically relevant, clinicians could potentially use such
severity of AKI during its acute phase.32,74 Recently, studies equations to risk stratify AKI survivors at highest risk for

Kidney International (2012) 82, 516524 521


mini review LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease

progression to CKD for therapeutic reasons or for inclusion balance of adaptive and maladaptive repair determine
in clinical trials.9 clinically apparent outcomes. The range of change in
In addition to clinical models, novel AKI biomarkers show response to differential injury and the balance of adaptive
promise in improving risk assessment of patients with severe and maladaptive repair in patients with different initial levels
AKI. Srisawat et al.77 have shown that plasma neutrophil of renal function comprise the entire spectrum of GFR
gelatinaseassociated lipocalin improves the discrimination of (Figure 3). Interventions to preserve function and enhance
clinical models to predict renal recovery. Future studies repair are presumably appropriate at several stages of the
should incorporate plasma and urine biomarkers in risk disease. Commonalities and differences in mechanisms
assessment strategies to determine the best approach to mediating the different courses of long-term outcome must
identify patients at risk for CKD progression. be delineated and translated into clinical trials and practice
over the next decade.
Summary The giants, such as Lowe and Merrill, on whose shoulders
The paradigm of AKI has markedly shifted over the past 5 we stand, were correct, even if their early findings were lost,
years. The previous conventional wisdom that survivors of misinterpreted, or incorrectly recalled. AKI and CKD
AKI tend to do well and fully recover renal function, perhaps comprise an intertwined clinical entity, a disease of
based on indistinct recollections of early, limited studies, diminished renal mass resulting in diminished GFR.
appears to be flawed.6,7 AKI can cause ESRD directly,8,10,78
and appears to increase the risk for incident CKD and worsen Recommendations and future directions
underlying CKD.12,22 In addition, severity and frequency of We recommend that hospital survivors of AKI who experience
AKI appear to be important predictors of poorer out- severe AKI (AKIN stage III) be followed up by a nephrologist
comes.9,26 Experimental data support the clinical observa- after discharge. Currently, this does not occur. At 30 days after
tions. Mechanistically, renal mass, vascular insufficiency, cell cycle discharge from the hospital with an episode of AKI, 74.5% of
disruption, and maladaptive repair appear to be important patients were seen by a primary physician compared with
modulators of outcome, and these pathophysiologic processes 11.9% and 29.5%, respectively, who were seen by a nephrologist
are likely operative in mediating the poor outcomes observed or a cardiologist.82 Surprisingly, only about one-third of AKI
in the clinical epidemiologic studies reviewed above. patients requiring dialysis (AKIN stage III) are seen by a
The public health impact of the long-term outcomes of nephrologist within 30 days of discharge. This increases to
AKI is significant. The population incidence of AKI is 48.6% within 1 year of discharge, which is likely a result of a
approximately 2100 per million population.1 Given the rising serum creatinine.82 Overall, a very small fraction of AKI
population of the developed world (USA, Canada, Western patients are currently followed up by nephrologists after
Europe, and Australia) of approximately 1 billion, there hospital discharge.82 To put this into context, an assessment
will be over 2 million cases of AKI this year, with an expected of the nephrology follow-up of AKI survivors as compared with
1.5 million AKI survivors. Of these patients, approxi- the cardiology follow-up of myocardial infarction survivors
mately 1520% will progress to advanced-stage CKD within reveals a stark difference (11.9% vs. 76%, respectively).31,82 At
24 months, resulting in approximately 300,000 cases of the very least, we should continue to see our patients.
advanced CKD per year. Given the increasing incidence of Monitoring patients for blood pressure control, the use of
AKI in the aging population,8,79,80 the projected incidence is drugs that interfere with the reninangiotensin system, and
expected to increase. The National Institute of Diabetes, avoidance of nephrotoxins may prove critical as public health
Digestive and Kidney Disease supports the Assessment, Serial measures, which may oppose current secular trends.
Evaluation, and Subsequent Sequelae of Acute Kidney Injury
(ASSESS AKI) study, which evaluates the long-term outcome DISCLOSURE
of hospitalized patients, with and without CKD, who All the authors declared no competing interests.
experience an episode of AKI, to determine the natural
ACKNOWLEDGMENTS
history of AKI and delineate risk factors for progression and We thank Paul Eggers and Richard Amdur for reviewing the
development of complications, including cardiovascular manuscript.
disease.81
We propose a new model integrating acute and CKD. The DISCLAIMER
separation of ARF and CKD into two distinct syndromes as The views expressed in this article do not necessarily represent the
taught in medical school is not in the best interest of an views of the Department of Health and Human Services, the National
Institutes of Health, the National Institute of Diabetes, Digestive and
integrated approach to the long-term care of patients with Kidney Diseases, or the United States Government.
kidney disease. Rather, we propose consideration of the state
of diminution in GFR as a clinical entity, which has REFERENCES
differential initiation and expression in time and along the 1. Ali T, Khan I, Simpson W et al. Incidence and outcomes in acute kidney
spectrum of magnitude of GFR (Figure 3). Phases of disease injury: a comprehensive population-based study. J Am Soc Nephrol 2007;
18: 12921298.
include initiation, repair, and long-term outcomes (Figure 3 2. Murugan R, Kellum JA. Acute kidney injury: whats the prognosis? Nat Rev
and/or 4). Differences in timing and severity of injury and the Nephrol 2011; 7: 209217.

522 Kidney International (2012) 82, 516524


LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease mini review

3. Uchino S, Kellum JA, Bellomo R et al. Acute renal failure in critically ill 31. Daugherty SL, Ho PM, Spertus JA et al. Association of early follow-up after
patients: a multinational, multicenter study. JAMA 2005; 294: 813818. acute myocardial infarction with higher rates of medication use. Arch
4. Kjellstrand CM, Ebben J, Davin T. Time of death, recovery of renal Intern Med 2008; 168: 485491 discussion 492.
function, development of chronic renal failure and need for chronic 32. Venkatachalam MA, Griffin KA, Lan R et al. Acute kidney injury: a
hemodialysis in patients with acute tubular necrosis. Trans Am Soc Artif springboard for progression in chronic kidney disease. Am J Physiol Renal
Intern Organs 1981; 27: 4550. Physiol 2010; 298: F1078F1094.
5. Liano F, Felipe C, Tenorio MT et al. Long-term outcome of acute tubular 33. Hsu CY, Chertow GM, McCulloch CE et al. Nonrecovery of kidney function
necrosis: a contribution to its natural history. Kidney Int 2007; 71: 679686. and death after acute on chronic renal failure. Clin J Am Soc Nephrol 2009;
6. Lowe KG. The late prognosis in acute tubular necrosis; an interim follow- 4: 891898.
up report on 14 patients. Lancet 1952: 10861088. 34. Goldstein SL, Devarajan P. Acute kidney injury in childhood: should we be
7. Finkenstaedt JT, Merrill JP. Renal function after recovery from acute renal worried about progression to CKD? Pediatr Nephrol 2011; 26: 509522.
failure. N Engl J Med 1956; 254: 10231026. 35. Goldstein SL, Devarajan P. Progression from acute kidney injury to
8. Xue JL, Daniels F, Star RA et al. Mortality and advancing to end-stage chronic kidney disease: a pediatric perspective. Adv Chronic Kidney Dis
renal disease in patients with hospital and non-hospital acquired renal 2008; 15: 278283.
failure. J Am Soc Nephrol 2007; 15: SA-PO965. 36. Hui-Stickle S, Brewer ED, Goldstein SL. Pediatric ARF epidemiology
9. Chawla LS, Amdur RL, Amodeo S et al. The severity of acute kidney injury at a tertiary care center from 1999 to 2001. Am J Kidney Dis 2005; 45:
predicts progression to chronic kidney disease. Kidney Int 2011; 79: 96101.
13611369. 37. Askenazi DJ, Feig DI, Graham NM et al. 35 Year longitudinal follow-up
10. Ishani A, Xue JL, Himmelfarb J et al. Acute kidney injury increases risk of of pediatric patients after acute renal failure. Kidney Int 2006; 69:
ESRD among elderly. J Am Soc Nephrol 2009; 20: 223228. 184189.
11. Lo LJ, Go AS, Chertow GM et al. Dialysis-requiring acute renal failure 38. Mitch WE, Walser M, Buffington GA et al. A simple method of estimating
increases the risk of progressive chronic kidney disease. Kidney Int 2009; progression of chronic renal failure. Lancet 1976; 2: 13261328.
76: 893899. 39. Shah BV, Levey AS. Spontaneous changes in the rate of decline in
12. Amdur RL, Chawla LS, Amodeo S et al. Outcomes following diagnosis of reciprocal serum creatinine: errors in predicting the progression of renal
acute renal failure in U.S. veterans: focus on acute tubular necrosis. Kidney disease from extrapolation of the slope. J Am Soc Nephrol 1992; 2:
Int 2009; 76: 10891097. 11861191.
13. Wald R, Quinn RR, Luo J et al. Chronic dialysis and death among survivors 40. Neugarten J, Feiner HD, Schacht RG et al. Aggravation of experimental
of acute kidney injury requiring dialysis. JAMA 2009; 302: 11791185. glomerulonephritis by superimposed clip hypertension. Kidney Int 1982;
14. Garg AX, Suri RS, Barrowman N et al. Long-term renal prognosis of 22: 257263.
diarrhea-associated hemolytic uremic syndrome: a systematic review, 41. Baldwin DS. Poststreptococcal glomerulonephritis. A progressive disease?
meta-analysis, and meta-regression. JAMA 2003; 290: 13601370. Am J Med 1977; 62: 111.
15. Chawla LS, Seneff MG, Nelson DR et al. Elevated plasma concentrations of 42. Ingelfinger JR. Disparities in renal endowment: causes and consequences.
IL-6 and elevated APACHE II score predict acute kidney injury in patients Adv Chronic Kidney Dis 2008; 15: 107114.
with severe sepsis. Clin J Am Soc Nephrol 2007; 2: 2230. 43. Hostetter TH. Progression of renal disease and renal hypertrophy. Annu
16. Engelman DT, Adams DH, Byrne JG et al. Impact of body mass index and Rev Physiol 1995; 57: 263278.
albumin on morbidity and mortality after cardiac surgery. J Thorac 44. Hostetter TH, Olson JL, Rennke HG et al. Hyperfiltration in remnant
Cardiovasc Surg 1999; 118: 866873. nephrons: a potentially adverse response to renal ablation. Jam Soc
17. Gariballa SE, Parker SG, Taub N et al. Influence of nutritional status on Nephrol 2001; 12: 13151325.
clinical outcome after acute stroke. Am J Clin Nutr 1998; 68: 275281. 45. Baldwin DS, Neugarten J. Hypertension and renal diseases. Am J Kidney
18. Owen Jr WF, Lew NL, Liu Y et al. The urea reduction ratio and serum Dis 1987; 10: 186191.
albumin concentration as predictors of mortality in patients undergoing 46. Shapiro JI, Elkins N, Reiss OK et al. Energy metabolism following reduction
hemodialysis. N Engl J Med 1993; 329: 10011006. of renal mass. Kidney Int Suppl 1994; 45: S100S105.
19. Friedman AN, Fadem SZ. Reassessment of albumin as a nutritional marker 47. Nath KA, Croatt AJ, Hostetter TH. Oxygen consumption and oxidant stress
in kidney disease. J Am Soc Nephrol 2010; 21: 223230. in surviving nephrons. Am J Physiol 1990; 258: F1354F1362.
20. Don BR, Kaysen G. Serum albumin: relationship to inflammation and 48. Tapp DC, Wortham WG, Addison JF et al. Food restriction retards
nutrition. Semin Dial 2004; 17: 432437. body growth and prevents end-stage renal pathology in remnant
21. Liu KD, Glidden DV, Eisner MD et al. Predictive and pathogenetic value of kidneys of rats regardless of protein intake. Lab Invest 1989; 60:
plasma biomarkers for acute kidney injury in patients with acute lung 184195.
injury. Crit Care Med 2007; 35: 27552761. 49. Risdon RA, Sloper JC, De Wardener HE. Relationship between renal
22. Ishani A, Nelson D, Clothier B et al. The magnitude of acute serum function and histological changes found in renal-biopsy specimens
creatinine increase after cardiac surgery and the risk of chronic kidney from patients with persistent glomerular nephritis. Lancet 1968; 2:
disease, progression of kidney disease, and death. Arch Intern Med 2011; 363366.
171: 226233. 50. Schainuck LI, Striker GE, Cutler RE et al. Structural-functional
23. James MT, Ghali WA, Knudtson ML et al. Associations between acute correlations in renal disease. II. The correlations. Hum Pathol 1970; 1:
kidney injury and cardiovascular and renal outcomes after coronary 631641.
angiography. Circulation 2011; 123: 409416. 51. Striker GE, Schainuck LI, Cutler RE et al. Structural-functional correlations
24. Coca SG, King Jr JT, Rosenthal RA et al. The duration of postoperative in renal disease. I. A method for assaying and classifying histopathologic
acute kidney injury is an additional parameter predicting long-term changes in renal disease. Hum Pathol 1970; 1: 615630.
survival in diabetic veterans. Kidney Int 2010; 78: 926933. 52. Azuma H, Nadeau K, Takada M et al. Cellular and molecular predictors of
25. Brown JR, Kramer RS, Coca SG et al. Duration of acute kidney injury chronic renal dysfunction after initial ischemia/reperfusion injury of a
impacts long-term survival after cardiac surgery. Ann Thorac Surg 2010; single kidney. Transplantation 1997; 64: 190197.
90: 11421148. 53. Azuma H, Nadeau K, Takada M et al. Initial ischemia/reperfusion injury
26. Thakar CV, Christianson A, Himmelfarb J et al. Acute kidney injury influences late functional and structural changes in the kidney. Transplant
episodes and chronic kidney disease risk in diabetes mellitus. Clin J Am Proc 1997; 29: 15281529.
Soc Nephrol 2011; 6: 25672572. 54. Liu KD, Glidden DV, Eisner MD et al. Predictive and pathogenetic value of
27. Hsu CY, Vittinghoff E, Lin F et al. The incidence of end-stage renal disease plasma biomarkers for acute kidney injury in patients with acute lung
is increasing faster than the prevalence of chronic renal insufficiency. Ann injury*. Crit Care Med 2007; 35: 27552761.
Intern Med 2004; 141: 95101. 55. Devarajan P. Update on mechanisms of ischemic acute kidney injury.
28. Thakar CV, Arrigain S, Worley S et al. A clinical score to predict acute renal J Am Soc Nephrol 2006; 17: 15031520.
failure after cardiac surgery. J Am Soc Nephrol 2005; 16: 162168. 56. Leelahavanichkul A, Huang Y, Hu X et al. Chronic kidney disease worsens
29. James MT, Hemmelgarn BR, Wiebe N et al. Glomerular filtration rate, sepsis and sepsis-induced acute kidney injury by releasing high mobility
proteinuria, and the incidence and consequences of acute kidney injury: group box protein-1. Kidney Int 2011; 80: 11981211.
a cohort study. Lancet 2010; 376: 20962103. 57. Bonventre JV, Weinberg JM. Recent advances in the pathophysiology of
30. Bidani AK, Griffin KA, Williamson G et al. Protective importance of the ischemic acute renal failure. J Am Soc Nephrol 2003; 14: 21992210.
myogenic response in the renal circulation. Hypertension 2009; 54: 58. Jang HR, Rabb H. The innate immune response in ischemic acute kidney
393398. injury. Clin Immunol 2009; 130: 4150.

Kidney International (2012) 82, 516524 523


mini review LS Chawla and PL Kimmel: Acute kidney injury and chronic kidney disease

59. Castano AP, Lin SL, Surowy T et al. Serum amyloid P inhibits fibrosis 70. Bonventre JV. Dedifferentiation and proliferation of surviving epithelial
through Fc gamma R-dependent monocyte-macrophage regulation cells in acute renal failure. J Am Soc Nephrol 2003; 14Suppl 1: S55S61.
in vivo. Sci Transl Med 2009; 1: 5ra13. 71. Price PM, Safirstein RL, Megyesi J. The cell cycle and acute kidney injury.
60. Nath KA. Tubulointerstitial changes as a major determinant in the Kidney Int 2009; 76: 604613.
progression of renal damage. Am J Kidney Dis 1992; 20: 117. 72. Hodeify R, Tarcsafalvi A, Megyesi J et al. Cdk2-dependent phosphorylation
61. Bohle A, von Gise H, Mackensen-Haen S et al. The obliteration of the of p21 regulates the role of Cdk2 in cisplatin cytotoxicity. Am J Physiol
postglomerular capillaries and its influence upon the function of both Renal Physiol 2011; 300: F1171F1179.
glomeruli and tubuli. Functional interpretation of morphologic findings. 73. Megyesi J, Price PM, Tamayo E et al. The lack of a functional p21(WAF1/
Klinische Wochenschrift 1981; 59: 10431051. CIP1) gene ameliorates progression to chronic renal failure. Proc Natl
62. Finn WF. Enhanced recovery from postischemic acute renal failure. Acad Sci USA 1999; 96: 1083010835.
Micropuncture studies in the rat. Circ Res 1980; 46: 440448. 74. Wynn TA. Fibrosis under arrest. Nat Med 2010; 16: 523525.
63. Yang L, Besschetnova TY, Brooks CR et al. Epithelial cell cycle arrest in G2/ 75. Bechtel W, McGoohan S, Zeisberg EM et al. Methylation determines fibroblast
M mediates kidney fibrosis after injury. Nat Med 2010; 16: 535543 531p activation and fibrogenesis in the kidney. Nat Med 2010; 16: 544550.
following 143. 76. Megyesi J, Udvarhelyi N, Safirstein RL et al. The p53-independent
64. Basile DP, Friedrich JL, Spahic J et al. Impaired endothelial proliferation activation of transcription of p21 WAF1/CIP1/SDI1 after acute renal
and mesenchymal transition contribute to vascular rarefaction failure. Am J Physiol 1996; 271: F1211F1216.
following acute kidney injury. Am J Physiol Renal Physiol 2011; 300: 77. Srisawat N, Murugan R, Lee M et al. Plasma neutrophil gelatinase-
F721F733. associated lipocalin predicts recovery from acute kidney injury following
65. Basile DP, Donohoe D, Roethe K et al. Renal ischemic injury results in community-acquired pneumonia. Kidney Int 2011; 80: 545552.
permanent damage to peritubular capillaries and influences long-term 78. Lassnigg A, Schmidlin D, Mouhieddine M et al. Minimal changes
function. Am J Physiol Renal Physiol 2001; 281: F887F899. of serum creatinine predict prognosis in patients after cardiothoracic
66. Yuan HT, Li XZ, Pitera JE et al. Peritubular capillary loss after mouse acute surgery: a prospective cohort study. J Am Soc Nephrol 2004; 15:
nephrotoxicity correlates with down-regulation of vascular endothelial 15971605.
growth factor-A and hypoxia-inducible factor-1 alpha. Am J Pathol 2003; 79. Chertow GM, Burdick E, Honour M et al. Acute kidney injury, mortality,
163: 22892301. length of stay, and costs in hospitalized patients. J Am Soc Nephrol 2005;
67. O0 Riordan E, Mendelev N, Patschan S et al. Chronic NOS inhibition 16: 33653370.
actuates endothelial-mesenchymal transformation. Am J Physiol Heart 80. Anderson S, Eldadah B, Halter JB et al. Acute kidney injury in older adults.
Circ Physiol 2007; 292: H285H294. J Am Soc Nephrol 2011; 22: 2838.
68. Horbelt M, Lee SY, Mang HE et al. Acute and chronic microvascular 81. Go AS, Parikh CR, Ikizler TA et al. The assessment, serial evaluation, and
alterations in a mouse model of ischemic acute kidney injury. Am J Physiol subsequent sequelae of acute kidney injury (ASSESS-AKI) study: design
Renal Physiol 2007; 293: F688F695. and methods. BMC Nephrol 2010; 11: 22.
69. Bechtel W, McGoohan S, Zeisberg EM et al. Methylation determines 82. USRDS Annual Report 2007 Department of Health and Human Services,
fibroblast activation and fibrogenesis in the kidney. Nat Med 2010; 16: NIDDK, United States Renal Data System (USRDS) (NIH publication no
544550. 07-3176) 2007; 1: 240241.

524 Kidney International (2012) 82, 516524


Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

Você também pode gostar