Sunteți pe pagina 1din 13

REVIEWS

Mechanisms of maladaptive repair after AKI


leading to accelerated kidney ageing and CKD
David A. Ferenbach and Joseph V. Bonventre
Abstract | Acute kidney injury is an increasingly common complication of hospital admission and is associated
with high levels of morbidity and mortality. A hypotensive, septic, or toxic insult can initiate a cascade of
events, resulting in impaired microcirculation, activation of inflammatory pathways and tubular cell injury
or death. These processes ultimately result in acutely impaired kidney function and initiation of a repair
response. This Review explores the various mechanisms responsible for the initiation and propagation of
acute kidney injury, the prototypic mechanisms by which a substantially damaged kidney can regenerate its
normal architecture, and how the adaptive processes of repair can become maladaptive. These mechanisms,
which include G2/M cell-cycle arrest, cell senescence, profibrogenic cytokine production, and activation of
pericytes and interstitial myofibroblasts, contribute to the development of progressive fibrotic kidney disease.
The end result is a state that mimics accelerated kidney ageing. These mechanisms present important
opportunities for the design of targeted therapeutic strategies to promote adaptive renal recovery and
minimize progressive fibrosis and chronic kidney disease after acute insults.
Ferenbach, D.A & Bonventre, J.V. Nat. Rev. Nephrol. advance online publication 3 February 2015; doi:10.1038/nrneph.2015.3

Introduction
Despite the advent of dialysis in the second half of the is baseline hypoxia even under normal conditions.7
20th century as a treatment for severe acute kidney injury Current treatment for AKI is supportive in nature, and
(AKI), the mortality associated with this condition trials of agents showing promise in experimental IRI
remains unacceptably high, especially in the intensive models (for example diuretics and dopamine) have failed
care unit population (>50%),13 with a paucity of effec- to ameliorate clinical AKI in translational studies.8,9
tive therapeutic interventions. The incidence of AKI has Although the high initial mortality associated with AKI
been steadily increasing related, in part, to the ageing is well recognized,13 for many years it was accepted that
of the population;4 the increasing prevalence of chronic normal kidney structure and function would return in
kidney disease (CKD), which predisposes to AKI;5 and the survivors of AKI. An increasing number of epidemiologi-
increasing number of invasive interventions that can result cal studies with both adequate statistical power and length
in haemodynamic compromise or septic complications. of follow-up1014 have, however, revealed that survivors of
Furthermore, contrast agents required for imaging studies AKI exhibit a persistently increased risk of progressive
and an increasing number of therapeutic agents in the CKD, proteinuria and an excess risk of cardiovascular
pharmacological armamentarium have varying degrees mortality. This finding complements results in labora-
of nephrotoxicity, which can precipitate or worsen AKI.4 tory animals demonstrating that renal injury produces
In many cases, progression of kidney failure is not a senescence-associated profibrotic secretory phenotype
due to worsening of primary renal disease, but rather a and a subsequent inflammatory milieu, which promotes
secondary insult, most commonly associated with tran- the gradual accumulation of renal fibrosis, vascular
sient intrarenal regional or generalized hypoperfusion rarefaction and CKD.1517 This Review summarizes our
or sepsis. Ischaemiareperfusion injury (IRI) and activa- emerging knowledge of the factors underlying both adap-
Renal Division
tion of inflammatory pathways initiate diverse processes tive kidney repair and the maladaptive repair linking
andDivision of resulting in acute tubular injury or necrosis, particularly AKI to CKD, and what therapeutic opportunities they
BiomedicalEngineering, in the outer stripe of the outer medulla6 where there present. Because of length constraints only a portion of
Department of
Medicine, Brigham the relevant data are included.
andWomens Hospital,
Harvard Medical Competing interests
School, 75 Francis J.V.B. is co-inventor on KIM1 patents that are assigned to
Adaptive repair after AKI
Street, Boston, Partners HealthCare and licensed to Johnson & Johnson, An acute renal insult affects the function of several dis-
MA02115, USA
Sekisui, Novartis, Biogen Idec., R & D, and Astute. He is a tinct cell populations within the kidney, which contrib-
(D.A.F., J.V.B.).
consultant for Astellas, Novartis, Roche and Sekisui regarding utes to the initiation and amplification of the kidney
the safety and efficacy of therapeutics or diagnostics for acute
Correspondence to: injury. These various cell types will be discussed along
J.V.B. kidney injury. He holds equity in MediBeacon, Sentien and
joseph_bonventre@ Thrasos, and has grant support from Novo Nordisk and Roche. with their potential relevance for the reparative phase
hms.harvard.edu D.A.F. declares no competing interests. of renal recovery. Although clinical AKI is associated

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 1


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points
Despite the high level of functional loss often seen in
patients with AKI, it is known that in humans the func-
Acute injury to the kidney is often associated with maladaptive repair and
tional loss can be transient. The kidney has the ability to
incomplete resolution, leading to residual abnormalities in kidney structure
andfunction
return to normal function following an insult (Figure1),
Increasing age, and chronic low-grade insults to the tubular epithelium although there is evidence from experimental models
increase epithelial cell sensitivity to episodes of acute kidney injury, leading and in humans that complete functional recovery is less
tomaladaptive repair and progression of chronic kidney disease likely with ageing.11,18 It must be recognized that func-
Maladaptive repair is characterized by fibrosis, vascular rarefaction, tubular tional recovery is usually assessed by measuring levels of
loss, glomerulosclerosis and the presence of a chronic inflammatory infiltrate serum creatinine, which is an insensitive tool.
within the kidney Whether tubular regeneration after injury arises from
Injured renal tubular epithelial cells become arrested at G2/M and adopt a
proliferation of surviving mature cells or from renal stem
profibrotic phenotype, which affects other epithelial cells, pericytes and the
immune system
cells has been debated.19,20 Long-lived label-retaining
Myofibroblasts that likely arise from renal pericytes proliferate and contribute tubular cells have been reported to be found within the
to the deposition of extracellular matrix and resulting fibrosis within the renal papilla21 and bear surface markers associated with
injuredkidney stem cell properties.22,23 However, a study from our lab
Maladaptive repair after acute kidney insults shares many common features oratory using genetic fate-mapping of renal tubular epi-
with kidney ageing and can be thought of as a state of accelerated kidney thelial cells provided strong evidence against renal stem
ageing cells being the main source of new cells after renalinjury,
even after repeated IRI. 24 This study confirms prior
with high morbidity and mortality, kidney biopsy is hypotheses19 that mature surviving cells dedifferentiate
seldom performed. In addition, when a biopsy is avail- and proliferate to replace those tubular epithelial cells
able it often does not sample the outer medulla where that have been lost,25 with no tubular cell subpopulation
a considerable component of the pathology may reside. (for example, a stem cell population), preferentially par-
This paucity of outer medullary tissue, together with the taking in the repair process in response to insults that
fact that the biopsy is often performed during the recov- injure the proximal tubule.25,26
ery phase rather than the injury phase, likely explains
why the injury to the tubules seen on biopsy may be Endothelial injury and oxygen delivery
less than one would expect from the functional impair- An important approach to facilitating recovery and
ment of the kidney. The presence of casts, tubular cells minimizing long-term sequelae of injury is to minimize
and high levels of kidney injury molecule1 (KIM1) the injury itself. After an initial insult there is an exten-
in the urine confirm the presence of severe proximal sion phase during which it might be possible to inter-
tubule injury. vene after recognizing the injury and hence change the

Tissue repair

Healthy Injured Repaired


kidney Tubular injury kidney Macrophage M1 to M2 switching kidney

Endothelial cell activation


Resolution of inflammatory infiltrate

Tubular obstruction

Tubular proliferation
Leucocyte recruitment

Endothelial repair
Vascular injury and regeneration

Scarred
kidney

Impaired repair and fibrosis

Figure 1 | A summary of some of the mechanisms involved in initial tissue injury and subsequent repair of the kidney after
Nature Reviews | Nephrology
acute kidney injury. Maladaptive and incomplete repair leads to the development of fibrosis and, ultimately, chronic kidney
disease.

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Normal structure AKI damage and dysfunction and initiate a repair process that
Tubular Injured, adhesive can be adaptive (Figure2) or maladaptive (Figure3).
epithelial Endothelial cell endothelium
cell Necrotic Both arterial pressure and segmental vascular resistance
Apoptotic
tubular cell tubular cell will influence kidney oxygen delivery and it is increasingly
recognized that alterations in tissue hypoxia can vary
substantially in different regions of the injured kidney.28
Factors such as sodium reabsorption, nitric oxide release,
angiotensin II generation, tissue haemoglobin concentra-
tion and oxygen saturation have been shown in humans
or experimental models to influencethe balance between
oxygen delivery and consumption in the injured organ27
Capillary
Pericyte with the potential to modulate hypoxic injury. In addi-
Reduced M1
Resident vascular macrophage Neutrophil tion to these factors, altered production of prostaglandins
macrophage NO release recruitment recruitment and reactive oxygen species due to injury of neighbour-
ing tubules27,29 further impair tissue oxygen delivery,
leading to a local no-reflow phenomenon where the
occluded microvasculature further amplifies initial
Repair Resolution injury. Administration or generation of agents that act
Tubular cell
Full tubular Intact on the vasculature, such as adenosine, in experimental
proliferation
repair vasculature animals have shown promise in promoting early vascu-
lar patency and hence minimizing resultant hypoxia in
experimentalIRI.30,31
Studies in rats demonstrate that following acute IRI,
vascular function remains impaired for several days.32
During this period there is impaired autoregulation
ofblood flow, potentially reflecting in part the failureof
endothelial nitric oxide synthase to generate nitric
oxide.33 Injured endothelial cells and changes to the
Macrophage integrity of the glycocalyx of the peritubular capillar-
Pericytes phagocytosis
remain and M1 to M2 Resolution of ies can result in exposure of a range of surface markers
in situ switching inflammatory infiltrate
that promote the recruitment and adhesion of leuco-
Figure 2 | The evolution of tissue injury, death and subsequent adaptive| Nephrology
Nature Reviews repair cytes34,35 and platelets, which can interfere with perfu-
after AKI. Following an episode of AKI, the kidney is capable of repair back to sion and contribute to additional endothelial cell injury
normal or near-normal structure and function despite apparently severe damage.
and inflammation. Vascular permeability is increased,
Initial injury is characterized by endothelial activation, recruitment of myeloid
leucocytes and widespread tubular cell injury and death. In adaptive repair, over
leading to interstitial tissue swelling, further compromis-
aperiod of days, debris is cleared by tubular cells and recruited macrophages, and ing microvascular perfusion.29 The availability ofintra-
epithelial dedifferentiation occurs followed by proliferation to restore the integrity vital microscopy has enabled the visualization of the
of the tubular epithelial cell layer. Macrophages support renal growth and post-ischaemic tissue, revealing that blood flow in cor-
recoveryby adopting an M2 phenotype before leaving the kidney. Pericytes remain tical peritubular capillaries becomes sluggish and may
in contact with the capillary network and do not give rise to new myofibroblasts or even reverse in the first 30min after injury.36,37
if they do, this myofibroblast proliferation is reversible. Abbreviations: AKI, acute Following apparently recovered IRI in the rat, there is
kidney injury; NO, nitric oxide.
a long-term reduction in peritubular capillary density,17
which precedes the development of visible fibrosis. Renal
long-term outcome by targeting the injury phase rather hypoxia is a common feature of diverse chronic renal inju-
than the recovery phase. Strategies to improve long-term ries, and is likely to contribute to the progression of renal
outcomes after AKI should therefore be based on a thor- failure.37 Early tubular regeneration after IRI is associ
ough understanding of the injury phase as well as the ated with high levels of transforming growth factor
recovery phase. Endothelial cell injury and dysfunction (TGF), but reduced vascular endothelial growth factor
may have an important role in the extension phase. (VEGF), suggesting that factors involved in proliferation
In the clinical setting, initiating insults include, among of the tubular compartment impede repair or contribute
a large list, shock, sepsis, surgery and administration of to ongoing loss in the microvascular compartment, in
contrast and other nephrotoxic agents. Many of these which the endothelial cells show little early proliferation
insults converge on a common ischaemiareperfusion after injury.38 Studies examining the response to kidney
pathway with localized or generalized renal hypoxia.6,27 injury have implicated the pericyte, which is an important
The resultant inflammation and ischaemic changes mediator of vascular stability in embryogenesis, as a key
affect tissue oxygenation, the local generation of nitric contributor to vascular homeostasis.38,39 Animals with
oxide and a number of profibrotic and proinflammatory defective pericytes that lack the matrix metalloproteinase
cytokines and growth factors, and the subsequent pro- inhibitor TIMP3 spontaneously develop a microvascular
duction of deleterious reactive oxygen species. Each of phenotype in the kidney,40 with both vascular rarefaction
these factors, as well as others, further potentiate tissue and increased fibrosis.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 3


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Healthy kidney AKI DNA damage CKD


Increasing age
Previous AKI/CKD
Sustained cell stress

G2/M cycle arrest Progressive


fibrosis

Maladaptive repair Progressive scarring


Injured, adhesive endothelium Pericyte/capillary Persisting M1 Chronic
dissociation macrophages Myofibroblast inflammation
Apoptotic Necrotic
tubular cell tubular cell

Tubular loss
Collagen
M1 macrophage Neutrophil Pericyte proliferation G2/M-arrested deposition Secretion of profibrotic
recruitment recruitment and differentiation tubular cells factors by G2/M cells

Figure 3 | Maladaptive repair of AKI leads to CKD. Studies have highlighted the importance of G2/MNaturecell-cycle
Reviews |arrest in
Nephrology
response to severe, repeated and genotoxic renal insults. In the initial repair phase after injury, cells may become
arrested in G2/M phase and release cytokines and growth factors that promote inflammatory cell retention within the
kidney and ongoing inflammation. Injury and pro-inflammatory stimuli lead to pericyte dissociation from the endothelium,
resulting in microvascular rarefaction and the progressive deposition of collagen I by myofibroblasts arising from activated
pericytes. Ageing sensitizes tubular cells to G2/M arrest in response to cell stress and DNA damage, providing a potential
explanation for the increased risk of CKD progression after AKI in the elderly. Abbreviations: AKI, acute kidney injury; CKD,
chronic kidney disease.

The cellular immune response Monocytes and macrophages


IRI results in a rapid immune response and the recruit- The early influx of neutrophils is followed by mono-
ment of leucocytes to the injured kidney. These cells cyte recruitment, predominantly of the Ly6Chi subset.43
may have dual roles: potentiating early injury, but also Macrophages exhibit phagocytic properties for both dying
being necessary for the subsequent successful resolu- tubular cells and neutrophils.52 Studies where macro
tion of damage.41 Kidneys contain an extensive network phages were depleted before AKI have demonstrated
of resident mononuclear phagocytes that mount an protection from injury or a net neutral effect depend-
early response to injury by releasing a systemic burst of ing on the approach used.5355 It is generally agreed that
tumour necrosis factor.42 The combination of chemotac- recruited macrophages amplify initial injury. While initi
tic signals from resident immune cells, injured tubular ally of the Ly6Chi M1 inflammatory subset, macrophages
cells and the altered characteristics of injured endothelial persist in the kidney but the population progressively
cells promotes vascular adhesion and transmigration of loses Ly6C and adopts M2 polarization over the course
neutrophils within the first 24h after injury.43 of the first 5days,41 after which their numbers decline.43
Macrophages of the Ly6Clo M2 subtype are believed to be
Neutrophils beneficial in the repair of the kidney after injury.
The presence and accumulation of interstitial and mar- Macrophages play key roles in supporting kidney
ginating neutrophils in very early experimental IRI is growth during nephrogenesis,56 and undertake the vital
well established,44,45 with one study demonstrating that removal of cellular debris and dying neutrophils after renal
cytokines, such as IL17, that are released from neutro- injury.57 After adopting an M2 phenotype in the recovery
phils contribute to downstream immune activation.46 phase of AKI,41 macrophages secrete compounds, includ-
Whether their presence within the acutely injured ing growth factors, fibronectin, Wnt-7b and IL1 receptor
kidney actively promotes injury remains controversial, antagonist, all of which support epithelial proliferation and
with various approaches to inhibit or deplete neutrophil repair.57,58 While pre-injury depletion of macrophages is
function demonstrating either protection44,47,48 or lack often protective, depletion of macrophages from mice
of protection4951 depending on the technique employed with established AKI results in the absence of M2-secreted
and the species studied. products and prolongation of renal injury.59 Furthermore,

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

polarization towards a proinflammatory M1 phenotype Administration of these agents resulted in reduced


resulted in failure to resolve the injury. This finding pro- leucocyte accumulation and blocked Toll-like-receptor-
vides further evidence for the importance of macrophages mediated macrophage activation. Administration of
in facilitating eventual renal repair.41,58,60 a lipoxin A4 analogue invivo downregulated inflam-
matorycytokine expression while upregulating anti-
Lymphocytes inflammatory cytokine expression, potentially via an
Neither B lymphocytes nor T lymphocytes are present effect on interactions between resident cells and recruited
in large numbers in the kidney before or after AKI;43 leucocytes.75 Hepatocyte growth factor (HGF) has also
however, studies of mouse models do support their been evaluated for potential beneficial effects to protect
influence on the evolution of renal injury. 61 Bcell- against injury. Overexpression of HGF in kidneys was
deficient (MT) and CD4/CD8-deficient mice are both associated with reduced injury, leucocyte recruitment
protected from AKI, whereas Rag1-knockout mice and TGF1 expression after ischaemic injury, and treat-
demonstrate no alteration in susceptibility to injury.6264 ment of epithelial cells invitro with HGF was associated
Inanother study using Rag1-deficient mice, protection with reduced production of CC motif chemokine 2, sup-
from injury was restored by adoptive transfer of either porting a key role for the epithelium in the recruitment of
Bcells or Tcells alone, but not by both,49 suggesting that myeloid inflammatorycells.7577
complex and poorly understood interactions between Serum complement is an evolutionarily conserved
lymphocyte populations contribute to the injury pheno- activator of the innate immune response. Widespread
type. Regulatory Tcells positive for FoxP3 and CD25 tubular deposition of complement has been documented
have been reported to limit tissue injury.65 Mice with a in the first few hours following IRI,78 and, in the rodent,
depleted regulatory Tcell population and those lacking is associated with a loss of the complement inhibitor Crry
regulatory Tcells exhibit heightened levels of immune from the tubular basolateral surface.79 Insitu activation
activation and tissue damage after IRI.65 of the complement system may also be important, where
The role of lymphocytes in mediating tissue repair production of C3 by renal dendritic cells promotes their
remains incompletely understood. Work using MT subsequent inflammatory activation and ability to acti-
mice demonstrated that chimeric animals had a more vate T lymphocytes.80 Unlike other solid organs, comple-
rapid recovery than did control mice after experimen- ment deposition in the kidney is documented to occur
tal IRI, suggesting that Bcells themselves inhibit tissue via the alternative pathway,78 activating in response to
repair.66 Other work reveals that lymphocytes of animals hydrolysis of C3 rather than the presence of antigen
previously exposed to severe IRI can induce albumin antibody complexes. Selective inhibition of this alterna-
uria after transfer to naive recipients, indicating that tive pathway protects against experimental ischaemic
immunological memory may contribute to the develop- injury.81 Evidence also exists for the importance of the
ment of proteinuria after AKI67 and this proteinuria may terminal phase of complement deposition (formation
have secondary effects to facilitate the onset of CKD. of the C5b9 complex) in mediating the severity of
ischaemic AKI.82
Circulating and endogenous compounds
A number of circulating and endogenous compounds Tubular cell injury and death
have been implicated in both tissue protection and Tubular cells in the S3 segment of the proximal tubule are
exacerbation of AKI. Many immune cells express the characterized by high metabolic activity with low baseline
A2A receptor for circulating adenosine, which down- ambient oxygen delivery from the capillary network.7 In
regulates inflammatory activation and tissue injury in the context of AKI they can experience profound hypoxia
multiple organs, including the kidney.68 In addition to and subsequent rebound reoxygenation, followed by
beneficial effects of local adenosine production on the further fluctuations owing to capillary dysfunction and
survival and function of the endothelium,69 macrophages the recruitment of activated and cytotoxic leucocytes.
and dendritic cells have been shown to be sensitive to These multiple insults result in a loss of physical cellcell
local adenosine production,55,70 with regulatory Tcells interactions, and tubular cell death with apoptosis and
implicated as a local producer of adenosine, exerting prominent programmed and unprogrammed necrosis.83,84
anti-inflammatory effects.71 Cell death, combined with the detachment and loss of
The endogenous anti-inflammatory compound haem viable epithelial cells into the tubular lumen, leads to the
oxygenase1 (HO1) is induced in response to hypoxia denudation of areas of the S3 segment. Cell death can
and implicated as a cellular protector against AKI. extend to earlier segments of the proximal tubule also,
Several groups have demonstrated the protective effects and results in a failure of tubular function, structural
of HO1 induction before experimental AKI,54,55,72 which obstruction of flow, and the distal delivery of a solute-
seemed to be mediated via expression in macrophages rich fluid activating tubuloglomerular feedback, which
rather than tubular epithelial cells,72 where it promotes further inhibits glomerular filtration.
an anti-inflammatory M2 phenotype.73 Studies using surface markers have lent weight to the
Resolvins and protectin D1 are natural anti- importance of modification of the epithelial cell pheno
inflammatory compounds capable of reducing experimen- type to assume more mesenchymal characteristics
tal AKI and subsequent fibrosis,74 and exhibit protective during both acute and chronic kidney injury and repair.85
effects with both pre-injury and early post-injury dosing. Anumber of groups have demonstrated that surviving

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 5


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

tubular cells may transiently upregulate markers such as isolated tubular epithelial injury.16 It has been shown that
vimentin, smooth muscle actin and S100A4, which cJun Nterminal kinase (JNK) remains active for weeks
are often associated with the fibroblast lineage but not after injury to tubular epithelial cells.95 Our laboratory
indicative of transformation to fibroblasts.86,87 The coex- demonstrated that, in multiple models of AKI, arrest of
pression of these markers with proliferating cell nuclear tubular cells in G2/M resulted in JNK activation, with
antigen suggests that the dedifferentiated cells are subsequent fibrosis which was reduced by JNK inhib
undergoing active replication.87,88 ition.15 These findings are compatible with the hypothesis
One pathway of documented importance in the that progressive nephron loss and failed redifferentiation
repair process after injury is Wnt/-catenin signalling. with ageing may facilitate maladaptive repair after AKI.96
Transient IRI has been shown to induce a rapid induc-
tion of Wnt4 that returns to baseline by 24h.89 Factors Epigenetic changes after AKI
present in AKI, including disruption of cellcell junc- The role of epigenetic changes in the kidney follow-
tions, lead to nuclear translocation of catenin.89 This ing AKI is a new and rapidly developing field. There
finding echoes embryogenesis, where expression of are diverse mechanisms by which modifications to the
Wnt contributes to maintenance of the undifferentiated epigenome can alter the subsequent expression of pro-
embryonic state.90 The dedifferentiated cells respond to inflammatory and anti-inflammatory genes.97,98 Studies
various proliferative cues, including HGF, epidermal of patients with CKD have already implicated DNA
growth factor, TGF and VEGF, 85 with a restricted methylation and histone modifications as contributors
burst of TGF expression, potentially maximizing a to the progression of the fibrotic process.98 AKI has been
therapeutic rather than profibrotic outcome. With sub- associated with alterations in DNA methylation and
sequent increases in cell number and resultant increased histone modification,99,100 leading to altered transcription
cellcell contacts, the Wnt signalling pathway is sup- of genes implicated in renal injury, including TNF and
pressed. Studies of tubulogenesis invitro suggest that CC motif chemokine2. Advances in epigenetic assess-
sequential Wnt downregulation and expression of matrix ment technology in the kidney are aiding the search for
metalloproteinase is necessary forthe recovery ofa dif- changes after AKI.101 These approaches have the poten-
ferentiated phenotype,91,92 hinting at the importance of tial to reveal mechanisms by which adaptively repaired
cellmatrix interactions in achieving and maintaining kidneys may undergo long-lasting t ranscriptional
the mature state. changes that predispose to CKD.

Mechanisms of CKD development post AKI KIM1 expression and kidney fibrosis
As stated previously, it is now widely accepted that epi- KIM1 is an important biomarker for renal injury, with
sodes of AKI predispose to the development of CKD multiple log-order increases in expression and shedding
even in those patients judged to have normal pre-injury from injured tubular cells after renal insults in rodents and
renal function based on measurement of serum creatin humans.102,103 KIM1 promotes the clearance of apoptotic
ine levels.10 Several emerging theories to explain the cells by tubular epithelia,104 a finding of importance for
difference between adaptive (without scarring) and endogenous kidney repair after injury. With prolonged
maladaptive repair leading to CKD are discussed below. epithelial cell expression of KIM1, however, our recent
work has demonstrated a profibrotic phenotype associ-
Recurrent tubular injury promotes scarring ated with increased leucocyte recruitment to the injured
Advances in molecular biology have enabled the gen- kidney. Thus KIM1 may provide a mechanistic link
eration of transgenic animals susceptible to the selective between injury and fibrosis.105 While acute expression of
injury of particular cell types such as the renal tubular epi- KIM1 is adaptive and facilitates repair,106 chronic expres-
thelial cell.16 The generation of transgenic mice express- sion is maladaptive and promotes fibrosis.104,105 Taken
ing the simian diphtheria toxin receptor on the tubular together, these data support the hypothesis that a focused
epithelia has enabled interrogation of the role of specific and repeated tubular injury can be sufficient to initiate
injury to this cell type without associated concurrent progressive scarring.16
injury to surrounding capillaries, pericytes and other
nearby cells. Interestingly, a single dose of diphtheria toxin Origins of myofibroblasts in renal fibrosis
was followed by a vigorous inflammatory response and A key feature of maladaptive repair after AKI is the
subsequent complete repair. By contrast, three doses of appearance of increased numbers of myofibroblasts
diphtheria toxin over 3weeks resulted in persistent inflam- which contribute to the deposition of collagen and other
mation, microvascular rarefaction, increased expression components of the fibrotic matrix in the kidney.107 An
of TGF1, collagen1(I) and fibronectin, progressive understanding of the origin of these cells is therefore an
fibrogenesis, and secondaryglomerulosclerosis.16 important step towards the development of antifibrotic
Multiple sublethal injuries could lead to premature cell therapies. Although it is accepted that stellate cells in the
senescence and a failure to respond to subsequent injury liver act as pericytes and mediate collagen deposition
with adaptive proliferation. In addition, repeated insults after injury,108 in the kidney the source of the interstitial
may lead to tubular cells remaining arrested in a dedif- fibroblasts responsible for the deposition of scar tissue
ferentiated state with ongoing production of profibrotic is a topic of ongoing debate. Candidates include circu-
factors,93,94 which contribute to microvasular loss after lating progenitors (fibrocytes), epithelialmesenchymal

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Altered growth factor expression epithelia involving Six2-cre (to mark cells that are
descendents of the metanephric mesenchyme), HoxB7-
Chronic KIM-1 expression cre (to mark cells that descend from the ureteric bud), or
FoxD1-cre-positive mesenchymal cells (which are des-
Chronic inflammatory infiltrate
tined to primarily form the kidney interstitial fibroblasts,
Microvascular rarefaction vascular smooth muscle cells, pericytes of peritubular
capillaries and mesangial cells) has enabled fate tracing
Myofibroblast proliferation of cells after renal injury.24,39 These studies demonstrated
Maladaptive
repair post-AKI Tubular loss
Ageing no evidence for transdifferentiation of epithelial cells to
fibroblasts and pointed to the pericyte as an important
Senescent tubular epithelia precursor of myofibroblasts.39 Importantly, while renal
epithelia can be readily induced to express mesenchymal
Secretion of profibrotic factors
by G2/M-arrested cells markers invitro, expression of these markers by epithelial
cells invivo should be interpreted as a reflection of dedif
Interstitial collagen deposition
ferentiation rather than conversion to fibroblasts.19,87
Glomerulosclerosis Conversion of epithelial cells to fibroblasts may therefore
be primarily an invitro phenomenon.
Figure 4 | Common mechanisms in kidney ageing and progressive kidney injury.
Nature Reviews
Features of kidney ageing include tubular loss, glomerulosclerosis, | Nephrology
microvascular Renal pericytes
rarefaction and deposition of interstitial collagen. The number of senescent and
Pericytes are specialized cells present in close proximity
G2/M arrested cells present in the ageing kidney also progressively increases. All
to the endothelial cells of multiple organs.118 Although
the above cellular changes are also seen in progressive renal disease in younger
patients following acute renal insults. These shared features suggest that they were first noted in electron microscopic studies of
progressive chronic kidney disease is functionally equivalent to accelerated ageing the kidney in the early 1980s, over the past decade there
of the kidney. Abbreviation: AKI, acute kidney injury. has been a rapid increase in research interest in the role
of the pericyte in embryogenesis and tissue homeostasis,
and its potential role in tissue fibrosis and vascular rare
transdifferentiation (EMT) and, more recently, renal faction after renal injury.38 Pericytes maintain vascular
pericytes or perivascular fibroblasts as the cells giving stability via cellcell communication and released factors
rise to pathogenic myofibroblasts in injury. including PDGF,119 angiopoietin,120 TGF,121 VEGF122
and sphingosine1-phosphate.123
Fibrocytes It has been proposed that the renal pericyte has
Fibrocytes are circulating cells of myeloid lineage that are a central role in the development of fibrosis after
proposed to be precursors for the fibroblasts responsible AKI 39,117,124 by migrating from its perivascular loca-
for tissue scarring in fibrotic disease of various organs tion and acquiring the markers and phenotype of the
including the kidney.109111 The contribution of these myofibroblast. The propensity of the injury-associated
cells to the direct deposition of matrix within scarred activated pericyte to break contact with endothelial
tissues has been challenged, however, both historically cells and migrate may deliver a double hit of vascular
by the lack of bone-marrow-derived cells in a para instability and collagen deposition, which are both hall-
biotic rat model of skin scarring,112 and more recently by marks of progressive CKD (Figure3). Studies demon-
fate-mapping techniques.113,114 Although bone-marrow- strate reduced vascular stability with loss of TIMP3 and
derived cells do not seem to be major contributors tothe ADAM-TS1, which is associated with worsened injury.40
direct transdifferentiation to myofibroblasts within As such, the pericyte is now the subject of active research
thekidney, similar to macrophages, these cells may aimed at targeting these cells to inhibit fibrosis and pre-
function in a paracrine fashion to support or promote serving the vascular network. Blockade of the PDGF
renal scarring by interacting with other kidney cells or receptor and VEGF receptor2 protects against fibro-
by producing factors that other cells respond to, thereby sis and vascular rarefaction.117 Recently Kramann et al.
contributing to the fibrotic response. identified a perivascular Gli-1(+) population of cells as
a major cellular origin of fibrosis.125
EMT
The role of EMT as the source of myofibroblasts and Ageing and kidney repair after injury
subsequent fibrosis after renal injury has generated The incidence of AKI increases with each decade of
much interest.115 While there is abundant documenta- life.4,126 Patients with in-hospital AKI are often elderly;
tion of expression of mesenchymal markers when epi- in one study of Medicare recipients the mean age was
thelial cells are injured or exposed to certain cytokines approximately 75years.4 The increased comorbidities
invitro, genetic lineage tracing studies supporting and associated polypharmacy might account for some
the existence of EMT invivo in the kidney have been of the increased risk of AKI associated with increasing
much more limited.116 Most studies have challenged age; however, there are likely to be underlying poorly
the conclusion that epithelial cells transdifferentiate to understood biological changes that contribute to sus-
myofibroblasts. Fluorescence labelling of cells express- ceptibility. Recent work has sought to define the under-
ing collagen1(I)117 or use of genetic crosses in renal lying processes and cellular responses that represent

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 7


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Normal ageing Oxidative stress DNA damage Mitochondrial injury Senescence of epithelial cells with ageing
With increasing age evidence exists for a reduced prolifer-
Stress-induced ative capacity within the kidney in the aftermath of injury
Replicative
senescence
premature senescence invivo and invitro.138 Microarray studies of ageing rat
Cellular senescence kidneys identified age-related alterations in multiple gene
networks at baseline.139 Multiple-fold induction of injury
biomarkers, such as KIM1, occurs in ageing animals,
Growth p16INK4a + p21WAF1 Cytokine Growth factor
arrest expression secretion secretion which is reflective of ongoing age-related kidney injury at
baseline. As discussed above, persistent KIM1 expression
may be maladaptive and facilitate chronic scarring, which
Recruitment of Promotion of Increased sensitivity Increase in can be exacerbated after denovo renal insults.
inflammatory cells further senescence to further injury fibrosis Increased rates of cellular senescence with ageing are
Figure 5 | Replicative and stress-induced premature senescence in kidney injury. associated with a state of growth arrest that can be trig-
Renal cells become senescent either through ageing and Nature Reviews
telomere | Nephrology
shortening, or gered by a critical shortening in telomere length or by
via genotoxic insults resulting in stress-induced premature senescence. While in stressors such as hypoxia or genotoxic insults.139 Senescent
terminal growth arrest, these senescent cells remain metabolically active and cells are resistant to apoptosis and, rather than being inert,
secrete a range of factors that contribute to the chronic inflammatory state, the they can secrete numerous cytokines and chemokines,
progression of renal fibrosis and increased susceptibility of other cells to
including (C-X-C motif) ligand 1 (CXCL1), IL6 and IL8,
subsequent insults and senescence. Modified with permission of American Society
of Nephrology, from Yang, H. & Fogo, A.B. J. Am. Soc. Nephrol. 21, 14362439
which all promote maintenance of a chronic inflammatory
(2010); permission conveyed through Copyright Clearance Center, Inc. state (Figure5).140 Senescence-associated galactosidase
has been used as a marker for senescence.141 Senescence
is associated with increased expression of p16INK4a and
the hallmarks of ageing,127 implicating factors such as p21WAF1, which contribute to growth arrest. Aged animals
genomic instability, loss of proteostasis and increas- have increased levels of p21WAF1 at baseline and levels are
ing cellular senescence as key contributors to the aged further increased in stress-induced premature senes-
phenotype. Disease-induced cellular senescence has cence, leading to proliferative arrest and apoptosis.142
been shown within the kidney and other organs.128130 Surprisingly, studies of p21-deficient mice exposed to IRI
Increased levels of cellular senescence have been showed a worsened injury phenotype.143 Increased levels
reported in patients with hypertension,131 in tubular of p16INK4a inhibit CDK4 and CDK6, leading to stabili-
cells of patients with IgA nephropathy 132 and in type2 zation of retinoblastoma-associated protein in its phos-
diabetes,133 where they correlate with severity of renal phorylated form, causing growth arrest. Pharmacological
disease. Our own data demonstrate growth-arrested blockade of both CDK4 and CDK6 in young animals pro-
tubular cellsto be predictive of the development of CKD tected against AKI, despite a transient reduction in cel-
subsequent to AKI.15 Registry data demonstrating the lular proliferation.144 Thus, particularly in young animals,
susceptibility of the CKD population and healthy elderly there may potentially be deleterious effects of excessive
individuals to AKI and progressive kidney disease10,13,14 proliferation in the immediate aftermath of IRI; however,
further point to shared mechanisms between ageing a persistent decrease in proliferative capacity may lead toa
and susceptibility to progressive kidney disease that is chronic profibrotic phenotype.
associated with maladaptive repair post AKI (Figure4). In contrast to what is seen in younger mice, reduced pro-
liferation was noted in the recovery phase after IRI in older
The ageing immune system mice, together with upregulation of zinc-2-glycoprotein
Immunosenescence refers to alterations in the func- (Azgp1); however, attempts to increase cell-cycle turnover
tion of the ageing immune system. With ageing there by knocking down Azgp1 resulted in markedly worse
is low-grade immune system activation, and a skewing fibrosis in recovery.138 Hence, while increasing levels of
towards increased myeloid cell responses together with senescent epithelial cells with either ageing or injury pro-
reduced function of Tlymphocytes.134 An aged human motes sensitivity to further injury and progressive fibro-
kidney attracts an increased quantity of inflammatory sis, it is important to select therapeutic targets to increase
infiltrates when transplanted into a young or old recipi- proliferation without a similar increase in fibrosis. It is
ent.135 While it has been suggested that this finding rep- possible, for example, that interventions that increase
resents the effects of increased expression of PECAM1 movement through the G0/G1 phase of the cell cycle
in the donor kidney,136 it remains plausible that this result without increasing movement through G2/M may then
is related to age-related increased cellular senescence. increase the number of cells trapped in G2/Mand enhance
Epithelial cell senescence in the aged kidney allograft the generation of pro-fibrotic cytokines.
may limit the ability of the kidney to mount an adequate Whether the above factors are fixed and intrinsic to the
adaptive repair response to the long-term immunological aged kidney itself or are influenced and hence modifiable
attack. As discussed previously, macrophage phenotypic by circulating factors remains unknown. Elegant studies
switching facilitates renal repair after AKI.41 This pheno- in other organs, such as the aged brain, skeletal muscle
typic change is impaired in macrophages in other organs and heart, using heterochronic parabiosis to create a
of the ageing rodent,137 but remains less well explored shared circulation, have demonstrated the modification
inthekidney. of aged injury phenotypes when mice are exposed to a

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

Renal tubular
INK CDK effects have been attributed to paracrine actions of bone-
CTGF epithelial cells
TGF-
inhibitors marrow-derived cells, either acting systemically or locally,
e.g. p16INK4a
IL-8 CDK6 CDK4 as some of these cells enter the kidney interstitium after
CDK1 Cyclin D KIP/CIP CDK injury.160 Importantly, although the number of MSCs in the
Cyclin B inhibitors
M e.g. p21WAF1/CIP1 blood of aged animals remain constant, it is reported that
G1 CDK2
Cyclin E
their secretion of TGF, bone morphogenetic protein2
and bone morphogenetic protein4 are reduced, while
G2/M arrest G2/M G1/S
checkpoint checkpoint secretion of proinflammatory IL6 is increased. Numbers
DNA damage of circulating endothelial progenitor cells are reduced with
Cell stress advancing age, as is their mobilization and incorporation
into the injured endothelium.161 Ageing is also associated
CDK1 G2 S CDK2 with the progressive accumulation of genomic damage,
Cyclin A S checkpoint Cyclin A which impacts negatively on stem cell function and likely
dedifferentiated epithelial cell function.162 These changes
contribute to a less reparative phenotype,163 indeed one
that may actually impair wound healing.164
Figure 6 | Eukaryotic cell-cycle checkpoints. Four phases of cell-cycle
Nature Reviewsprogression
| Nephrology
are seen in eukaryotic cells. Cellular DNA is replicated in the S phase, and cell
mitosis occurs in M phase, separated by two gap phases, G1 and G2. Critical
Maladaptive repair and G2/M arrest
checkpoints exist at G1/S and G2/M, where the actions of cyclins, CDKs and their Maladaptive repair leading to CKD is characterized by
inhibitors determine whether appropriate cell size, DNA replication and integrity persistent parenchymal inflammation, with increased
exist to allow the initiation of DNA synthesis or the completion of cell division, numbers of myofibroblasts and accumulation of extra-
respectively. Increased numbers of G2/M-arrested cells have been identified as a cellular matrix. 16 Risk factors for the maladaptive
common feature in models of progressive chronic kidney disease. Abbreviations: repair response include the type and duration of injury,
CDK, cyclin-dependent kinase; CTGF, connective tissue growth factor; TGF, thepre-injury glomerular filtration rate and the age ofthe
transforming growth factor.
patient.14 With recent studies failing to demonstrate con-
vincing evidence for epithelial-to-fibroblast transdifferen-
young circulation,145147 or by systemic depletion of senes- tiation to account for progressive fibrosis,24,39,117 increasing
cent cells.148 A study examining the effect of transplant- interest has focused on alterations in the injured tubular
ing young and old bone marrow into aged mice showed epithelium as the potential cause of ongoing activation
reduced levels of fibrosis and markers of senescence in and proliferation of pericytes, which may represent the
the resident renal cells of the recipients of young bone important precursor population for myofibroblasts.15 As
marrow.149 This observation raises the possibility of mod- discussed above, contrary to their name, senescent cells
ulating the levels of epithelial cell senescence by altering can remain metabolically active and adopt a senescence-
the systemic milieu to delay or prevent age-facilitated associated secretory phenotype, which is associated
renaldisease. with the release of connective tissue growth factor and
TGF, both contributors to chronic inflammation, col-
Growth factor production with ageing lagen deposition and vascular rarefaction.165,166 Senescent
In response to tubular injury, epithelial cells produce a cells are also documented to produce IL8, which may
number of growth factors, including epidermal growth potentiate cells entering G2/M arrest.167
factor, HGF, and insulin-like growth factor1.150 There Checkpoints in the cell cycle involving cyclins, cyclin-
is evidence that levels of renal epidermal growth factor dependent kinases and cyclin-dependent kinase inhibitors
decrease with increasing age,151 with similar reductions ensure the arrest of cell division in the presence of inad-
seen in VEGF expression, coupled with increased levels equate cell size, DNA damage or cell stress (Figure6).142
of thrombospondin1.152 An age-associated increase in Recent work in our laboratory supports G2/M arrest in
levels of TGF has also been shown.153 TGF- is potently tubular cells as an important driver of maladaptive repair
profibrotic with sustained release. These changes in and progressive CKD after AKI (Figure6).15,168,169 We
growth-factor expression could contribute to the increased examined multiple experimental models of murine kidney
susceptibility to tissue injury, fibrosis and vascular injury, including severe bilateral IRI, unilateral IRI, aristo
rarefaction seen in the elderly.154 lochic acid nephropathy and unilateral ureteral obstruc-
tion, and identified the accumulation of cells in G2/M
Circulating progenitor cells in ageing growth arrest as the common feature predicting progres-
The role of circulating stem cells, progenitor cells or sive fibrotic kidney disease.15 G2/M arrest in tubular epi-
marrow stromal cells (MSCs) in mediating repair after thelial cells in response to injury results in the acquisition
AKI is controversial. An initial report suggesting that of a pathogenic phenotype characterized by the sustained
MSCs were incorporated into the tubules of the repair- expression of profibrotic growth factors. This hypothesis
ing kidney 155 was followed by a study from our laboratory is supported by reports from other laboratories168170 and
that demonstrated no evidence of incorporation of non- by pharmacological inhibition of G2/M-arrested cells,
renal cells into the tubules of the repaired kidney.156 Several which reduced fibrosis, whereas increases in the propor-
studies have, however, reported partial protection against tion of G2/M-arrested cells in the cell cycle exacerbated
kidney injury with the administration of MSCs.157159 These fibrosis.2,168170 Notably, humans with the FAN1 mutation

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 9


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

and defects in DNA repair develop karyomegalic intersti- Clinical application in humans will, however, need careful
tial nephritis, with evidence of increased levels of DNA assessment of the safety of the drugs and pathways under
damage and cell-cycle arrest in the late G2 phase.171 As investigation given the function of G2/M in preventing the
ageing is associated with increased levels of DNA damage perpetuation of dangerous DNA mutations. Recent work
and cell senescence,172 predisposition to G2/M arrest pro- in rhabdomyosarcoma indicates that bypassing cell-cycle
vides another plausible mechanism to explain the height- checkpoints promotes tumourigenesis.176 Other strategies
ened risk of fibrotic kidney disease complicating AKI in could focus on either blocking the profibrotic signalling of
the elderly, which is likely to be related to a combination G2/M-arrested cells (as has been performed successfully
of factors as described in this Review. in rodents using JNK-pathway inhibition15), or promoting
their apoptosis or pharmacological depletion. Notably, in
Conclusions mice with a progeroid background of accelerated ageing,
Work published over the past few years has revealed new sustained depletion of senescent cells expressing p16INK4a
targets for antifibrotic therapies such as G2/M arrested significantly delayed the onset of age-related disorders
cells, immunosenescence, and modulation of growth in multiple tissues.148 This finding suggests that senes-
factors and inflammatory mediators. The role of EMT cent cells actively promote the disease phenotype rather
versus pericytes (or other interstitial cell populations) than simply representing a marker of an ongoing process.
in driving renal fibrosis has been re-evaluated. Targeting Another approach would be to target perivascular gli1(+)
perivascular cells may address both the deposition progenitors.125 Translation of these strategies to humans
offibrosis and microvascular rarefaction characteristic of will require the identification of novel markers permitting
progressive CKD after AKI.173 Various compounds that the specific targeting of these cells.
modulate pericyte function are being investigated to assess The contribution of ageing to immuosenescence and to
their ability to promote kidney recovery.174 the pool of senescent tubular cells within the kidney may
With recognition of the importance of abnormalities in explain the increased susceptibility to AKI displayed by
the epithelial cell cycle in determining the renal outcome the elderly as well as the age-influenced increased rates of
after injury comes the potential to target this process CKD progression. Progressive CKD may be mechanisti-
using novel therapeutic strategies. Experiments in rodents cally synonymous with accelerated ageing of the kidney.
suggest the therapeutic potential of blocking the initiation Given the ageing population and increasing burden of
of the G2/M checkpoint, or encouraging cells to transit age-related progressive CKD in both developed and devel-
through G2/M to complete mitosis, using agents such as oping nations, novel therapies for this pervasive disease
histone deacetylase inhibitors170 or p53 inhibition.15,175 deserve the highest priority.

1. Metcalfe, W. et al. Acute renal failure requiring a systematic review and meta-analysis. 20. Cantley, L.G. Adult stem cells in the repair of the
renal replacement therapy: incidence and KidneyInt. 81, 442448 (2012). injured renal tubule. Nat. Clin. Pract. Nephrol. 1,
outcome. QJM 95, 579583 (2002). 11. Ishani, A. et al. Acute kidney injury increases risk 2232 (2005).
2. Ali, T. et al. Incidence and outcomes in acute of ESRD among elderly. J. Am. Soc. Nephrol. 20, 21. Oliver, J.A., Maarouf, O., Cheema, F.H.,
kidney injury: A comprehensive population-based 223228 (2009). Martens,T.P. & Al-Awqati, Q. The renal papilla is
study. J. Am. Soc. Nephrol. 18, 12921298 12. Shimoi, T. et al. The significant impact of acute a niche for adult kidney stem cells. J. Clin. Invest.
(2007). kidney injury on CKD in patients who survived 114, 795804 (2004).
3. Noble, J.S., Simpson, K. & Allison, M.E. over 10years after myeloablative allogeneic 22. Bussolati, B, et al. Isolation of renal progenitor
Longterm quality of life and hospital mortality in SCT. Bone Marrow Transplant. 48, 8084 cells from adult human kidney. Am. J. Pathol. 166,
patients treated with intermittent or continuous (2013). 545555 (2005).
hemodialysis for acute renal and respiratory 13. Belayev, L.Y. & Palevsky, P.M. The link between 23. Dekel, B. et al. Isolation and characterization
failure. Ren. Fail. 28, 323330 (2006). acute kidney injury and chronic kidney disease. ofnontubular sca1+lin- multipotent stem/
4. Xue, J.L. et al. Incidence and mortality of acute Curr. Opin. Nephrol. Hypertens. (2013). progenitor cells from adult mouse kidney. J. Am.
renal failure in medicare beneficiaries, 1992 14. Chawla, L.S. & Kimmel, P.L. Acute kidney injury Soc. Nephrol. 17, 33003314 (2006).
to2001. J. Am. Soc. Nephrol. 17, 11351142 and chronic kidney disease: an integrated 24. Humphreys, B.D. et al. Intrinsic epithelial cells
(2006). clinical syndrome. Kidney Int. 82, 516524 repair the kidney after injury. Cell Stem Cell 2,
5. Wonnacott, A., Meran, S., Amphlett, B., (2012). 284291 (2008).
Talabani,B. & Phillips, A. Epidemiology and 15. Yang, L., Besschetnova, T.Y., Brooks, C.R., 25. Humphreys, B.D. et al. Repair of injured proximal
outcomes in community-acquired versus Shah, J.V. & Bonventre, J.V. Epithelial cell cycle tubule does not involve specialized progenitors.
hospital-acquired aki. Clin. J. Am. Soc. Nephrol. arrest in G2/M mediates kidney fibrosis after Proc. Natl Acad. Sci. USA 108, 92269231
9,10071014 (2014). injury. Nat. Med. 16, 535543 (2010). (2011).
6. Thadhani, R., Pascual, M. & Bonventre, J.V. 16. Grgic, I. et al. Targeted proximal tubule 26. Berger, K. & Moeller, M.J. Mechanisms of
Acute renal failure. N. Engl. J. Med. 334, injurytriggers interstitial fibrosis and epithelial repair and regeneration after acute
14481460 (1996). glomerulosclerosis. Kidney Int. 82, 172183 kidney injury. Semin. Nephrol. 34, 394403
7. Brezis, M. & Rosen, S. Hypoxia of the renal (2012). (2014).
medullaits implications for disease. N. Engl. J. 17. Basile, D.P., Donohoe, D., Roethe, K. 27. Evans, R.G. et al. Haemodynamic influences
Med. 332, 647655 (1995). &Osborn,J.L. Renal ischemic injury results onkidney oxygenation: clinical implications of
8. Friedrich, J.O., Adhikari, N., Herridge, M.S. inpermanent damage to peritubular capillaries integrative physiology. Clin. Exp. Pharmacol.
&Beyene, J. Meta-analysis: low-dose dopamine and influences long-term function. Am. J. Physiol. Physiol. 40, 106122 (2013).
increases urine output but does not prevent Renal Physiol. 281, F887F899 (2001). 28. Pallone, T.L., Robertson, C.R. & Jamison, R.L.
renal dysfunction or death. Ann. Intern. Med. 18. Schmitt, R. & Cantley, L.G. The impact of aging Renal medullary microcirculation. Physiol. Rev.
142, 510524 (2005). on kidney repair. Am. J. Physiol. Renal Physiol. 70, 885920 (1990).
9. Ho, K.M. & Sheridan, D.J. Meta-analysis of 294, F1265F1272 (2008). 29. Basile, D.P. The endothelial cell in ischemic acute
frusemide to prevent or treat acute renal failure. 19. Bonventre, J.V. Dedifferentiation and kidney injury: Implications for acute and chronic
BMJ 333, 420 (2006). proliferation of surviving epithelial cells in acute function. Kidney Int. 72, 151156 (2007).
10. Coca, S.G., Singanamala, S. & Parikh, C.R. renal failure. J. Am. Soc. Nephrol. 14 (Suppl. 1), 30. Kim, M. et al. The volatile anesthetic isoflurane
Chronic kidney disease after acute kidney injury: S55S61 (2003). induces ecto5'-nucleotidase (cd73) to protect

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

against renal ischemia and reperfusion injury. 51. Thornton, M.A., Winn, R., Alpers, C.E. 70. Li, L. et al. Dendritic cells tolerized with
Kidney Int. 84, 90103 (2013). &Zager,R.A. An evaluation of the neutrophil as adenosine A2 AR agonist attenuate acute kidney
31. Venkatachalam, M.A. & Weinberg, J.M. The a mediator of invivo renal ischemic-reperfusion injury. J. Clin. Invest. 122, 39313942 (2012).
conundrum of protection from AKI by adenosine injury. Am. J. Pathol. 135, 509515 (1989). 71. Kinsey, G.R. et al. Autocrine adenosine signaling
in rodent clamp ischemia models. Kidney Int. 84, 52. Savill, J., Smith, J., Sarraf, C., Ren, Y., Abbott, F. promotes regulatory T cell-mediated renal
1619 (2013). & Rees, A. Glomerular mesangial cells and protection. J. Am. Soc. Nephrol. 23, 15281537
32. Conger, J.D., Robinette, J.B. & Hammond, W.S. inflammatory macrophages ingest neutrophils (2012).
Differences in vascular reactivity in models of undergoing apoptosis. Kidney Int. 42, 924936 72. Ferenbach, D.A. et al. The induction of
ischemic acute renal failure. Kidney Int. 39, (1992). macrophage hemeoxygenase1 is protective
10871097 (1991). 53. Ferenbach, D.A. et al. Macrophage/monocyte during acute kidney injury in aging mice.
33. Noiri, E. et al. Oxidative and nitrosative stress depletion by clodronate, but not diphtheria toxin, KidneyInt. 79, 966976 (2011).
inacute renal ischemia. Am. J. Physiol. Renal improves renal ischemia/reperfusion injury in 73. Ferenbach, D.A. et al. Macrophages expressing
Physiol. 281, F948F957 (2001). mice. Kidney Int. 82, 928933 (2012). heme oxygenase1 improve renal function in
34. De Greef, K.E., Ysebaert, D.K., Persy, V., 54. Jo, S.K., Sung, S.A., Cho, W.Y., Go, K.J. & ischemia/reperfusion injury. Mol. Ther. 18,
Vercauteren, S.R. & De Broe, M.E. ICAM1 Kim,H.K. Macrophages contribute to the 17061713 (2010).
expression and leukocyte accumulation in initiation of ischaemic acute renal failure in rats. 74. Duffield, J.S. et al. Resolvin D series and
innerstripe of outer medulla in early phase Nephrol. Dial. Transplant. 21, 12311239 protectin D1 mitigate acute kidney injury.
ofischemic compared to hgCl2-induced ARF. (2006). J.Immunol. 177, 59025911 (2006).
Kidney Int. 63, 16971707 (2003). 55. Day, Y.J., Huang, L., Ye, H., Linden, J. 75. Leonard, M.O. et al. 15epi16-(para-
35. Kelly, K.J. et al. Minocycline inhibits apoptosis &Okusa,M.D. Renal ischemia-reperfusion fluorophenoxy)-lipoxin a(4)-methyl ester, a
and inflammation in a rat model of ischemic injury and adenosine 2a receptor-mediated synthetic analogue of 15epilipoxin A(4), is
renal injury. Am. J. Physiol. Renal Physiol. 287, tissue protection: Role of macrophages. Am. J. protective in experimental ischemic acute renal
F760F766 (2004). Physiol. Renal Physiol. 288, F722F731 (2005). failure. J. Am. Soc. Nephrol. 13, 16571662
36. Brodsky, S.V. et al. Endothelial dysfunction 56. Rae, F. et al. Characterisation and trophic (2002).
inischemic acute renal failure: rescue by functions of murine embryonic macrophages 76. Franquesa, M. et al. Tubular epithelial cells
transplanted endothelial cells. Am. J. Physiol. based upon the use of a csf1r-egfp transgene transfected with hHGF counteracts monocyte
Renal Physiol. 282, F1140F1149 (2002). reporter. Dev. Biol. 308, 232246 (2007). chemotactic protein1 up-regulation after
37. Yamamoto, T. et al. Intravital videomicroscopy of 57. Ricardo S.D., van Goor, H. & Eddy, A.A. hypoxia/reoxygenation insult. Transplant. Proc.
peritubular capillaries in renal ischemia. Am. J. Macrophage diversity in renal injury and repair. 41, 20692072 (2009).
Physiol. Renal Physiol. 282, F1150F1155 J.Clin. Invest. 118, 35223530 (2008). 77. Franquesa, M. et al. Direct electrotransfer of
(2002). 58. Lin, S.L. et al. Macrophage wnt7b is critical for hHGF gene into kidney ameliorates ischemic
38. Schrimpf, C. & Duffield, J.S. Mechanisms of kidney repair and regeneration. Proc. Natl Acad. acute renal failure. Gene Ther. 12, 15511558
fibrosis: the role of the pericyte. Curr. Opin. Sci. USA 107, 41944199 (2010). (2005).
Nephrol. Hypertens. 20, 297305 (2011). 59. Jang, H.S., Kim, J., Park, Y.K. & Park, K.M. 78. Thurman, J.M., Lucia, M.S., Ljubanovic, D.
39. Humphreys, B.D. et al. Fate tracing reveals Infiltrated macrophages contribute to recovery &Holers, V.M. Acute tubular necrosis is
thepericyte and not epithelial origin of after ischemic injury but not to ischemic characterized by activation of the alternative
myofibroblasts in kidney fibrosis. Am. J. Pathol. preconditioning in kidneys. Transplantation 85, pathway of complement. Kidney Int. 67, 524530
176, 8597 (2010). 447455 (2008). (2005).
40. Schrimpf, C. et al. Pericyte TIMP3 and ADAMTS1 60. Lech, M. et al. Macrophage phenotype controls 79. Thurman, J.M. et al. Altered renal tubular
modulate vascular stability after kidney injury. long-term AKI outcomeskidney regeneration expression of the complement inhibitor Crry
J.Am. Soc. Nephrol. 23, 868883 (2012). versus atrophy. J. Am. Soc. Nephrol. 25, 292304 permits complement activation after ischemia/
41. Lee, S. et al. Distinct macrophage phenotypes (2013). reperfusion. J. Clin. Invest. 116, 357368 (2006).
contribute to kidney injury and repair. J. Am. Soc. 61. Linfert, D., Chowdhry, T. & Rabb, H. Lymphocytes 80. Peng, Q., Li, K., Patel, H., Sacks, S.H. & Zhou,W.
Nephrol. 22, 317326 (2011). and ischemia-reperfusion injury. Transplant. Rev. Dendritic cell synthesis of C3 is required for
42. Dong, X. et al. Resident dendritic cells are 23, 110 (2009). fullTcell activation and development of a th1
thepredominant TNF-secreting cell in early 62. Burne-Taney, M.J. et al. Bcell deficiency confers phenotype. J. Immunol. 176, 33303341 (2006).
renalischemia-reperfusion injury. Kidney Int. 71, protection from renal ischemia reperfusion 81. Thurman, J.M. et al. Treatment with an inhibitory
619628 (2007). injury. J. Immunol. 171, 32103215 (2003). monoclonal antibody to mouse factor b protects
43. Ysebaert, D.K. et al. Identification and kinetics 63. Rabb, H. et al. Pathophysiological role of mice from induction of apoptosis and renal
of leukocytes after severe ischaemia/ Tlymphocytes in renal ischemia-reperfusion ischemia/reperfusion injury. J. Am. Soc. Nephrol.
reperfusion renal injury. Nephrol. Dial. Transplant. injury in mice. Am. J. Physiol. Renal Physiol. 27, 17, 707715 (2006).
15, 15621574 (2000). F525F531 (2000). 82. Renner, B. et al. The complement inhibitors
44. Kelly, K.J. et al. Intercellular adhesion 64. Park, P. et al. Injury in renal ischemiareperfusion Crryand factor H are critical for preventing
molecule1deficient mice are protected is independent from immunoglobulins and autologous complement activation on renal
againstischemic renal injury. J. Clin. Invest. 97, Tlymphocytes. Am. J. Physiol. Renal Physiol. 282, tubular epithelial cells. J. Immunol. 185,
10561063 (1996). F352F357 (2002). 30863094 (2010).
45. Bolisetty, S. & Agarwal, A. Neutrophils in acute 65. Kinsey, G.R. et al. Regulatory T cells suppress 83. Linkermann, A. et al. Two independent pathways
kidney injury: not neutral any more. Kidney Int. innate immunity in kidney ischemia-reperfusion of regulated necrosis mediate ischemia-
75, 674676 (2009). injury. J. Am. Soc. Nephrol. 20, 17441753 reperfusion injury. Proc. Natl Acad. Sci. USA 110,
46. Li, L. et al. IL17 produced by neutrophils (2009). 1202412029 (2013).
regulates IFNmediated neutrophil migration 66. Jang, H.R. et al. Bcells limit repair after 84. Linkermann, A., De Zen, F., Weinberg, J.,
inmouse kidney ischemia-reperfusion injury. ischemic acute kidney injury. J. Am. Soc. Nephrol. Kunzendorf, U. & Krautwald, S. Programmed
J.Clin. Invest. 120, 331342 (2010). 21, 654665 (2010). necrosis in acute kidney injury. Nephrol. Dial.
47. Chaturvedi, S. et al. Slit2 prevents neutrophil 67. Burne-Taney, M.J. et al. Transfer of lymphocytes Transplant. 27, 34123419 (2012).
recruitment and renal ischemia-reperfusion injury. from mice with renal ischemia can induce 85. Ishibe, S. & Cantley, L.G. Epithelial
J. Am. Soc. Nephrol. 24, 12741287 (2013). albuminuria in naive mice: A possible mesenchymalepithelial cycling in kidney
48. Awad, A.S. et al. Compartmentalization of mechanism linking early injury and progressive repair.Curr. Opin. Nephrol. Hypertens. 17,
neutrophils in the kidney and lung following renal disease? Am. J. Physiol. Renal Physiol. 291, 379385 (2008).
acute ischemic kidney injury. Kidney Int. 75, F981F986 (2006). 86. Villanueva, S., Cespedes, C. & Vio, C.P.
689698 (2009). 68. Day, Y.J. et al. Renal protection from ischemia Ischemicacute renal failure induces the
49. Burne-Taney, M.J., Yokota-Ikeda, N. & Rabb, H. mediated by a2a adenosine receptors on expression of a wide range of nephrogenic
Effects of combined T and Bcell deficiency bonemarrow-derived cells. J. Clin. Invest. 112, proteins. Am. J. Physiol. Regul. Integr. Comp.
onmurine ischemia reperfusion injury. Am. J. 883891 (2003). Physiol. 290, R861R870 (2006).
Transplant. 5, 11861193 (2005). 69. Basile, D.P. et al. Impaired endothelial 87. Witzgall, R., Brown, D., Schwarz, C.
50. Melnikov, V.Y. et al. Neutrophil-independent proliferation and mesenchymal transition &Bonventre,J.V. Localization of proliferating
mechanisms of caspase1 and IL18mediated contribute to vascular rarefaction following acute cell nuclear antigen, vimentin, cfos, and
ischemic acute tubular necrosis in mice. J. Clin. kidney injury. Am. J. Physiol. Renal Physiol. 300, clusterin in the postischemic kidney. Evidence
Invest. 110, 10831091 (2002). F721F733 (2011). for a heterogenous genetic response among

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 11


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

nephron segments, and a large pool of 105. Humphreys, B.D. et al. Chronic epithelial kidney 124. Chen, Y.T. et al. Platelet-derived growth factor
mitotically active and dedifferentiated cells. injury molecule1 expression causes murine receptor signaling activates pericyte
J.Clin. Invest. 93, 21752188 (1994). kidney fibrosis. J. Clin. Invest. 123, 40234035 myofibroblast transition in obstructive and post-
88. Witzgall, R., OLeary, E., Gessner, R., (2013). ischemic kidney fibrosis. Kidney Int. 80, 1170
Ouellette,A.J. & Bonventre, J.V. Kid1, a 106. Yang, L. et al. KIM-1/TIM-1 mediated- 1181 (2011).
putative renal transcription factor: Regulation phagocytosis reduces acute injury to the kidney. 125. Kramann, R. et al. Perivascular gli1(+)
during ontogeny and in response to ischemia J. Clin. Invest. (in press). progenitors are key contributors to injury-induced
and toxic injury. Mol. Cell. Biol. 13, 19331942 107. Bonventre, J.V. & Yang, L. Cellular organ fibrosis. Cell Stem Cell 16, 5166 (2015).
(1993). pathophysiology of ischemic acute kidney injury. 126. Leung, K.C., Tonelli, M. & James, M.T. Chronic
89. Price, V.R., Reed, C.A., Lieberthal, W. & J. Clin. Invest. 121, 42104221 (2011). kidney disease following acute kidney injury-risk
Schwartz, J.H. ATP depletion of tubular cells 108. Mederacke, I. et al. Fate tracing reveals hepatic and outcomes. Nat. Rev. Nephrol. 9, 7785
causes dissociation of the zonula adherens and stellate cells as dominant contributors to liver (2013).
nuclear translocation of -catenin and LEF1. fibrosis independent of its aetiology. Nature 127. Lopez-Otin, C., Blasco, M.A., Partridge, L.,
J.Am. Soc. Nephrol. 13, 11521161 (2002). Commun. 4, 2823 (2013). Serrano, M. & Kroemer, G. The hallmarks of
90. Kuure, S., Popsueva, A., Jakobson, M., Sainio, K. 109. Williams, S.M. et al. Class I HDACs regulate aging. Cell 153, 11941217 (2013).
& Sariola, H. Glycogen synthase kinase3 angiotensin ii-dependent cardiac fibrosis via 128. Clements, M.E., Chaber, C.J., Ledbetter, S.R.
inactivation and stabilization of -catenin induce fibroblasts and circulating fibrocytes. J. Mol. Cell. &Zuk, A. Increased cellular senescence and
nephron differentiation in isolated mouse and Cardiol. 67, 112125 (2014). vascular rarefaction exacerbate the progression
rat kidney mesenchymes. J. Am. Soc. Nephrol. 110. Sahebally, S.M. et al. Circulating fibrocytes and of kidney fibrosis in aged mice following transient
18, 11301139 (2007). Crohns disease. Br. J. Surg. 100, 15491556 ischemic injury. PLoS ONE 8, e70464 (2013).
91. Ishibe, S., Haydu, J.E., Togawa, A., Marlier, A. (2013). 129. Tabibian, J.H., OHara, S.P., Splinter, P.L.,
&Cantley, L.G. Cell confluence regulates 111. Wada, T. et al. Involvement of bonemarrow- Trussoni, C.E. & Larusso, N.F. Cholangiocyte
hepatocyte growth factor-stimulated cell derived cells in kidney fibrosis. Clin. Exp. senescence via Nras activation is a
morphogenesis in a catenindependent Nephrology. 15, 813 (2011). characteristic of primary sclerosing cholangitis.
manner. Mol. Cell. Biol. 26, 92329243 (2006). 112. Ross, R., Everett, N.B. & Tyler, R. Wound healing Hepatology 59, 22632275 (2014).
92. OBrien, L.E. et al. ERK and MMPs sequentially and collagen formation. VI. The origin of the 130. Marongiu, F., Serra, M.P., Sini, M., Angius, F. &
regulate distinct stages of epithelial tubule wound fibroblast studied in parabiosis. J. Cell Laconi, E. Clearance of senescent hepatocytes
development. Develop. Cell 7, 2132 (2004). Biol. 44, 645654 (1970). in a neoplastic-prone microenvironment delays
93. Kimura, M. et al. Role of atrophic changes in 113. Dulauroy, S., Di Carlo, S.E., Langa, F., Eberl, G. the emergence of hepatocellular carcinoma.
proximal tubular cells in the peritubular deposition &Peduto, L. Lineage tracing and genetic ablation Aging 6, 2634 (2014).
of type IV collagen in a rat renal ablation model. of adam12(+) perivascular cells identifya major 131. Westhoff, J.H. et al. Hypertension induces
Nephrol. Dial. Transplant. 20, 15591565 (2005). source of profibrotic cells during acute tissue somatic cellular senescence in rats and humans
94. Suzuki, T., Kimura, M., Asano, M., Fujigaki, Y. injury. Nat. Med. 18, 12621270 (2012). by induction of cell cycle inhibitor p16ink4a.
&Hishida, A. Role of atrophic tubules in 114. Lin, S.L., Kisseleva, T., Brenner, D.A. & Hypertension 52, 123129 (2008).
development of interstitial fibrosis in Duffield,J.S. Pericytes and perivascular 132. Liu, J. et al. Accelerated senescence of renal
microembolism-induced renal failure in rat. fibroblasts are the primary source of collagen- tubular epithelial cells is associated with
Am.J. Pathol. 158, 7585 (2001). producing cells in obstructive fibrosis of the disease progression of patients with
95. de Borst, M.H. et al. CJun NH2-terminal kinase kidney. Am. J. Pathol. 173, 16171627 (2008). immunoglobulin A (IgA) nephropathy. Transl. Res.
is crucially involved in renal tubulo-interstitial 115. Zeisberg, M. & Duffield, J.S. Resolved: EMT 159, 454463 (2012).
inflammation. J. Pharmacol. Exp. Ther. 331, produces fibroblasts in the kidney. J. Am. Soc. 133. Verzola, D. et al. Accelerated senescence in
896905 (2009). Nephrol. 21, 12471253 (2010). thekidneys of patients with type2 diabetic
96. Venkatachalam, M.A. et al. Acute kidney injury: 116. Iwano, M. et al. Evidence that fibroblasts derive nephropathy. Am. J. Physiol. Renal Physiol. 295,
a springboard for progression in chronic kidney from epithelium during tissue fibrosis. J. Clin. F1563F1573 (2008).
disease. Am. J. Physiol. Renal Physiol. 298, Invest. 110, 341350 (2002). 134. Franceschi, C. et al. Inflamm-aging. An
F1078F1094 (2010). 117. Lin, S.L. et al. Targeting endotheliumpericyte evolutionary perspective on immunosenescence.
97. Bomsztyk, K. & Denisenko, O. Epigenetic cross talk by inhibiting VEGF receptor signaling Ann. NY Acad. Sci. 908, 244254 (2000).
alterations in acute kidney injury. Semin. Nephrol. attenuates kidney microvascular rarefaction and 135. Oien, C.M. et al. Living donor kidney
33, 327340 (2013). fibrosis. Am. J. Pathol. 178, 911923 (2011). transplantation: the effects of donor age and
98. Wing, M.R., Ramezani, A., Gill, H.S., 118. Smith, S.W., Chand, S. & Savage, C.O. Biology gender on short- and long-term outcomes.
Devaney,J.M. & Raj, D.S. Epigenetics of of the renal pericyte. Nephrol. Dial. Transplant. Transplantation 83, 600606 (2007).
progression of chronic kidney disease: fact or 27, 21492155 (2012). 136. Reutzel-Selke, A. et al. Donor age intensifies the
fantasy? Semin. Nephrol. 33, 363374 (2013). 119. Betsholtz, C. Insight into the physiological early immune response after transplantation.
99. Naito, M., Zager, R.A. & Bomsztyk, K. BRG1 functions of PDGF through genetic studies in Kidney Int. 71, 629636 (2007).
increases transcription of proinflammatory mice. Cytokine Growth Factor Rev. 15, 215228 137. Kelly, J., Ali Khan, A., Yin, J., Ferguson, T.A. &
genes in renal ischemia. J. Am. Soc. Nephrol. 20, (2004). Apte, R.S. Senescence regulates macrophage
17871796 (2009). 120. Sundberg, C., Kowanetz, M., Brown, L.F., activation and angiogenic fate at sites of tissue
100. Zager, R.A. & Johnson, A.C. Renal ischemia- Detmar,M. & Dvorak, H.F. Stable expression injury in mice. J. Clin. Invest. 117, 34213426
reperfusion injury upregulates histone-modifying ofangiopoietin1 and other markers by cultured (2007).
enzyme systems and alters histone expression at pericytes: phenotypic similarities to a 138. Schmitt, R., Marlier, A. & Cantley, L.G.
proinflammatory/profibrotic genes. Am. J. Physiol. subpopulation of cells in maturing vessels Zagexpression during aging suppresses
Renal Physiol. 296, F1032F1041 (2009). during later stages of angiogenesis invivo. proliferation after kidney injury. J. Am. Soc.
101. Yu, J., Feng, Q., Ruan, Y., Komers, R., Kiviat, N. Lab.Invest. 82, 387401 (2002). Nephrol. 19, 23752383 (2008).
&Bomsztyk, K. Microplate-based platform for 121. Carvalho, R.L. et al. Defective paracrine 139. Kwekel, J.C., Desai, V.G., Moland, C.L., Vijay, V.
combined chromatin and DNA methylation signalling by TGF in yolk sac vasculature of & Fuscoe, J.C. Life cycle analysis of kidney gene
immunoprecipitation assays. BMC Mol. Biol. 12, endoglin mutant mice: A paradigm for hereditary expression in male f344 rats. PLoS ONE 8,
49 (2011). haemorrhagic telangiectasia. Development 131, e75305 (2013).
102. Bonventre, J.V. Kidney injury molecule1 (Kim1): 62376247 (2004). 140. Kuilman, T., Michaloglou, C., Mooi, W.J.
a urinary biomarker and much more. Nephrol. 122. Benjamin, L.E., Hemo, I. & Keshet, E. &Peeper, D.S. The essence of senescence.
Dial. Transplant. 24, 32653268 (2009). Aplasticitywindow for blood vessel remodelling Genes Dev. 24, 24632479 (2010).
103. Ichimura, T. et al. Kidney injury molecule1 is defined by pericyte coverage of the preformed 141. Itahana, K., Campisi, J. & Dimri, G.P. Methods
(Kim1), a putative epithelial cell adhesion endothelial network and is regulated by PDGFb todetect biomarkers of cellular senescence:
molecule containing a novel immunoglobulin and VEGF. Development 125, 15911598 thesenescence-associated -galactosidase
domain, is up-regulated in renal cells after injury. (1998). assay. Methods Mol. Biol. 371, 2131 (2007).
J. Biol. Chem. 273, 41354142 (1998). 123. Chae, S.S., Paik, J.H., Allende, M.L., 142. Yang, H. & Fogo, A.B. Cell senescence in
104. Ichimura, T. et al. Kidney injury molecule1 is Proia,R.L. & Hla, T. Regulation of limb theaging kidney. J. Am. Soc. Nephrol. 21,
aphosphatidylserine receptor that confers a development by the sphingosine 1phosphate 14361439 (2010).
phagocytic phenotype on epithelial cells. J. Clin. receptor S1p1/EDG1 occurs via the hypoxia/ 143. Megyesi, J., Andrade, L., Vieira, J.M. Jr,
Invest. 118, 16571668 (2008). VEGF axis. Dev. Biol. 268, 441447 (2004). Safirstein, R.L. & Price, P.M. Positive effect

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrneph


2015 Macmillan Publishers Limited. All rights reserved
REVIEWS

ofthe induction of p21WAF1/CIP1 on the 156. Duffield, J.S. et al. Restoration of tubular 167. Acosta, J.C. et al. Chemokine signaling via the
courseofischemic acute renal failure. Kidney epithelial cells during repair of the postischemic CXCR2 receptor reinforces senescence. Cell
Int. 60, 21642172 (2001). kidney occurs independently of bone marrow- 133, 10061018 (2008).
144. Dirocco, D. et al. CDK4/6 inhibition induces derived stem cells. J. Clin. Invest. 115, 17431755 168. Wu, C.F. et al. Transforming growth factor 1
epithelial cell cycle arrest and ameliorates acute (2005). stimulates profibrotic epithelial signaling to
kidney injury. Am. J. Physiol. Renal Physiol. 306, 157. Morigi, M. et al. Mesenchymal stem cells are activate pericyte-myofibroblast transition in
F379F388 (2013). renotropic, helping to repair the kidney and obstructive kidney fibrosis. Am. J. Pathol. 182,
145. Conboy, I.M. et al. Rejuvenation of aged improve function in acute renal failure. J. Am. 118131 (2013).
progenitor cells by exposure to a young systemic Soc. Nephrol. 15, 17941804 (2004). 169. Tang, J. et al. Sustained activation of EGFR
environment. Nature 433, 760764 (2005). 158. Semedo, P. et al. Early modulation of inflammation triggers renal fibrogenesis after acute kidney
146. Loffredo, F.S. et al. Growth differentiation factor by mesenchymal stem cell after acute kidney injury. Am. J. Pathol. 183, 160172 (2013).
11 is a circulating factor that reverses age-related injury. Int. Immunopharmacol. 9, 677682 (2009). 170. Cianciolo Cosentino, C. et al. Histone
cardiac hypertrophy. Cell 153, 828839 (2013). 159. Hagiwara, M., Shen, B., Chao, L. & Chao, J. deacetylase inhibitor enhances recovery
147. Villeda, S.A. et al. The ageing systemic milieu Kallikrein-modified mesenchymal stem cell afterAKI. J. Am. Soc. Nephrol. 24, 943953
negatively regulates neurogenesis and cognitive implantation provides enhanced protection (2013).
function. Nature 477, 9094 (2011). against acute ischemic kidney injury by inhibiting 171. Zhou, W. et al. FAN1 mutations cause
148. Baker, D.J et al. Clearance of p16(Ink4a)-positive apoptosis and inflammation. Hum. Gene Ther. karyomegalic interstitial nephritis, linking
senescent cells delays ageing-associated 19, 807819 (2008). chronic kidney failure to defective DNA damage
disorders. Nature 479, 232236 (2011). 160. Humphreys, B.D. & Bonventre, J.V. repair. Nat. Genet. 44, 910915 (2012).
149. Yang, H.C. et al. Cells derived from young bone Mesenchymal stem cells in acute kidney injury. 172. Yang, H.C. et al. Cells derived from young bone
marrow alleviate renal aging. J. Am. Soc. Nephrol. Annu. Rev. Med. 59, 311325 (2008). marrow alleviate renal aging. J. Am. Soc. Nephrol.
22, 20282036 (2011). 161. Thum, T. et al. Age-dependent impairment of 22, 20282036 (2011).
150. Devarajan, P. Update on mechanisms of endothelial progenitor cells is corrected by 173. Kramann, R. & Humphreys, B.D. Kidney
ischemic acute kidney injury. J. Am. Soc. Nephrol. growthhormonemediated increase of insulin-like pericytes: roles in regeneration and repair.
17, 15031520 (2006). growthfactor1. Circ. Res. 100, 434443 (2007). Semin. Nephrol. 34, 374383 (2014).
151. Shurin, G.V. et al. Dynamic alteration of soluble 162. Behrens, A., van Deursen, J.M., Rudolph, K.L. 174. Duffield, J.S. & Lupher, M.L. Jr. PRM151
serum biomarkers in healthy aging. Cytokine 39, &Schumacher, B. Impact of genomic damage (recombinant human serum amyloid p/pentraxin2)
123129 (2007). and ageing on stem cell function. Nat. Cell Biol. for the treatment of fibrosis. DrugNews Perspect.
152. Kang, D.H. et al. Impaired angiogenesis in the 16, 201207 (2014). 23, 305315 (2010).
aging kidney: vascular endothelial growth factor 163. Stolzing, A. & Scutt, A. Age-related impairment of 175. Zhou, L. et al. Activation of p53 promotes renal
and thrombospondin1 in renal disease. Am. J. mesenchymal progenitor cell function. Aging Cell injury in acute aristolochic acid nephropathy.
Kidney Dis. 37, 601611 (2001). 5, 213224 (2006). J.Am. Soc. Nephrol. 21, 3141 (2010).
153. Ruiz-Torres, M.P. et al. Age-related increase 164. Schatteman, G.C. & Ma, N. Old bone marrow 176. Crose, L.E. et al. Alveolar rhabdomyosarcoma-
inexpression of TGF-1 in the rat kidney: cells inhibit skin wound vascularization. Stem Cells associated PAX3FOXO1 promotes
relationship to morphologic changes. J. Am. Soc. 24, 717721 (2006). tumorigenesis via hippo pathway suppression.
Nephrol. 9, 782791 (1998). 165. Gewin, L. & Zent, R. How does TGF- mediate J.Clin. Invest. 124, 285296 (2014).
154. Karam, Z. & Tuazon, J. Anatomic and physiologic tubulointerstitial fibrosis? Semin. Nephrol. 32,
changes of the aging kidney. Clin. Geriatr. Med. 228235 (2012). Author contributions
29, 555564 (2013). 166. Qi, W., Chen, X., Poronnik, P. & Pollock, C.A. The authors contributed equally to researching
155. Poulsom, R. et al. Bone marrow contributes to Transforming growth factor-beta/connective datafor the article, discussion of its content, writing
renal parenchymal turnover and regeneration. tissue growth factor axis in the kidney. Int. J. and reviewing and/or editing of the manuscript
J.Pathol. 195, 229235 (2001). Biochem. Cell Biol. 40, 913 (2008). beforesubmission.

NATURE REVIEWS | NEPHROLOGY ADVANCE ONLINE PUBLICATION | 13


2015 Macmillan Publishers Limited. All rights reserved

S-ar putea să vă placă și