You are on page 1of 17

Journal of Nephrology

https://doi.org/10.1007/s40620-020-00793-2

REVIEW

AKI: an increasingly recognized risk factor for CKD development


and progression
J. T. Kurzhagen1   · S. Dellepiane2   · V. Cantaluppi3   · H. Rabb1 

Received: 14 April 2020 / Accepted: 26 June 2020


© Italian Society of Nephrology 2020

Abstract
Acute kidney injury (AKI) is an increasing health burden with high morbidity and mortality rates worldwide. AKI is a risk
factor for chronic kidney disease (CKD) development and progression to end stage renal disease (ESRD). Rapid action is
required to find treatment options for AKI, plus to anticipate the development of CKD and other complications. Therefore,
it is essential to understand the pathophysiology of AKI to CKD transition. Over the last several years, research has revealed
maladaptive repair to be an interplay of cell death, endothelial dysfunction, tubular epithelial cell senescence, inflammatory
processes and more—terminating in fibrosis. Various pathological mechanisms have been discovered and reveal targets for
potential interventions. Furthermore, there have been clinical efforts measures for AKI prevention and progression including
the development of novel biomarkers and prediction models. In this review, we provide an overview of pathophysiological
mechanisms involved in kidney fibrosis. Furthermore, we discuss research gaps and promising therapeutic approaches for
AKI to CKD progression.

Keywords  AKI · CKD · AKD · Renal fibrosis · Mechanisms

Abbreviations bFGF Basic fibroblast growth factor


ADQI Acute disease quality initiative BM Bone marrow
Ag-presentation Antigen presentation BUN Blood urea nitrogen
AKD Acute kidney disease CCL2 CC-chemokine ligand 2
AKI Acute kidney injury CCR2 C–C chemokine receptor type 2
Akt Protein kinase B, PKB CDDO-dhTFEA CDDO-9,11-dihydro-trifluoroethyl
Alpha-SMA α-Smooth muscle actin Amide
AQP2 Aquaporine 2 CDDO-Im CDDO-imidazole
ASC Adipose mesenchymal stem cells CDDO-Me CDDO-methyl ester
ATN Acute tubular necrosis CI Confidence interval
BC-SC Blood cord stem cells CKD Chronic kidney disease
CRAC channel Ca2+ release-activated ­Ca2+ channel
CRRT​ Continuous renal replacement therapy
V. Cantaluppi: On behalf of the AKI and CRRT Project Group of
the Italian Society of Nephrology (SIN). CTGF CCN2 or connective tissue growth
factor
* H. Rabb Cxcl12 CXC-Motif-Chemokin 12
hrabb1@jhmi.edu CVD Cardiovascular diseases
1
Division of Nephrology, Johns Hopkins Hospital, Johns DAMPs Damaged-associated molecular
Hopkins University School of Medicine, Ross 965, 720 patterns
Rutland Ave, Baltimore, MD 21206, USA DCs Dendritic cells
2
Icahn School of Medicine at Mount Sinai, New York, NY, DGF Delayed graft function
USA DM Diabetes mellitus
3
Nephrology and Kidney Transplantation Unit, Department DN αβTCR double negative T cells
of Translational Medicine, University of Piemonte Orientale ECM Extracellular matrix
(UPO), “Maggiore Della Carità” University Hospital, EGF Epidermal growth factor
Novara, Italy

13
Vol.:(0123456789)
Journal of Nephrology

EMP Epithelial-mesenchymal plasticity RANTES Regulated upon activation, normal


EMT Epithelial-mesenchymal transition T-cell expressed and secreted
EndMT Endothelial-to-mesenchymal transition RIPK1 Receptor-interacting protein kinase-1
EPC Endothelial progenitor cells SC Stem cells
ER Endoplasmic reticulum Snai1 Snail Family Transcriptional Repressor
ERK 1/2 Extracellular-signal-regulated kinase 1
1/2 T2DM Type 2 diabetes mellitus
ESRD End stage renal disease TEC Tubular epithelial cells
EVs Extracellular vesicles TGF-β Transforming growth factor β
FGF-2 Fibroblast growth factor Th cells T helper cells,
GFR Glomerular filtration rate Th1 T helper cell 1
Glom Glomerular Th2 T helper cell 2
HGF Hepatocyte growth factor Th17 T helper cell 17
HPSE Heparanase TIMP-2 Tissue inhibitor of metalloproteinases-2
HR Hazard ratio TNF Tumor necrosis factor
HSC Hematopoietic SC TRADD TNF-receptor dead domain
IFN-γ Interferon γ Treg Regulatory T cells
IGF1 Insulin-like growth factor 1 Twist1 Twist Family BHLH Transcription Fac-
IGFBP7 Insulin-like growth factor-binding tor 1
protein 7 UUO Unilateral ureter obstruction
IL Interleukin VEGF Vascular endothelial growth factor
iPS Induced pluripotent SC
IRI Ischemic reperfusion injury
ISN International Society of Nephrology Introduction
KC Keratinocyte-derived chemokine
KDIGO Kidney disease improving global Acute kidney injury (AKI) is an enormous health burden
outcomes worldwide, and associated with end stage renal disease
Keap1 Kelch-like ECH-associated protein 1 (ESRD), cardiovascular diseases (CVD), hypertension and
KIM-1 Kidney injury molecule-1 death [1–3]. A recent meta-analysis including 154 studies
KO Knock out with more than 3.5 million AKI patients from mostly high-
LSC Liver stem cells income countries found a pooled incidence as high as 21.6%
M2 M2 macrophages and a mortality rate of 23.9% [4]. These numbers are even
MCP-1 Monocyte chemoattractant protein-1 higher for patients in intensive care units, where the inci-
MHC Major histocompatibility complex dence is up to 50% [5]. In high-income countries, AKI is
MLKL Mixed lineage kinase domain-like mainly seen in hospitalized patients, whereas in low-income
protein countries, it is mostly community-acquired due to dehydra-
MMP Matrix metalloprotease tion caused by gastroenteritis, acute infections and toxins
MSC Mesenchymal stem cells in combination with limited access to health care services
NET Neutrophil extracellular traps [6]. In 2015, the International Society of Nephrology (ISN)
NF-κB Nuclear factor ’kappa-light-chain- stated the treatment of AKI as a human right [7].
enhancer’ of activated B-cells AKI can result in a broad spectrum of renal outcomes
NGAL Neutrophil gelatinase-associated ranging from full recovery to ESRD. Historically, an
lipocalin improvement in serum creatinine and blood urea nitro-
NK cells Natural killer cells gen (BUN) was presumed full recovery of AKI. Over the
NO Nitric oxide last decade, there is an appreciation that many cases of
Nrf2 Nuclear factor erythroid 2-related fac- AKI lead to the development of chronic kidney disease
tor 2 (CKD). Even hospitalized patients with a small serum
Orai1 Calcium release-activated calcium creatinine increase (1.2 to < 1.5 times), that does meet
channel protein 1 the AKI Kidney Disease Improving Global Outcomes
PDGF Platelet-derived growth factor (KDIGO) guidelines 2012, have been shown to have a sig-
PIIINP Procollagen type III N-terminal peptide nificantly increased risk for CVD development and long-
PTEN Phosphatase and Tensin homolog term mortality [8]. In order to cover situations of kidney
injury that neither meet AKI (defined for the timeframe

13
Journal of Nephrology

Recurrence of AKI
Death

Recovery Progression
Sustained
reversal

Transient AKI Persistent AKI


<48h

AKI<7d
AKD CKD >90d

Risk factors: Severity/frequency of AKI, albuminuria, age, gender, pre-exisng CKD, hypoalbuminemia, arterial
hypertension, heart failure, obesity and diabetes mellitus

Fig. 1  Range of renal outcomes following AKI. According to Kid- (retrospectively day 0–day 7). AKI and AKD are overlapping syn-
ney Disease Improving Global Outcomes (KDIGO) guideline 2012, dromes. AKI can either be followed by adaptive repair processes
acute kidney injury (AKI) is defined as an increase in serum creati- leading to recovery or maladaptive repair resulting in progression to
nine (SCr) by 50% within 7 days or an increase in SCr by 0.3 mg/dl CKD. Hence, these processes are a continuum and multiple outcomes
within 2 days or oliguria. Chronic kidney disease (CKD) criteria are in between full recovery and progression to CKD are known. AKD
abnormalities of kidney structure or function for > 90 days based on has been defined as a syndrome covering the timeframe between AKI
cause, glomerular filtration rate (GFR) category, and albuminuria cat- and CKD. Known risk factors for AKI to CKD transition are severity
egory (CGA). Acute kidney disease (AKD) definition applies if AKI and frequency of AKI, albuminuria, age, gender, pre-existing CKD,
criteria were met or GFR < 60  ml/min per 1.73 ­m2 for < 3  months hypoalbuminemia, and chronic disease e.g. arterial hypertension,
or a decrease in GFR by ≥ 35% or increase in SCr by > 50% for < heart failure and diabetes mellitus
3 months occurred. AKD start point depends on which criteria apply

within the first 7 days) nor CKD criteria (defined for the (Canada), AKI-related healthcare costs were estimated to be
time > 90 days) the term acute kidney disease (AKD) has about 200 million Canadian dollars ($140 million) per year
been proposed [9–11] (Fig. 1). Though AKI is now rec- [19]. Moreover, the progression from AKI to CKD leads to
ognized as an important risk factor for CKD, the extent to comorbidity development and to a further medical expense
which AKI contributes to the incidence of CKD remains rise.
unknown. A recent meta-analysis of 82 studies including Given the heavy health burden of AKI and CKD, there
more than 2 million patients showed an elevated risk for is an urgent need to develop treatments for AKI and to pre-
hospitalized patients with AKI for new or progressive vent CKD. In this review, we provide a brief summary of
CKD (HR 2.67, 95% CI 1.99–3.58), ESRD (HR 4.81, 95% molecular mechanisms involved in maladaptive repair pro-
CI 3.04–7.62) and mortality (HR 1.8, 95% CI 1.61–2.02), cesses causing AKI to CKD transition and their potential
with an increased risk for CKD development following interventions. We will also suggest possible approaches for
an increased AKI stage [12]. Besides the severity of AKI, improvement of clinical strategies and patient follow up.
its frequency, as well as albuminuria, age, gender, pre-
existing CKD, hypoalbuminemia, and other chronic dis-
eases like arterial hypertension, heart failure, obesity and
diabetes mellitus (DM) have been identified as risk factors Mechanisms
for AKI to CKD progression [13–17].
The high hospitalization rate of AKI leads to enormous Maladaptive repair causing AKI to CKD transition
health care costs. In 2017, United States´ AKI costs were involves many pathophysiological processes. Mechanisms
estimated to be between $5.4 and $24 billion; in particular, of renal fibrosis involve renal tubular epithelial cells and
the in-hospital dialysis need during AKI was associated with the vascular system in the form of endothelial dysfunction
a cost increase of $40,000 per patient [18]. In a popula- and microvascular rarefaction. Inflammation involving T
tion-based study on 239,906 adults hospitalized in Alberta cells, B cells, macrophages, neutrophils, mast cells as well

13
Journal of Nephrology

Fig. 2  Overview of different
mechanisms involved in AKI
to CKD progression. Different
etiologic agents concur in acute C O
kidney injury (AKI) pathogene- O
sis (upper half of the figure), the Sepsis
Post-renal
most common are: endogenous Ischemia
and exogenous toxins, ischemia/ Failure
Inflammatory
hypoxia, microbial products, Response
immune molecules (e.g. com-
plement fragments, interleukins,
chemokines), urinary tract
obstruction and factors related
to organ cross-talk (e.g. hor-
mones, cytokines, fluid unbal- Intrinsic
ance etc.) and multi-organ fail- AKI
ure. When the injury overturns Endo or Exogenous
the compensatory response,
Nephrotoxins
tissue repair is defective (lower Mulorgan
half). Angiogenesis is impaired,
failure
cell cycle becomes dysfunc-
tional, tubular epithelium loses
its intracellular structure and
dedifferentiates towards a myo- Maladapve Repair
fibroblast phenotype, the extra-
cellular matrix is expanded and
post-injury immune infiltrate
may persist. Altogether, these
factors cause the progression to Cell cycle arrest
chronic kidney disease (CKD). Organelle damage
EMT  Epithelial-mesenchymal
transition, EndMT Endothelial-
to-mesenchymal transition

Capillary rarefaction
EndMT
EMT, apoptosis,
senescence

Matrix expansion and remodeling

Inflammatory cell infiltraon

as dendritic cells (DCs) occurs (Fig. 2). However, there Cell death


are differences in the molecular mechanisms depending on
the etiology of AKI and, therefore, to which extent each At least five different pathways can induce cellular death:
process contributes to the emerging fibrosis. necrosis, apoptosis, necroptosis, ferroptosis and pyroptosis.
In AKI, acute tubular necrosis (ATN), seen commonly with
ischemic and toxic injury, is characterized by accumulation
of dead epithelial cells in the lumen with the classical muddy

13
Journal of Nephrology

brown casts. Most cases of ATN (except severe forms like inflammasome can be over-activated during AKI and lead
cortical necrosis) resolve with the classical sequence: oligo- to pyroptosis with cell membrane fragmentation and mas-
anuria plus AKI, improvement in urine output or polyuria sive release of IL-1β and IL-18 [24]. There is emerging
and decreases in serum creatinine. This common clinical data describing an autoamplification loop fired by inflam-
scenario parallels the proliferation of the surviving cells as matory processes and necrosis. This process—called
elegantly shown by cell-fate tracking. However, the putative necroinflammation—plays a major role in tubular injury
role of local stem elements is still questioned [20]. [23].
Regulated necrosis as well as apoptosis are mechanisms
involved in cell death in AKI. Two different pathways can Extracellular matrix (ECM)
induce apoptosis: extrinsic and intrinsic. In the first case
immune cells release soluble mediators (e.g. tumor necro- Renal fibrosis is characterized by accumulation of extracel-
sis factor—TNF) or engage dead-receptor signaling (e.g. lular matrix (ECM) causing structural and therefore func-
Fas, a membrane homologue of TNF), thus activating cas- tional damage of the kidney. Immune cells and tubular cells
pase-8. Intrinsic apoptosis occurs when intracellular injury release profibrotic factors after AKI, which results in the
is irreversible and mitochondrial membrane is disrupted. activation and proliferation of myofibroblasts which pro-
Consequently, cytochromes are released in the cytoplasm duce ECM. The origins of myofibroblasts include endothe-
and promote the assembling of the apoptosome, a qua- lial cells (Endothelial-to-mesenchymal transition, EndMT),
ternary protein structure that activates caspase-9. Both epithelial cells (epithelial-mesenchymal transition, EMT)
pathways converge on the sequential activation of cas- and potentially immune cells [25].
pase-3, 7 and 6 and induce cell self-digestion and death.
Subsequently, immune scavengers phagocytize the remain- Endothelial‑to‑mesenchymal transition (EndMT)
ing apoptotic bodies without activating an inflammatory
response. In renal tubules, apoptotic cells are also shed in Peritubular endothelial cells represent a critical target dur-
the lumen and lost in urine. ing AKI. The alterations of endothelial function are mainly
Regulated necrosis in AKI includes necroptosis, fer- driven by a decreased bioavailability of nitric oxide and lead
roptosis, pyroptosis and G2/M cell cycle arrest. Necrop- to dysregulated blood flow, increased vascular permeability
tosis is an immunogenic form of programmed cell death and inflammatory cell recruitment. Recent studies suggested
and is activated by inflammatory cytokines, autophagy that EndMT plays a key role in capillary rarefaction and
failure and viruses expressing caspase-8 inhibitors. When CKD progression. EndMT is defined as the loss of typical
extrinsic apoptosis signaling prevails, receptor-interacting endothelial markers with the transition into a pro-fibrotic
protein kinase-1 (RIPK1) binds the TNF-receptor dead phenotype [26]; this phenomenon is a key feature in both
domain (TRADD) and activates caspase-8. If the inflam- ischemic and septic AKI [27, 28]. In a swine model of renal
matory signaling prevails (e.g. via interferon—IFN) or IRI, the activation of complement cascade induced EndMT
caspase-8 is inhibited, RIPK1 binds protein kinase R and via Akt (protein kinase B, PKB) pathway. Moreover, in vitro
phosphorylates RIPK3, which in turn activates mixed the anaphylatoxins C3a and C5a downregulated endothelial
lineage kinase domain-like protein (MLKL) and initiates antigen expression and promoted a fibroblast-like pheno-
necroptosis. Of note, RIPK3 is a major determinant of type [28]. As consequence, therapeutic strategies targeting
renal mitochondrial damage and is detectable in the urine EndMT, such as complement inhibition or stem cell infu-
of patients with sepsis-AKI [21]. MLKL is overexpressed sion, may enhance adaptive repair, thus limiting AKI to
by tubular cells following ischemia or tubular cast accu- CKD progression.
mulation and its inhibition is protective from AKI, and
MLKL can activate necroptosis in tubular epithelium inde- (Partial) epithelial‑to‑mesenchymal transition (EMT)
pendently from RIPK3 [22].
Ferroptosis results from lethal lipid peroxidation due Epithelial-to-mesenchymal transition (EMT) occurs when
to dysfunction of glutathione-peroxidase 4 and blocking terminally differentiated epithelial cells acquire a mesen-
has been shown to protect from renal ischemic reperfusion chymal phenotype. According to the original renal model
injury (IRI) in mice [23]. Another cell death pathway is of EMT, tubular cells lose epithelial markers and start
pyroptosis, a “hyper-inflammatory” cell death. Different expressing mesenchymal ones and therefore trigger fibrotic
cells respond to inflammation by assembling multimeric remodeling. Disruption of the cytoskeleton, breakdown of
protein complexes called inflammasomes that cleave and epithelial adhesion and basement membrane and synthesis
activate different cytokines and inflammatory caspases. of α-smooth muscle actin (α-SMA) have been described [29,
The description of the inflammasome response is beyond 30]. However, studies assessing cell lineages in a unilateral
the scope of this review. However, in brief, the canonical ureter obstruction (UUO) model as well as an IRI-model

13
Journal of Nephrology

resulted in contradicting findings and could not confirm Inflammatory processes


tubular epithelial cells to be the source for myofibroblasts.
Instead, pericytes and fibroblasts were found to develop Although the inflammatory processes in AKI have been
towards myofibroblasts [31, 32]. extensively studied, the data on soluble inflammatory fac-
More recent findings revealed the concept of epithelial- tors and immune cells contributing to AKI to CKD transi-
mesenchymal plasticity (EMP): a partial change in the tion is limited. It is important to note that the data has to
tubular-cell-phenotype towards mesenchymal status and be interpreted in the context of the experimental model. In
vice versa occurs in a cycle of dedifferentiation and repair. many studies, unilateral ischemic model is used, sometimes
Chronic triggers and tissue damage can disrupt this bal- with aggravation through following nephrectomy of the con-
ance and lead to development of a permanent mesenchymal tralateral kidney or high salt diet. Other common models
phenotype. Cell cycle arrest has been found to be a major include the UUO and bilateral ischemic models. Chronic
mechanism. Cells remain in G2 phase and are unable to re- changes—such as fibrosis—are often assessed after 6 weeks
differentiate as they do not pass the G2/M checkpoint. The [42]. Studies with more long-term follow-ups are rare. Stud-
G2/M checkpoint is the last control step before cells start ies on macrophages and DCs include glomerular as well as
mitosis. Traditionally this step was understood to be a con- interstitial fibrosis models which further complicates data
trol mechanism in response to DNA damage. In AKI to CKD interpretation.
transition it was also shown that transforming growth factor T cells—CD4+ and CD8+—infiltrate the kidney early
β (TGF-β), Snail Family Transcriptional Repressor 1 (Snai1) within hours after IRI, and have additional waves 5–6 weeks
and Twist Family BHLH Transcription Factor 1 (Twist1) later, when the expression of IL-1β and “regulated upon acti-
have been found to be major regulators in these pathophysi- vation, normal T-cell expressed and secreted” (RANTES)—
ologic pathways by inducing cellular arrest [33–35]. as general inflammation markers occurs in IRI kidneys, com-
Interestingly, heparanase (HPSE) is an endonuclease pared to contralateral and sham kidneys [42–44]. When T
regulating fibroblast growth factor (FGF-2) and TGF-β and cells from mice, that had undergone unilateral 60 min-IRI,
has been demonstrated to negatively impact the development were adoptively transferred into WT mice, the histological
of IRI-AKI induced EMT [36]. assessment showed no statistically significant difference in
fibrosis scores. However, a significant increase in albumi-
Tubular epithelial cell senescence nuria was measured after 12 weeks compared to sham mice
[45]. Interestingly, T cells in spleen of IRI mice as well as
Renal tubular epithelial cells play a key role in post-AKI IRI-primed lymphocyte recipients showed changes suggest-
transition to CKD. The identified mechanisms of this mala- ing a systemic proinflammatory response—in mice with kid-
daptive repair include: cell cycle arrest in G2/M phase EMT ney IRI, an increase of the proinflammatory cytokine Inter-
and increased production of profibrogenic factors [37]. All feron γ (IFN-γ) occurred in splenic T cells, even though
these biological processes induce tissue fibrosis and tubular there were no changes in numbers [42]. In mice that had
senescence. The latter is a dysfunctional phenotype of tubu- undergone adoptive transfer of IRI-primed lymphocytes,
lar cells characterized by organelle rarefaction, loss of solute CD3+ and CD4+ lymphocytes, more specifically acti-
carriers and upregulation of myo-fibroblast pathways like vated T cells (CD3+ CD25+) and memory T cells (CD4+
Wnt4/β-catenin [38]. A further mechanism of renal senes- CD44+), were significantly elevated in spleen compared to
cence is the down-regulation of the anti-aging molecule sham mice. These findings indicate a mechanistic role of
Klotho. Klotho protein is expressed as membrane recep- T cells in the pathogenesis of renal fibrosis following AKI
tor or released as soluble hormone by tubular convoluted [45]. Depletion of CD4+ and CD8+ T cells 24 h before
segments. Its precise mechanism of action is still unknown, unilateral ischemia resulted in less medullary damage and
however, circulating Klotho downregulates TGF-β signaling, reduced kidney IFN-γ expression. When depleting T cells
promotes phosphate clearance and maintains both endothe- after ischemia, IL-1β in the kidney was upregulated [44].
lial and tubular physiologic phenotype [39, 40]. As reported When assessing CD4+ T cells in AKI, the T helper cell
for endothelial cell dysfunction, complement activation has a (Th) 1 subset of CD4+ T cells has been demonstrated to
pivotal role in tubular injury: in a swine model of renal IRI, play a role in pathogenesis. An additional inflammatory
C1 inhibitor preserved tubular Klotho expression and C5a subgroup of CD4+ T cells—Th17 cells—mediates AKI to
in vitro reduced Klotho expression through nuclear factor CKD transition [46]. When treating rats with high salt diet
’kappa-light-chain-enhancer’ of activated B-cells (NF-κB) after AKI, an exacerbation was found. Interestingly, arterial
signaling and induced tubular senescence through epigenetic hypertension was found and CD4+ T cells showed higher
mechanisms [38]. Consistently, decreased Klotho expression amount of activation and Th17, Th1 and Th2 subgroups in
in kidney allografts was associated with delayed graft func- rats treated with high salt diet. When unilateral nephrectomy
tion (DGF) and earlier functional loss [41]. was performed, kidneys of rats that had been exposed to the

13
Journal of Nephrology

high salt diet, showed more fibrosis and cell volume in the It can be difficult to differentiate kidney DCs and mono-
interstitium. Losartan treatment reversed these effects and cytes/macrophages. In a model of glomerular disease, it
resulted in reduced CD4+ T cell kidney infiltration, includ- has been demonstrated that DCs aggravate progression
ing Th17, Th1 and Th2 subgroups. However, the percentage of kidney disease by promoting CD4+ T cell recruitment
of activated or memory T cells was not affected [43]. Cal- [53]. However, in a UUO model, a direct role in tissue
cium release-activated calcium channel protein 1 (Orai1) is fibrosis was not identified even though DCs were acti-
a subunit of Ca2+ release-activated C ­ a2+ (CRAC) channel, vated with a higher antigen presenting phenotype [54].
a membrane protein involved in the control of intracellular Cytokines that are produced by DCs are known to play
Ca2+ concentration. Orai1 was found to play an important a role in maladaptive repair. The extent to which those
role in the mechanism of CD4+ IL17+ T cells in AKI patho- released by DCs contribute to fibrosis is unknown. In
genesis. Its expression is increased after AKI and subsequent human fibrotic kidneys, the number of DCs is elevated,
intracellular ­Ca2+ accumulation leads to an upregulation of displaying a heightened expression of activation markers
ROR𝛾T activity which induces skewing towards IL-17 pro- and TGF-β production [55].
ducing T cells. Blocking of Orai1 through YM58483/BPT2 Albumin has been identified as a potential antigen in
resulted in attenuation of AKI and most importantly, to a rat CKD model that is reabsorbed from the tubules and
decrease in renal fibrosis. after proteasomal processing is presented by DCs leading
Following AKI, B cell numbers increase in mouse kid- to activation of CD8+ T cells, that produce IFN-γ. Dam-
neys and proliferation as well as differentiation into CD126+ aged-associated molecular patterns (DAMPs) seem to be
plasma cells occurs [47]. B cell-deficient mice have been required for this reaction since ovalbumin administration
shown to be protected from renal IRI, suggesting a patho- in healthy mice kidneys did not lead to an proinflammatory
genic contribution of B cells in AKI. However, adoptive immune response [56]. Depleting CD8+ T cells alone was
transfer of B cells into those B cell-deficient mice did not also shown to decrease fibrosis following AKI as CD8+ T
restore the susceptibility to AKI, whereas transfer of serum cells ameliorate monocyte-to-fibroblast transition driven
did [48]. In contrast, a study using a similar methodologic by CD4+ T cells [57].
approach described B cell-deficient mice to present worse Mast cells are early responders of inflammation and
AKI with decreased IL-10 in serum [49]. After kidney release large amounts of proinflammatory factors such
IRI, B cell-deficient mice present higher IL-10 and vascu- as cytokines, chemokines, growth factors, proteases and
lar endothelial growth factor levels, indicating that B cells profibrotic mediators (TGF-β, MMPs). Chymase, one of
might block repair processes and therefore contribute to the released proteases, has a unique role since it is also
fibrosis [47]. Overall, cellular and soluble response might involved in the activation of the angiotensin II pathway.
lead into different directions of repair and fibrosis. It might In steady state, there is only a small number of mast cells
also be the case that different subgroups of B cells represent found in the kidney, which increases during inflammatory
different roles in tissue fibrosis and repair processes [49]. conditions and correlates with fibrosis—mostly in the
T lymphocytes in mouse kidneys have been found months interstitium and rarely around glomeruli. There have been
after IRI and a population of CD19+ ∕low CD45R− B lympho- conflicting outcomes using different knock out (KO) and
cytes 16–18 months after bilateral IRI in the kidney with disease models suggesting the existence of different sub-
high expression of IL-6-Receptor (CD126) and Cxcl12- types of macrophages that require further observation [58].
Receptor (CD184). Cxcl12 is expressed by renal stromal Overall—similar to other immune cells described here—it
cells in AKI to CKD transition. Tertiary lymphoid organs is believed that the pro-fibrotic phenotype rather occurs
occur in the kidney after IRI [50]. in the setting of extended and chronic inflammation [59].
Macrophages also consist of different subgroups with Besides CD4+ T cells, γδ T cells, macrophages, DCs
different roles in AKI and AKI to CKD transition. Tradi- and natural killer (NK) cells are also known to release
tionally, M1 macrophages are regarded as proinflammatory IL-17, which promotes neutrophil invasion and therefore
and M2 macrophages are known to contribute to repair of renal fibrosis [60, 61]. Neutrophils accumulate in AKI
AKI. However, in sustained tubular injury, an uncontrolled kidneys and depletion in an UUO mouse model leads
hyperproliferation of M2 macrophages occurs and CD11b + to decreased M2 macrophage invasion and less fibrosis.
Ly6C low macrophages, a profibrotic M2 macrophage sub- Through production of neutrophil extracellular traps
population, is predominant [51]. Recruitment of fibroblasts (NET), which consist of pro-inflammatory cytokines, pro-
through ECM and pro-fibrotic factors as well as an increase teases and chromatin, they might contribute to necrosis
of matrix metalloprotease (MMP) expression contribute to in the kidney [23]. The pro-fibrotic roles of neutrophils
the development of renal fibrosis [52]. has recently been described to be MMP-9 dependent [62].
In contrast, several immune cells have been shown to
ameliorate fibrosis and contribute to tissue repair. Tregs

13
Journal of Nephrology

Tregs
Memory- Profibroc cytokines Monocytes
(CD44+), (TGF-β, IL-1 β, TNF,
Acvaon IL-6, IL-12)
DNs (CD25+),
IFN-γ, DCs M2
RANTES,
IL-1β Ag-presentaon repair

Acvaon (IL-4, IL-


B cells 6, Ag-presentaon) T cells M1 Macrophages
in early AKI
Acvaon
IFN-γ (IL-17)

CD8+
Profibroc
cytokines (NO,
Injury leads to TNFα, IL17)
release of IL-1, Il-6, Monocyte-to- M2
IL-17, IL-23, MCP-1, Promoon fibroblast ly6C low
B cells KC, IFN-γ CD4+ transion
Acvaon
CD19⁺/lowCD45R¯
Profibroc cytokines
Acvaon (TNFα, IL-4, IL-13)
IL-1 from monocytes, (IL-17) Transion
Acvaon NaCl promote MMP-9
(IL-6, differenciaon Neutrophils Mast Cells
Monoclonal Ab Cxcl12)
Profibroc Profibroc factors
Th1 Cytokines, (TGF-β1, PDGF, VEGF,
NET IGF1, Galacn3) Profibroc factors
Th17 Orai1 (TGF-β1, PDGF,
CTGF), Tryptase,
Chymas (AngII

Fibroblasts
acvaon)

Parenchymal
Transformaon/
acvaon
ECM
cells EMT,

Myofibroblasts
EndMT

Fig. 3  Immune cells involved in AKI to CKD transition. Various Dendritic cells, M2 M2 macrophages, Th1 T helper cell 1, Th17 T
immune cells contribute to pro-fibrotic processes in acute kidney helper cell 17, Treg regulatory T cells, DN αβTCR double negative
injury (AKI). Main mechanisms include the release of pro-fibrotic T cells, ECM Extra cellular matrix, Ag-presentation Antigen presen-
cytokines and profibrotic factors. Additionally, differentiation towards tation, RANTES Regulated upon activation, normal T-cell expressed
pro-fibrotic immune cell types and transition to fibroblasts or myofi- and secreted, Il interleukin, KC keratinocyte-derived chemokine,
broblasts have also been described. Immune cells that are involved MCP-1 Monocyte chemoattractant protein-1, MMP9 Matrix met-
in AKI to chronic kidney disease (CKD) transition are marked blue alloprotease-9, NET Neutrophil extracellular traps, Cxcl12 CXC-
and blue arrows display differentiation into subgroups. Production of Motif-Chemokin 12, Orai1 Calcium release-activated calcium chan-
pro-fibrotic cytokines, antigen-presentation and activation of other nel protein 1, PDGF Platelet-derived growth factor, VEGF Vascular
immune cells are shown in orange. Other pro-fibrotic factors, that Endothelial Growth Factor, CTGF CCN2 or connective tissue growth
are involved in myofibroblast-activation, are marked in brown. Cells factor, IGF1 Insulin-like growth factor 1
and pathways with anti-fibrotic functions are shown in green. DC

have been investigated extensively. αβTCR double nega- several clinical trials. Data from experimental studies
tive T cells (DN) lack CD4 and CD8 receptors and have suggest administration of growth factors, anti-oxidants
been shown to benefit in AKI [63] (Fig. 3). Even though and anti-fibrotic molecules. However, clinical results are
there is lack of knowledge of the specific roles of most of still lacking given that translation from experimental to
these cells in AKI to CKD progression, they hold signifi- clinical studies has usually failed in AKI development
cant potential for positive effects with possible treatment and progression. Recent experimental studies suggested
options in kidney fibrosis and further research should be new potential tools and OMICS technologies allowed the
conducted. identification of new molecules and pathways involved in
AKI to CKD transition, which has led to the discovery of
new biomarkers, that should greatly assist clinical trials
Potential interventions in this area.
We arbitrarily highlight two promising approaches that
Different therapeutic strategies have been proposed to limit our teams are studying: The first is the modulation of the
AKI to CKD progression: Established options include low Kelch-like ECH-associated protein 1/Nuclear factor eryth-
salt and protein intake or pharmacological inhibition of roid 2-related factor 2 (Keap1/Nrf2) pathway: Nrf2 is one of
the renin–angiotensin–aldosterone system confirmed by the most relevant anti-oxidant and anti-senescent molecules

13
Journal of Nephrology

expressed by renal resident cells and immune cells, sug- Pharmacologic Nrf2 activators have shown promising
gesting its putative role in the mechanisms of immune- benefit in mouse and human studies. Systemic administration
kidney interaction associated with adaptive or maladaptive of CDDO-imidazole (CDDO-Im) protected mice from AKI.
repair following AKI. The second approach is based on the However, contrasting effects using high dosage of CDDO-
expanding field of regenerative medicine: Stem cell-based 9,11-dihydro-trifluoroethyl Amide (CDDO-dhTFEA) have
therapies are the cornerstone of regenerative medicine and been described and kidney fibrosis was worsened, potentially
represent one of the ways to enhance tissue repair in acute through an activation of NF-κB, which is also regulated by
and chronic organ dysfunction. The field of stem cell therapy Keap1. CDDO-methyl ester (CDDO-Me) has been already
has recently received a major advance from the observa- used in clinical trials for treatment of patients with renal dis-
tion that their regenerative potential is mainly ascribed to eases and has been shown to increase eGFR in type 2 diabe-
the release of paracrine mediators. Among these paracrine tes mellitus (T2DM) patients with CKD stages 3–4 (BEAM
factors, extracellular vesicles (EV) represent a new way to study). However, adverse cardiovascular effects occurred in
enlighten the mechanisms of cell-to-cell communication: a similar study including T2DM participants with advanced
stem cell-derived EV have been evaluated in experimen- CKD stage 4 (BEACON study). Additionally, an increase of
tal and clinical studies as therapeutic approach for acute albuminuria was observed in both studies [66].
and chronic organ injury. Recent reports shed light into Since T cells could be a direct mediator of AKI to CKD
the mechanisms of EV biogenesis and formation and more progression, they constitute a particularly attractive thera-
innovative protocols have been developed for isolation and peutic target. It could be shown, that adoptive transfer of T
characterization of EV: for all these reasons, stem EV are cells from Keap1- KO mice leads to protection from AKI in
currently under evaluation in clinical trials as regenerative the recipient mice [68]. However, whether this approach is
strategy of several organs including the kidney. also applicable to prevent AKI to CKD progression has yet
to be investigated. Successful Keap1-KO using CRISPR/
Nrf2 modification Cas9 technique in human T cells has already been demon-
strated and might pave the way for future immunotherapy
The Keap1/Nrf2 pathway is a promising therapeutic target [71].
for kidney diseases. In steady state, Keap1 inhibits the activ-
ity of Nrf2, a major player in cytotoxic stress protection. Regenerative role of stem cell‑derived extracellular
This connection is released by oxidative stress leading to an vesicles (EVs)
upregulation of Nrf2 target genes. In AKI, a high oxidative
stress level occurs along with IRI [64] and CKD has been Different types of stem cells have been investigated as
shown to be associated with decreased Nrf2 activity [65]. potential therapeutic strategy in experimental models of
Therefore, in both conditions an increase of the Nrf2 expres- AKI: However, the results of clinical trials mainly based
sion is considered a promising therapeutic strategy and dif- on mesenchymal stromal cell (MSC) administration to pre-
ferent approaches, such as pharmacologic modulation and vent AKI in patients at high risk after cardiac surgery were
gene editing, are being investigated in basic research studies conflicting. Recently, it has been shown that the regenera-
as well as clinical trials [66]. tive potential of stem cells is primarily ascribed to parac-
In murine model, Keap1 whole body KO mice are not rine mechanisms including the release of growth factors
viable, but the hypomorphic mutant has been shown to be and EVs. EVs are microparticles released by different
protected from AKI and to develop less fibrosis, indicating types of progenitors and able to induce epigenetic repro-
a beneficial effect of Nrf2 upregulation for AKI to CKD gramming of injured renal cells through the direct transfer
transition in a murine model [67]. Tissue specific KO of of proteins, lipids, mRNA and microRNA [72]. Compared
Keap1 gene protects from AKI: Keap1-KO in immune cells to cellular therapies, EVs have minimal immunological
was shown to ameliorate AKI [68]. However, deleting Keap1 and neoplastic risk while being extremely malleable. Most
in adult tubular cells may be a viable option with potential of stem cell derived EVs do not express major histocom-
for protection [67]. Yet, there is data showing that Keap1 patibility complex (MHC) molecules, thus can be repeat-
gene in tubular epithelial cells may be involved in homeo- edly infused without inducing allo-immunization. Vesicle
stasis and development of the kidneys. Kidney epithelial cell content varies according to the parental cell phenotype and
specific Keap1-KO resulted in hydronephrosis, suggesting culture conditions and, as EVs do not carry coding DNA,
Keap1 gene to play an important role in development of kid- parental gene editing may enhance specific EV proprieties
neys, particularly when water/Aquaporine 2 (AQP2) trans- without substantial risk of transformation in target cells
port is impaired [69, 70]. Thus, caution is necessary when [73]. Various EV types have been tested as therapeutic
deciding which cell type to edit. tools in pre-clinical models of AKI; given the kidneys
being mesodermic organs, most studies have investigated

13
Journal of Nephrology

Table 1  Pre-clinical studies about extracellular vesicles (EVs) as acute kidney injury (AKI) therapy
Author References EV source AKI model Mechanism

C. Wang [76] HSC IRI miR-199a-5p inhibits ER stress in tubular cells


T. Pan [77] myocytes Sepsis miR-21 inhibits Nf-κB and PTEN in tubular cells
X. Wu [78] BC-SC Cardiac surgery Reduced inflammatory infiltration
J.L. Viñas [79] EPC IRI miR486-5p transfer promotes regenerative cell chemotaxis
J.M. Dominguez 2nd [80] TEC IRI Modulation of > 300 proteins differentially expressed in IRI
G. Zhang [81] Kidney vein—TEC IRI Apoptosis downregulation
B. Wang [82] MSC Cisplatin Autophagy induction
A. Ranghino [83] Glom MSC IRI miRNA modulation (most upregulated: hsa-miR-299-5p)
J.H. Dominguez [84] TEC IRI IRI altered transcript were reduced from 3100 to 1600
S. Bruno [85] MSC Glycerol miRNA transfer, miR-486-5p most enriched
X. Zou [86] MSC IRI Downregulation of NK cell migration to kidneys
X. Zou [87] MSC IRI VEGF and angiogenesis upregulation
G. Zhang [88] BC-SC IRI Nrf2 and heme oxygenase 1 upregulation
J. L Viñas [89] EPC IRI miR486-5p transfer curbs PTEN activation
B. Shen [90] MSC IRI CCR2 exosome clear CCL2 and prevent macrophage migration
K.C. Lin [91] ASC IRI Apoptosis downregulation
D. Gu [92] MSC IRI miR-30 inhibits mitochondrial damage
D. C. de Almeida [93] MSC Cisplatin miRNA transfer (miR-880, miR-141, miR-377, and miR-21)
reduces apoptosis
G. Q. Ju [94] MSC IRI HGF induction via miRNA transfer
D. Burger [95] EPC IRI Apoptosis inhibition
X. Zou [96] BC-SC IRI Suppression of CX3CL1
G. Zhang [97] BC-SC IRI Downregulation of inducible NO synthase
M.B. Herrera Sanchez [98] LSC Glycerol Improvement of tubular proliferation
H. Y. Choi [99] Kidney SC IRI Transfer of growth factor RNA (VEGF, bFGF etc.)
Y. Zhou [100] BC-SC Cisplatin ERK pathway upregulation
L. Kilpinen [101] BC-SC IRI Transfer of growth factors
V. Cantaluppi [74] EPC IRI Angiogenic miRNA (miR-126 and miR-296) delivery
S. Bruno [102] MSC Cisplatin Apoptosis inhibition
S. Gatti [103] MSC IRI miRNA transfer, apoptosis inhibition
S. Bruno [104] MSC Glycerol EVs adhere to tubules via CD44 and induce tubular cell pro-
liferation
V. Cantaluppi [105] EPC Glomerulonephritis Complement inhibition via miRNA transfer

AKI Acute kidney injury, ASC Adipose mesenchymal stem cells, BC-SC Blood cord (Warton Jelly) stem cells, bFGF Basic fibroblast growth
factor, CCL2 CC-chemokine ligand 2, CCR2 C–C chemokine receptor type  2, EPC Endothelial progenitor cells, ER Endoplasmic reticulum,
ERK 1/2 Extracellular-signal-regulated kinase 1/2, EVs Extracellular vesicles, Glom Glomerular, HGF Hepatocyte growth factor, HSC Hemat-
opoietic stem cells, IRI Ischemia reperfusion injury, LSC Liver stem cells, MSC Mesenchymal stem cells, NK cells Natural Killer cells, NO
Nitric oxide, Nrf2 Nuclear factor erythroid 2-related factor 2, PTEN Phosphatase and Tensin homolog, TEC Tubular epithelial cells, VEGF Vas-
cular Endothelial Growth Factor

EVs of mesenchymal origin, including MSC, hematopoi- and histology during AKI and reduced the inflammatory
etic SC (HSC), endothelial progenitors and tubular cell burst. Of note, EV infusion was non-inferior to stem cell-
lines (Table 1) [74, 75]. A recent meta-analysis on EV based therapies without potential adverse effects such as
therapy in experimental AKI was performed. However, tumorigenesis and maldifferentiation (Table 1). In experi-
the paper was limited by the heterogeneity of pre-clinical mental renal IRI, EVs from endothelial progenitors local-
investigations and acknowledged a significant publication ized in both the renal peri-tubular capillaries and tubular
bias [75]. Nevertheless, 31 studies met the pre-specified epithelial cells and prevented AKI to CKD progression. Of
quality criteria and the authors found that stem cell EVs note, both RNAse treatment of EVs and silencing of Dicer
(mainly from MSC) consistently improved renal function (a key enzyme in microRNA biogenesis) in origin cells

13
Journal of Nephrology

Fig. 4  Stem cells and extracellular vesicles as treatment options for with a viral transfection of specific oncogenes). SC can be directly
AKI. Various stem cell types have been investigated in preclinical infused as therapeutic agents or can be cultivated to collect their ben-
models to treat acute kidney injury (AKI). Bone marrow stem cells eficial soluble factors. In particular, extracellular vesicles can be iso-
(SC) and progenitors are by far the most studied and include endothe- lated by ultra-centrifugation or specific enrichment membranes. BM
lial progenitor cells, mesenchymal SC and hematopoietic SC. Other bone marrow, SC stem cells, EPC endothelial progenitor cells, MSC
sources are: blood cord, embryonic tissue, various mesenchymal tis- mesenchymal stem cells, HSC hematopoietic stem cells, BC blood
sues (including adipose SC), kidney progenitors (that express CD133 cord, ASC adipose stem cells, iPS induced pluripotent stem cells, EV
surface marker) and the induced pluripotent SC (usually obtained extracellular vesicles

completely abrogated this protective effect. These results To pave the way for timely diagnosis of AKI and evalu-
suggest the key role of RNA transfer from EVs to target ation of prognosis, clinical multivariant models as well as
cells in order to facilitate kidney repair [74] (Fig. 4). Bas- novel biomarkers have been investigated.
ing on these results, stem cell-derived EVs may represent Hodgson et al. recently summarized efforts of the past
an intriguing option for limiting AKI to CKD progression. decades regarding prediction models and AKI alerts and
concluded that early diagnosis of AKI can only improve
Clinical interventions outcomes if adequate interventions follow. In a prospective
cohort analysis, the outcome of patients suffering from hos-
In order to address optimal care for the high-risk popula- pital-acquired AKI could be improved using this approach.
tion of AKI patients, attempts have been made for improve- A sophisticated flow chart advised for actions depending
ment in clinical management. Most importantly, the need on the risk level, such as further assessment of underlying
for preventive actions and early detection measures has to be disease and fluid status, further blood and urinary labora-
emphasized. This could enable proactive management and tory assessments, imaging, reviewing of drugs, and patient
anticipation of disease progression. education. However, study participants with community-
acquired AKI showed no benefit, which, according to the
authors, might be due to lack of correct documentation of

13
Journal of Nephrology

kidney risk factors before hospital admission. The impor- follow-up visits in this collective [111, 112]. Harel et al.
tance of a transfer of information between primary and sec- found decreased mortality in AKI patients that were under
ondary care systems was emphasized [106]. Another review nephrological care [113]. Nonetheless, causality has not
similarly found that the combination of care bundles, e-alerts been validated.
and education led to the best outcomes [107]. Patients under Due to limited evidence, there is no detailed guideline
intensive medical care, benefited from a recently developed yet for the management of AKI survivors. The current
risk prediction score [108, 109]. KDIGO guideline recommends an evaluation of patients
The current gold standard for AKI diagnosis is still rep- three months after AKI [114]. The Acute Disease Quality
resented by serum creatinine, which has major limitations Initiative (ADQI) suggests the intensity and frequency of
since it detects AKI on an advanced stage and gives no surveillance be proportionate to the risk of future outcomes.
insight in the underlying pathogenesis. In order to evalu- The follow-up management recommendations include
ate the risk for AKI to CKD transition, it would be par- patient education, medication reconciliation, nephrotoxin
ticularly useful to find novel biomarkers that detect early avoidance, and kidney function monitoring [9]. Basing on
stages of AKI and provide information about maladaptive the previous considerations, the AKI and continuous renal
repair processes. Following these approaches, not only have replacement therapy (CRRT) Project Group of the Italian
biomarkers displaying early AKI as well as tubular injury Society of Nephrology (SIN) started an ongoing study with
and inflammatory processes been the subject of research, the following aims: (1) to evaluate the “real-life” AKI inci-
but also prognostic markers that help distinguish following dence in hospitalized patients; (2) to start a collaborative
adaptive repair and fibrosis. Urinary neutrophil gelatinase- program between Nephrologists and General Practitioners
associated lipocalin (NGAL) is already used in clinical for an integrated strategy of AKI follow-up; (3) to create a
practice in Europe and Asia as a marker of early tubular biobank for the early identification of biomarkers of AKI to
injury. Furthermore, kidney injury molecule-1 (KIM-1) has CKD progression.
been investigated as a tubular injury biomarker and its abil-
ity to predict development of CKD has been demonstrated.
However, for CKD progression of advanced stages of CKD Conclusion
following AKI insults, advantages over established mark-
ers were not found. Tissue inhibitor of metalloproteinases-2 AKI is an increasingly recognized risk factor for CKD devel-
(TIMP-2) and insulin-like growth factor-binding protein 7 opment and progression. There have been developments in
(IGFBP7) are commercially available urine biomarkers that elucidating the molecular and cellular mechanisms involved
are used for AKI risk assessment in critically ill patients and in maladaptive repair processes revealing possible targets
harbor potential for further clinical applications. As poten- for treatment. In addition, approaches in public health, pre-
tial detectors of inflammatory processes proinflammatory diction models and clinical care initiatives have also been
cytokines (e.g. IL-18, IL-6, IL-10), monocyte chemoattract- studied with promising results. To decrease of AKI to CKD
ant protein-1 (MCP-1) and TNF receptors are investigated. progression worldwide, a combination of all these strategies
For distinction of adaptive and maladaptive repair, there is will be necessary. However, further research is required to
urinary procollagen type III N-terminal peptide (PIIINP), make a major impact to help our patients with AKI to CKD
which is a product of collagen synthesis and might be a progression.
promising representator of kidney fibrosis. Yet, urinary
epidermal growth factor (EGF) is inversely correlated with
interstitial fibrosis and shows potential as a biomarker rep- Funding  This study was (partially) funded by the Italian Ministry of
resenting renal repair processes [110]. Education, University and Research (MIUR) program “Departments of
Excellence 2018-2022”, AGING Project—Department of Translational
Additionally, consistent follow up management of AKI as Medicine, Università del Piemonte Orientale (UPO). HR was supported
secondary or tertiary preventive measures is required. Thus, by grants from the NIDDK and philanthropic gifts from Rogelio Miro
several interesting studies have been performed to investi- and Sammy Eldin. JTK was supported with a scholarship by the Dr.
gate patient and health care provider awareness after AKI. Werner Jackstädt-Foundation (project number S 134–10.117).
It has been shown that the majority of patients with AKI
lack awareness of their condition as well as knowledge of Compliance with ethical standards 
risk factors after hospital discharge [16]. Additionally, the
Conflict of interest  The authors have declared that no conflict of inter-
compliance of patients for nephrological follow-ups after est exists.
hospital discharge is very low, although it has been demon-
strated that automatic scheduling can improve this issue. The Ethical approval  This article does not contain any studies with human
same study also stated 70% of the observed patients required participants performed by any of the authors.
at least one intervention, emphasizing the importance of

13
Journal of Nephrology

References risk of adverse outcomes after acute kidney injury: a system-


atic review and meta-analysis of cohort studies using consensus
definitions of exposure. Kidney Int 95(1):160–172. https​://doi.
1. Ishani A, Xue JL, Himmelfarb J, Eggers PW, Kimmel PL, Molit-
org/10.1016/j.kint.2018.08.036
oris BA, Collins AJ (2009) Acute kidney injury increases risk of
13. Parr SK, Matheny ME, Abdel-Kader K, Greevy RA Jr, Bian
ESRD among elderly. J Am Soc Nephrol 20(1):223–228. https​
A, Fly J, Chen G, Speroff T, Hung AM, Ikizler TA, Siew ED
://doi.org/10.1681/asn.20070​80837​
(2018) Acute kidney injury is a risk factor for subsequent pro-
2. Pechman KR, De Miguel C, Lund H, Leonard EC, Basile DP,
teinuria. Kidney Int 93(2):460–469. https​://doi.org/10.1016/j.
Mattson DL (2009) Recovery from renal ischemia-reperfusion
kint.2017.07.007
injury is associated with altered renal hemodynamics, blunted
14. Thakar CV, Christianson A, Himmelfarb J, Leonard AC (2011)
pressure natriuresis, and sodium-sensitive hypertension. Am J
Acute kidney injury episodes and chronic kidney disease risk in
Physiol Regul Integr Comp Physiol 297(5):R1358–1363. https​
diabetes mellitus. Clin J Am Soc Nephrol 6(11):2567–2572. https​
://doi.org/10.1152/ajpre​gu.91022​.2008
://doi.org/10.2215/cjn.01120​211
3. James MT, Ghali WA, Knudtson ML, Ravani P, Tonelli M,
15. Amdur RL, Chawla LS, Amodeo S, Kimmel PL, Palant CE
Faris P, Pannu N, Manns BJ, Klarenbach SW, Hemmelgarn
(2009) Outcomes following diagnosis of acute renal failure
BR (2011) Associations between acute kidney injury and car-
in US veterans: focus on acute tubular necrosis. Kidney Int
diovascular and renal outcomes after coronary angiography.
76(10):1089–1097. https​://doi.org/10.1038/ki.2009.332
Circulation 123(4):409–416. https​://doi.org/10.1161/circu​latio​
16. Chawla LS, Seneff MG, Nelson DR, Williams M, Levy H, Kim-
naha.110.97016​0
mel PL, Macias WL (2007) Elevated plasma concentrations of
4. Susantitaphong P, Cruz DN, Cerda J, Abulfaraj M, Alqahtani
IL-6 and elevated APACHE II score predict acute kidney injury
F, Koulouridis I, Jaber BL (2013) World incidence of AKI: a
in patients with severe sepsis. Clin J Am Soc Nephrol 2(1):22–
meta-analysis. Clin J Am Soc Nephrol 8(9):1482–1493. https​://
30. https​://doi.org/10.2215/cjn.02510​706
doi.org/10.2215/cjn.00710​113
17. Coca SG, Singanamala S, Parikh CR (2012) Chronic kidney
5. Hoste EA, Bagshaw SM, Bellomo R, Cely CM, Colman R, Cruz
disease after acute kidney injury: a systematic review and meta-
DN, Edipidis K, Forni LG, Gomersall CD, Govil D, Honore PM,
analysis. Kidney Int 81(5):442–448. https​://doi.org/10.1038/
Joannes-Boyau O, Joannidis M, Korhonen AM, Lavrentieva A,
ki.2011.379
Mehta RL, Palevsky P, Roessler E, Ronco C, Uchino S, Vazquez
18. Silver SA, Chertow GM (2017) The economic consequences of
JA, Vidal Andrade E, Webb S, Kellum JA (2015) Epidemiology
acute kidney injury. Nephron. https:​ //doi.org/10.1159/000475​ 607
of acute kidney injury in critically ill patients: the multinational
19. Collister D, Pannu N, Ye F, James M, Hemmelgarn B, Chui B,
AKI-EPI study. Intensive Care Med 41(8):1411–1423. https​://
Manns B, Klarenbach S (2017) Health care costs associated
doi.org/10.1007/s0013​4-015-3934-7
with AKI. Clin J Am Soc Nephrol 12(11):1733–1743. https​://
6. Cerda J, Bagga A, Kher V, Chakravarthi RM (2008) The con-
doi.org/10.2215/cjn.00950​117
trasting characteristics of acute kidney injury in developed and
20. Kusaba T, Lalli M, Kramann R, Kobayashi A, Humphreys BD
developing countries. Nat Clin Pract Nephrol 4(3):138–153. https​
(2014) Differentiated kidney epithelial cells repair injured proxi-
://doi.org/10.1038/ncpne​ph072​2
mal tubule. Proc Natl Acad Sci US A 111(4):1527–1532. https:​ //
7. Mehta RL, Cerda J, Burdmann EA, Tonelli M, Garcia-Garcia
doi.org/10.1073/pnas.13106​53110​
G, Jha V, Susantitaphong P, Rocco M, Vanholder R, Sever
21. Sureshbabu A, Patino E, Ma KC, Laursen K, Finkelsztein EJ,
MS, Cruz D, Jaber B, Lameire NH, Lombardi R, Lewington
Akchurin O, Muthukumar T, Ryter SW, Gudas L, Choi AMK,
A, Feehally J, Finkelstein F, Levin N, Pannu N, Thomas B,
Choi ME (2018) RIPK3 promotes sepsis-induced acute kidney
Aronoff-Spencer E, Remuzzi G (2015) International Society of
injury via mitochondrial dysfunction. JCI Insight. https​://doi.
Nephrology’s 0by25 initiative for acute kidney injury (zero pre-
org/10.1172/jci.insig​ht.98411​
ventable deaths by 2025): a human rights case for nephrology.
22. Liu W, Chen B, Wang Y, Meng C, Huang H, Huang XR, Qin J,
Lancet 385(9987):2616–2643. https​://doi.org/10.1016/s0140​
Mulay SR, Anders HJ, Qiu A, Yang B, Freeman GJ, Lu HJ, Lin
-6736(15)60126​-x
HY, Zheng ZH, Lan HY, Huang Y, Xia Y (2018) RGMb protects
8. Losito A, Nunzi E, Pittavini L, Zampi I, Zampi E (2018) Car-
against acute kidney injury by inhibiting tubular cell necroptosis
diovascular morbidity and long term mortality associated with in
via an MLKL-dependent mechanism. Proc Natl Acad Sci USA
hospital small increases of serum creatinine. J Nephrol 31(1):71–
115(7):E1475–E1484. https:​ //doi.org/10.1073/pnas.171695​ 9115​
77. https​://doi.org/10.1007/s4062​0-017-0413-y
23. Mulay SR, Linkermann A, Anders HJ (2016) Necroinflammation
9. Chawla LS, Bellomo R, Bihorac A, Goldstein SL, Siew ED, Bag-
in kidney disease. J Am Soc Nephrol 27(1):27–39. https​://doi.
shaw SM, Bittleman D, Cruz D, Endre Z, Fitzgerald RL, Forni
org/10.1681/asn.20150​40405​
L, Kane-Gill SL, Hoste E, Koyner J, Liu KD, Macedo E, Mehta
24. Komada T, Muruve DA (2019) The role of inflammasomes in
R, Murray P, Nadim M, Ostermann M, Palevsky PM, Pannu N,
kidney disease. Nat Rev Nephrol 15(8):501–520. https​://doi.
Rosner M, Wald R, Zarbock A, Ronco C, Kellum JA (2017)
org/10.1038/s4158​1-019-0158-z
Acute kidney disease and renal recovery: consensus report of the
25. Yuan Q, Tan RJ, Liu Y (2019) Myofibroblast in kidney fibrosis:
Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat Rev
origin, activation, and regulation. Adv Exp Med Biol 1165:253–
Nephrol 13(4):241–257. https​://doi.org/10.1038/nrnep​h.2017.2
283. https​://doi.org/10.1007/978-981-13-8871-2_12
10. KDIGO (2012) (Kidney Disease: Improving Global Outcomes).
26. Jourde-Chiche N, Fakhouri F, Dou L, Bellien J, Burtey S, Frimat
Clinical Practice. Guideline for the Evaluation and Management
M, Jarrot PA, Kaplanski G, Le Quintrec M, Pernin V, Rigothier
of Chronic Kidney Disease. Kidney Int Suppl 3:1–150
C, Sallee M, Fremeaux-Bacchi V, Guerrot D, Roumenina LT
11. Levey AS, de Jong PE, Coresh J, El Nahas M, Astor BC, Mat-
(2019) Endothelium structure and function in kidney health and
sushita K, Gansevoort RT, Kasiske BL, Eckardt KU (2011)
disease. Nat Rev Nephrol 15(2):87–108. https:​ //doi.org/10.1038/
The definition, classification, and prognosis of chronic kidney
s4158​1-018-0098-z
disease: a KDIGO Controversies Conference report. Kidney Int
27. Stasi A, Intini A, Divella C, Franzin R, Montemurno E, Granda-
80(1):17–28. https​://doi.org/10.1038/ki.2010.483
liano G, Ronco C, Fiaccadori E, Pertosa GB, Gesualdo L, Cas-
12. See EJ, Jayasinghe K, Glassford N, Bailey M, Johnson DW,
tellano G (2017) Emerging role of Lipopolysaccharide binding
Polkinghorne KR, Toussaint ND, Bellomo R (2019) Long-term

13
Journal of Nephrology

protein in sepsis-induced acute kidney injury. Nephrol Dial V, Lucarelli G, Ditonno P, Battaglia M, Crovace A, Staffieri
Transplant 32(1):24–31. https​://doi.org/10.1093/ndt/gfw25​0 F, Oortwijn B, van Amersfoort E, Pertosa G, Grandaliano G,
28. Curci C, Castellano G, Stasi A, Divella C, Loverre A, Gigante Gesualdo L (2016) Complement modulation of anti-aging factor
M, Simone S, Cariello M, Montinaro V, Lucarelli G, Ditonno P, Klotho in ischemia/reperfusion injury and delayed graft function.
Battaglia M, Crovace A, Staffieri F, Oortwijn B, van Amersfoort Am J Transpl 16(1):325–333. https​://doi.org/10.1111/ajt.13415​
E, Gesualdo L, Grandaliano G (2014) Endothelial-to-mesenchy- 41. Castellano G, Franzin R, Stasi A, Divella C, Sallustio F, Pontrelli
mal transition and renal fibrosis in ischaemia/reperfusion injury P, Lucarelli G, Battaglia M, Staffieri F, Crovace A, Stallone G,
are mediated by complement anaphylatoxins and Akt pathway. Seelen M, Daha MR, Grandaliano G, Gesualdo L (2018) Com-
Nephrol Dial Transpl 29(4):799–808. https​://doi.org/10.1093/ plement activation during ischemia/reperfusion injury induces
ndt/gft51​6 pericyte-to-myofibroblast transdifferentiation regulating peritu-
29. Liu Y (2004) Epithelial to mesenchymal transition in renal fibro- bular capillary lumen reduction through pERK signaling. Front
genesis: pathologic significance, molecular mechanism, and Immunol 9:1002. https​://doi.org/10.3389/fimmu​.2018.01002​
therapeutic intervention. J Am Soc Nephrol 15(1):1–12. https​:// 42. Burne-Taney MJ, Yokota N, Rabb H (2005) Persistent
doi.org/10.1097/01.asn.00001​06015​.29070​.e7 renal and extrarenal immune changes after severe ischemic
30. Liu Y (2010) New insights into epithelial-mesenchymal transi- injury. Kidney Int 67(3):1002–1009. https​://doi.org/10.111
tion in kidney fibrosis. J Am Soc Nephrol 21(2):212–222. https​ 1/j.1523-1755.2005.00163​.x
://doi.org/10.1681/asn.20081​21226​ 43. Mehrotra P, Patel JB, Ivancic CM, Collett JA, Basile DP (2015)
31. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Th-17 cell activation in response to high salt following acute
Bonventre JV, Valerius MT, McMahon AP, Duffield JS (2010) kidney injury is associated with progressive fibrosis and attenu-
Fate tracing reveals the pericyte and not epithelial origin of ated by AT-1R antagonism. Kidney Int 88(4):776–784. https​://
myofibroblasts in kidney fibrosis. Am J Pathol 176(1):85–97. doi.org/10.1038/ki.2015.200
https​://doi.org/10.2353/ajpat​h.2010.09051​7 44. Ascon M, Ascon DB, Liu M, Cheadle C, Sarkar C, Racusen L,
32. Kriz W, Kaissling B, Le Hir M (2011) Epithelial-mesenchymal Hassoun HT, Rabb H (2009) Renal ischemia-reperfusion leads
transition (EMT) in kidney fibrosis: fact or fantasy? J Clin Invest to long term infiltration of activated and effector-memory T lym-
121(2):468–474. https​://doi.org/10.1172/jci44​595 phocytes. Kidney Int 75(5):526–535. https​://doi.org/10.1038/
33. Fintha A, Gasparics Á, Rosivall L, Sebe A (2019) Therapeu- ki.2008.602
tic targeting of fibrotic epithelial-mesenchymal transition-an 45. Burne-Taney MJ, Liu M, Ascon D, Molls RR, Racusen L, Rabb
outstanding challenge. Front Pharmacol 10:388. https​://doi. H (2006) Transfer of lymphocytes from mice with renal ischemia
org/10.3389/fphar​.2019.00388​ can induce albuminuria in naive mice: a possible mechanism
34. Grande MT, Sánchez-Laorden B, López-Blau C, De Frutos CA, linking early injury and progressive renal disease? Am J Physiol
Boutet A, Arévalo M, Rowe RG, Weiss SJ, López-Novoa JM, Renal Physiol 291(5):F981–986. https​://doi.org/10.1152/ajpre​
Nieto MA (2015) Snail1-induced partial epithelial-to-mesenchy- nal.00229​.2005
mal transition drives renal fibrosis in mice and can be targeted to 46. Yokota N, Burne-Taney M, Racusen L, Rabb H (2003) Contrast-
reverse established disease. Nat Med 21(9):989–997. https​://doi. ing roles for STAT4 and STAT6 signal transduction pathways in
org/10.1038/nm.3901 murine renal ischemia-reperfusion injury. Am J Physiol Renal
35. Lovisa S, LeBleu VS, Tampe B, Sugimoto H, Vadnagara K, Physiol 285(2):F319–325. https:​ //doi.org/10.1152/ajpren​ al.00432​
Carstens JL, Wu CC, Hagos Y, Burckhardt BC, Pentcheva-Hoang .2002
T, Nischal H, Allison JP, Zeisberg M, Kalluri R (2015) Epi- 47. Jang HR, Gandolfo MT, Ko GJ, Satpute SR, Racusen L, Rabb
thelial-to-mesenchymal transition induces cell cycle arrest and H (2010) B cells limit repair after ischemic acute kidney injury.
parenchymal damage in renal fibrosis. Nat Med 21(9):998–1009. J Am Soc Nephrol 21(4):654–665. https​://doi.org/10.1681/
https​://doi.org/10.1038/nm.3902 asn.20090​20182​
36. Masola V, Zaza G, Gambaro G, Onisto M, Bellin G, Vischini G, 48. Burne-Taney MJ, Ascon DB, Daniels F, Racusen L, Baldwin W,
Khamaysi I, Hassan A, Hamoud S, Nativ O, Heyman SN, Lupo Rabb H (2003) B cell deficiency confers protection from renal
A, Vlodavsky I, Abassi Z (2016) Heparanase: a potential new ischemia reperfusion injury. J Immunol 171(6):3210–3215. https​
factor involved in the renal epithelial mesenchymal transition ://doi.org/10.4049/jimmu​nol.171.6.3210
(EMT) induced by ischemia/reperfusion (I/R) injury. PLoS ONE 49. Renner B, Strassheim D, Amura CR, Kulik L, Ljubanovic D,
11(7):e0160074. https​://doi.org/10.1371/journ​al.pone.01600​74 Glogowska MJ, Takahashi K, Carroll MC, Holers VM, Thurman
37. Ferenbach DA, Bonventre JV (2015) Mechanisms of maladaptive JM (2010) B cell subsets contribute to renal injury and renal pro-
repair after AKI leading to accelerated kidney ageing and CKD. tection after ischemia/reperfusion. J Immunol 185(7):4393–4400.
Nat Rev Nephrol 11(5):264–276. https​://doi.org/10.1038/nrnep​ https​://doi.org/10.4049/jimmu​nol.09032​39
h.2015.3 50. Cippa PE, Liu J, Sun B, Kumar S, Naesens M, McMahon AP
38. Castellano G, Franzin R, Sallustio F, Stasi A, Banelli B, Romani (2019) A late B lymphocyte action in dysfunctional tissue repair
M, De Palma G, Lucarelli G, Divella C, Battaglia M, Crovace following kidney injury and transplantation. Nat Commun
A, Staffieri F, Grandaliano G, Stallone G, Ditonno P, Cravedi 10(1):1157. https​://doi.org/10.1038/s4146​7-019-09092​-2
P, Cantaluppi V, Gesualdo L (2019) Complement component 51. Clements M, Gershenovich M, Chaber C, Campos-Rivera J,
C5a induces aberrant epigenetic modifications in renal tubular Du P, Zhang M, Ledbetter S, Zuk A (2016) Differential Ly6C
epithelial cells accelerating senescence by Wnt4/betacatenin expression after renal ischemia-reperfusion identifies unique
signaling after ischemia/reperfusion injury. Aging (Albany NY) macrophage populations. J Am Soc Nephrol 27(1):159–170.
11(13):4382–4406. https​://doi.org/10.18632​/aging​.10205​9 https​://doi.org/10.1681/asn.20141​11138​
39. Hu MC, Shi M, Gillings N, Flores B, Takahashi M, Kuro OM, 52. Baek JH (2019) The impact of versatile macrophage functions
Moe OW (2017) Recombinant alpha-Klotho may be prophylactic on acute kidney injury and its outcomes. Front Physiol 10:1016.
and therapeutic for acute to chronic kidney disease progression https​://doi.org/10.3389/fphys​.2019.01016​
and uremic cardiomyopathy. Kidney Int 91(5):1104–1114. https​ 53. Heymann F, Meyer-Schwesinger C, Hamilton-Williams EE,
://doi.org/10.1016/j.kint.2016.10.034 Hammerich L, Panzer U, Kaden S, Quaggin SE, Floege J, Grone
40. Castellano G, Intini A, Stasi A, Divella C, Gigante M, Pontrelli HJ, Kurts C (2009) Kidney dendritic cell activation is required
P, Franzin R, Accetturo M, Zito A, Fiorentino M, Montinaro for progression of renal disease in a mouse model of glomerular

13
Journal of Nephrology

injury. J Clin Invest 119(5):1286–1297. https:​ //doi.org/10.1172/ protein 1 (Keap1) causes hydronephrosis in mice. BMC Nephrol
jci38​399 17(1):110. https​://doi.org/10.1186/s1288​2-016-0310-y
54. Snelgrove SL, Kausman JY, Lo C, Lo C, Ooi JD, Coates PT, 70. Suzuki T, Seki S, Hiramoto K, Naganuma E, Kobayashi EH,
Hickey MJ, Holdsworth SR, Kurts C, Engel DR, Kitching AR Yamaoka A, Baird L, Takahashi N, Sato H, Yamamoto M (2017)
(2012) Renal dendritic cells adopt a pro-inflammatory phenotype Hyperactivation of Nrf2 in early tubular development induces
in obstructive uropathy to activate T cells but do not directly nephrogenic diabetes insipidus. Nat Commun 8:14577. https​://
contribute to fibrosis. Am J Pathol 180(1):91–103. https​://doi. doi.org/10.1038/ncomm​s1457​7
org/10.1016/j.ajpat​h.2011.09.039 71. Noel S, Lee SA, Sadasivam M, Hamad ARA, Rabb H (2018)
55. Kitching AR (2014) Dendritic cells in progressive renal dis- KEAP1 editing using CRISPR/Cas9 for therapeutic NRF2 activa-
ease: some answers, many questions. Nephrol Dial Transpl tion in primary human T lymphocytes. J Immunol 200(5):1929–
29(12):2185–2193. https​://doi.org/10.1093/ndt/gfu07​6 1936. https​://doi.org/10.4049/jimmu​nol.17008​12
56. Rees A (2009) Cross dendritic cells anger T cells after kidney 72. Deregibus MC, Cantaluppi V, Calogero R, Lo Iacono M, Tetta C,
injury. J Am Soc Nephrol 20(1):3–5. https​://doi.org/10.1681/ Biancone L, Bruno S, Bussolati B, Camussi G (2007) Endothe-
asn.20081​11200​ lial progenitor cell derived microvesicles activate an angiogenic
57. Dong Y, Yang M, Zhang J, Peng X, Cheng J, Cui T, Du J (2016) program in endothelial cells by a horizontal transfer of mRNA.
Depletion of CD8+ T cells exacerbates CD4+ T cell-induced Blood 110(7):2440–2448. https​://doi.org/10.1182/blood​-2007-
monocyte-to-fibroblast transition in renal fibrosis. J Immunol 03-07870​9
196(4):1874–1881. https​://doi.org/10.4049/jimmu​nol.15012​32 73. Kim H, Kim D, Nam H, Moon S, Kwon YJ, Lee JB (2019) Engi-
58. Meng XM, Nikolic-Paterson DJ, Lan HY (2014) Inflammatory neered extracellular vesicles and their mimetics for clinical trans-
processes in renal fibrosis. Nat Rev Nephrol 10(9):493–503. https​ lation. Methods. https​://doi.org/10.1016/j.ymeth​.2019.10.005
://doi.org/10.1038/nrnep​h.2014.114 74. Cantaluppi V, Gatti S, Medica D, Figliolini F, Bruno S, Deregi-
59. Bradding P, Pejler G (2018) The controversial role of mast bus MC, Sordi A, Biancone L, Tetta C, Camussi G (2012)
cells in fibrosis. Immunol Rev 282(1):198–231. https​://doi. Microvesicles derived from endothelial progenitor cells protect
org/10.1111/imr.12626​ the kidney from ischemia-reperfusion injury by microRNA-
60. Mehrotra P, Collett JA, McKinney SD, Stevens J, Ivancic CM, dependent reprogramming of resident renal cells. Kidney Int
Basile DP (2017) IL-17 mediates neutrophil infiltration and renal 82(4):412–427. https​://doi.org/10.1038/ki.2012.105
fibrosis following recovery from ischemia reperfusion: compen- 75. Liu C, Wang J, Hu J, Fu B, Mao Z, Zhang H, Cai G, Chen X,
satory role of natural killer cells in athymic rats. Am J Physiol Sun X (2020) Extracellular vesicles for acute kidney injury in
Renal Physiol 312(3):F385–F397. https​://doi.org/10.1152/ajpre​ preclinical rodent models: a meta-analysis. Stem Cell Res Ther
nal.00462​.2016 11(1):11. https​://doi.org/10.1186/s1328​7-019-1530-4
61. Cortvrindt C, Speeckaert R, Moerman A, Delanghe JR, Speec- 76. Wang C, Zhu G, He W, Yin H, Lin F, Gou X, Li X (2019)
kaert MM (2017) The role of interleukin-17A in the pathogen- BMSCs protect against renal ischemia-reperfusion injury by
esis of kidney diseases. Pathology 49(3):247–258. https​://doi. secreting exosomes loaded with miR-199a-5p that target BIP to
org/10.1016/j.patho​l.2017.01.003 inhibit endoplasmic reticulum stress at the very early reperfu-
62. Wang H, Gao M, Li J, Sun J, Wu R, Han D, Tan J, Wang J, sion stages. FASEB J 33(4):5440–5456. https​://doi.org/10.1096/
Wang B, Zhang L, Dong Y (2019) MMP-9-positive neutro- fj.20180​1821R​
phils are essential for establishing profibrotic microenviron- 77. Pan T, Jia P, Chen N, Fang Y, Liang Y, Guo M, Ding X (2019)
ment in the obstructed kidney of UUO mice. Acta Physiol (Oxf) Delayed remote ischemic preconditioning confersrenoprotection
227(2):e13317. https​://doi.org/10.1111/apha.13317​ against septic acute kidney injury via exosomal miR-21. Thera-
63. D’Alessio FR, Kurzhagen JT, Rabb H (2019) Reparative T lym- nostics 9(2):405–423. https​://doi.org/10.7150/thno.29832​
phocytes in organ injury. J Clin Invest 129(7):2608–2618. https​ 78. Wu X, Yan T, Wang Z, Wu X, Cao G, Zhang C, Tian X, Wang
://doi.org/10.1172/jci12​4614 J (2018) Micro-vesicles derived from human Wharton’s Jelly
64. Kim J, Seok YM, Jung KJ, Park KM (2009) Reactive oxygen spe- mesenchymal stromal cells mitigate renal ischemia-reperfusion
cies/oxidative stress contributes to progression of kidney fibrosis injury in rats after cardiac death renal transplantation. J Cell Bio-
following transient ischemic injury in mice. Am J Physiol Renal chem 119(2):1879–1888. https​://doi.org/10.1002/jcb.26348​
Physiol 297(2):F461–470. https:​ //doi.org/10.1152/ajpren​ al.90735​ 79. Vinas JL, Spence M, Gutsol A, Knoll W, Burger D, Zimpel-
.2008 mann J, Allan DS, Burns KD (2018) Receptor-ligand interac-
65. Stenvinkel P, Meyer CJ, Block GA, Chertow GM, Shiels PG tion mediates targeting of endothelial colony forming cell-
(2019) Understanding the role of the cytoprotective transcription derived exosomes to the kidney after ischemic injury. Sci Rep
factor nuclear factor erythroid 2-related factor 2-lessons from 8(1):16320. https​://doi.org/10.1038/s4159​8-018-34557​-7
evolution, the animal kingdom and rare progeroid syndromes. 80. Dominguez JM 2nd, Dominguez JH, Xie D, Kelly KJ (2018)
Nephrol Dial Transpl. https​://doi.org/10.1093/ndt/gfz12​0 Human extracellular microvesicles from renal tubules reverse
66. Nezu M, Suzuki N, Yamamoto M (2017) Targeting the KEAP1- kidney ischemia-reperfusion injury in rats. PLoS ONE
NRF2 system to prevent kidney disease progression. Am J Neph- 13(8):e0202550. https​://doi.org/10.1371/journ​al.pone.02025​50
rol 45(6):473–483. https​://doi.org/10.1159/00047​5890 81. Zhang G, Yang Y, Huang Y, Zhang L, Ling Z, Zhu Y, Wang F,
67. Nezu M, Souma T, Yu L, Suzuki T, Saigusa D, Ito S, Suzuki N, Zou X, Chen M (2017) Hypoxia-induced extracellular vesicles
Yamamoto M (2017) Transcription factor Nrf2 hyperactivation mediate protection of remote ischemic preconditioning for renal
in early-phase renal ischemia-reperfusion injury prevents tubu- ischemia-reperfusion injury. Biomed Pharmacother 90:473–478.
lar damage progression. Kidney Int 91(2):387–401. https​://doi. https​://doi.org/10.1016/j.bioph​a.2017.03.096
org/10.1016/j.kint.2016.08.023 82. Wang B, Jia H, Zhang B, Wang J, Ji C, Zhu X, Yan Y, Yin
68. Noel S, Martina MN, Bandapalle S, Racusen LC, Potteti HR, L, Yu J, Qian H, Xu W (2017) Pre-incubation with hucMSC-
Hamad AR, Reddy SP, Rabb H (2015) T lymphocyte-specific exosomes prevents cisplatin-induced nephrotoxicity by activating
activation of Nrf2 protects from AKI. J Am Soc Nephrol autophagy. Stem Cell Res Ther 8(1):75. https​://doi.org/10.1186/
26(12):2989–3000. https​://doi.org/10.1681/asn.20141​00978​ s1328​7-016-0463-4
69. Noel S, Arend LJ, Bandapalle S, Reddy SP, Rabb H (2016) Kid- 83. Ranghino A, Bruno S, Bussolati B, Moggio A, Dimuccio V,
ney epithelium specific deletion of kelch-like ECH-associated Tapparo M, Biancone L, Gontero P, Frea B, Camussi G (2017)

13
Journal of Nephrology

The effects of glomerular and tubular renal progenitors and 95. Burger D, Vinas JL, Akbari S, Dehak H, Knoll W, Gutsol A,
derived extracellular vesicles on recovery from acute kidney Carter A, Touyz RM, Allan DS, Burns KD (2015) Human
injury. Stem Cell Res Ther 8(1):24. https:​ //doi.org/10.1186/s1328​ endothelial colony-forming cells protect against acute kidney
7-017-0478-5 injury: role of exosomes. Am J Pathol 185(8):2309–2323. https​
84. Dominguez JH, Liu Y, Gao H, Dominguez JM 2nd, Xie D, ://doi.org/10.1016/j.ajpat​h.2015.04.010
Kelly KJ (2017) Renal tubular cell-derived extracellular vesicles 96. Zou X, Zhang G, Cheng Z, Yin D, Du T, Ju G, Miao S, Liu G,
accelerate the recovery of established renal ischemia reperfu- Lu M, Zhu Y (2014) Microvesicles derived from human Whar-
sion injury. J Am Soc Nephrol 28(12):3533–3544. https​://doi. ton’s Jelly mesenchymal stromal cells ameliorate renal ischemia-
org/10.1681/asn.20161​21278​ reperfusion injury in rats by suppressing CX3CL1. Stem Cell Res
85. Bruno S, Tapparo M, Collino F, Chiabotto G, Deregibus MC, Ther 5(2):40. https​://doi.org/10.1186/scrt4​28
Soares Lindoso R, Neri F, Kholia S, Giunti S, Wen S, Quesen- 97. Zhang G, Zou X, Miao S, Chen J, Du T, Zhong L, Ju G, Liu G,
berry P, Camussi G (2017) Renal regenerative potential of differ- Zhu Y (2014) The anti-oxidative role of micro-vesicles derived
ent extracellular vesicle populations derived from bone marrow from human Wharton-Jelly mesenchymal stromal cells through
mesenchymal stromal cells. Tissue Eng Part A 23(21–22):1262– NOX2/gp91(phox) suppression in alleviating renal ischemia-
1273. https​://doi.org/10.1089/ten.TEA.2017.0069 reperfusion injury in rats. PLoS ONE 9(3):e92129. https​://doi.
86. Zou X, Gu D, Zhang G, Zhong L, Cheng Z, Liu G, Zhu Y (2016) org/10.1371/journ​al.pone.00921​29
NK cell regulatory property is involved in the protective role of 98. Herrera Sanchez MB, Bruno S, Grange C, Tapparo M, Can-
msc-derived extracellular vesicles in renal ischemic reperfusion taluppi V, Tetta C, Camussi G (2014) Human liver stem cells
injury. Hum Gene Ther 27(11):926–935. https:​ //doi.org/10.1089/ and derived extracellular vesicles improve recovery in a murine
hum.2016.057 model of acute kidney injury. Stem Cell Res Ther 5(6):124. https​
87. Zou X, Gu D, Xing X, Cheng Z, Gong D, Zhang G, Zhu Y ://doi.org/10.1186/scrt5​14
(2016) Human mesenchymal stromal cell-derived extracellular 99. Choi HY, Moon SJ, Ratliff BB, Ahn SH, Jung A, Lee M, Lee S,
vesicles alleviate renal ischemic reperfusion injury and enhance Lim BJ, Kim BS, Plotkin MD, Ha SK, Park HC (2014) Micro-
angiogenesis in rats. Am J Transl Res 8(10):4289–4299 particles from kidney-derived mesenchymal stem cells act as
88. Zhang G, Zou X, Huang Y, Wang F, Miao S, Liu G, Chen M, Zhu carriers of proangiogenic signals and contribute to recovery
Y (2016) Mesenchymal stromal cell-derived extracellular vesi- from acute kidney injury. PLoS ONE 9(2):e87853. https​://doi.
cles protect against acute kidney injury through anti-oxidation org/10.1371/journ​al.pone.00878​53
by enhancing Nrf2/ARE activation in rats. Kidney Blood Press 100. Zhou Y, Xu H, Xu W, Wang B, Wu H, Tao Y, Zhang B, Wang M,
Res 41(2):119–128. https​://doi.org/10.1159/00044​3413 Mao F, Yan Y, Gao S, Gu H, Zhu W, Qian H (2013) Exosomes
89. Vinas JL, Burger D, Zimpelmann J, Haneef R, Knoll W, Camp- released by human umbilical cord mesenchymal stem cells pro-
bell P, Gutsol A, Carter A, Allan DS, Burns KD (2016) Transfer tect against cisplatin-induced renal oxidative stress and apoptosis
of microRNA-486-5p from human endothelial colony forming in vivo and in vitro. Stem Cell Res Ther 4(2):34. https​://doi.
cell-derived exosomes reduces ischemic kidney injury. Kidney org/10.1186/scrt1​94
Int 90(6):1238–1250. https​://doi.org/10.1016/j.kint.2016.07.015 101. Kilpinen L, Impola U, Sankkila L, Ritamo I, Aatonen M,
90. Shen B, Liu J, Zhang F, Wang Y, Qin Y, Zhou Z, Qiu J, Fan Y Kilpinen S, Tuimala J, Valmu L, Levijoki J, Finckenberg P, Sil-
(2016) CCR2 positive exosome released by mesenchymal stem jander P, Kankuri E, Mervaala E, Laitinen S (2013) Extracellu-
cells suppresses macrophage functions and alleviates ischemia/ lar membrane vesicles from umbilical cord blood-derived MSC
reperfusion-induced renal injury. Stem Cells Int 2016:1240301. protect against ischemic acute kidney injury, a feature that is lost
https​://doi.org/10.1155/2016/12403​01 after inflammatory conditioning. J Extracell Vesicles. https:​ //doi.
91. Lin KC, Yip HK, Shao PL, Wu SC, Chen KH, Chen YT, Yang org/10.3402/jev.v2i0.21927​
CC, Sun CK, Kao GS, Chen SY, Chai HT, Chang CL, Chen CH, 102. Bruno S, Grange C, Collino F, Deregibus MC, Cantaluppi V,
Lee MS (2016) Combination of adipose-derived mesenchymal Biancone L, Tetta C, Camussi G (2012) Microvesicles derived
stem cells (ADMSC) and ADMSC-derived exosomes for protect- from mesenchymal stem cells enhance survival in a lethal model
ing kidney from acute ischemia-reperfusion injury. Int J Cardiol of acute kidney injury. PLoS ONE 7(3):e33115. https​://doi.
216:173–185. https​://doi.org/10.1016/j.ijcar​d.2016.04.061 org/10.1371/journ​al.pone.00331​15
92. Gu D, Zou X, Ju G, Zhang G, Bao E, Zhu Y (2016) Mesenchymal 103. Gatti S, Bruno S, Deregibus MC, Sordi A, Cantaluppi V, Tetta
stromal cells derived extracellular vesicles ameliorate acute renal C, Camussi G (2011) Microvesicles derived from human adult
ischemia reperfusion injury by inhibition of mitochondrial fis- mesenchymal stem cells protect against ischaemia-reperfusion-
sion through miR-30. Stem Cells Int 2016:2093940. https​://doi. induced acute and chronic kidney injury. Nephrol Dial Transpl
org/10.1155/2016/20939​40 26(5):1474–1483. https​://doi.org/10.1093/ndt/gfr01​5
93. de Almeida DC, Bassi EJ, Azevedo H, Anderson L, Origassa 104. Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S,
CS, Cenedeze MA, de Andrade-Oliveira V, Felizardo RJ, da Collino F, Morando L, Busca A, Falda M, Bussolati B, Tetta C,
Silva RC, Hiyane MI, Semedo P, Dos Reis MA, Moreira-Filho Camussi G (2009) Mesenchymal stem cell-derived microvesi-
CA, Verjovski-Almeida S, Pacheco-Silva A, Camara NO (2016) cles protect against acute tubular injury. J Am Soc Nephrol
A regulatory miRNA-mRNA network is associated with tissue 20(5):1053–1067. https​://doi.org/10.1681/asn.20080​70798​
repair induced by mesenchymal stromal cells in acute kidney 105. Cantaluppi V, Medica D, Mannari C, Stiaccini G, Figliolini F,
injury. Front Immunol 7:645. https​://doi.org/10.3389/fimmu​ Dellepiane S, Quercia AD, Migliori M, Panichi V, Giovannini
.2016.00645​ L, Bruno S, Tetta C, Biancone L, Camussi G (2015) Endothelial
94. Ju GQ, Cheng J, Zhong L, Wu S, Zou XY, Zhang GY, Gu D, progenitor cell-derived extracellular vesicles protect from com-
Miao S, Zhu YJ, Sun J, Du T (2015) Microvesicles derived from plement-mediated mesangial injury in experimental anti-Thy1.1
human umbilical cord mesenchymal stem cells facilitate tubu- glomerulonephritis. Nephrol Dial Transpl 30(3):410–422. https​
lar epithelial cell dedifferentiation and growth via hepatocyte ://doi.org/10.1093/ndt/gfu36​4
growth factor induction. PLoS ONE 10(3):e0121534. https:​ //doi. 106. Hodgson LE, Selby N, Huang TM, Forni LG (2019) The role of
org/10.1371/journ​al.pone.01215​34 risk prediction models in prevention and management of AKI.

13
Journal of Nephrology

Semin Nephrol 39(5):421–430. https​://doi.org/10.1016/j.semne​ a prospective time series study. Nephron 131(1):43–50. https​://
phrol​.2019.06.002 doi.org/10.1159/00043​8871
107. Sykes L, Nipah R, Kalra P, Green D (2018) A narrative review 112. Silver SA, Siew ED (2017) Follow-up care in acute kidney injury:
of the impact of interventions in acute kidney injury. J Nephrol lost in transition. Adv Chronic Kidney Dis 24(4):246–252. https​
31(4):523–535. https​://doi.org/10.1007/s4062​0-017-0454-2 ://doi.org/10.1053/j.ackd.2017.05.008
108. Malhotra R, Kashani KB, Macedo E, Kim J, Bouchard J, Wynn 1 13. Harel Z, Wald R, Bargman JM, Mamdani M, Etchells E, Garg
S, Li G, Ohno-Machado L, Mehta R (2017) A risk prediction AX, Ray JG, Luo J, Li P, Quinn RR, Forster A, Perl J, Bell CM
score for acute kidney injury in the intensive care unit. Nephrol (2013) Nephrologist follow-up improves all-cause mortality of
Dial Transpl 32(5):814–822. https​://doi.org/10.1093/ndt/gfx02​6 severe acute kidney injury survivors. Kidney Int 83(5):901–908.
109. Abdelaziz TS, Fouda R, Hussin WM, Elyamny MS, Abdelha- https​://doi.org/10.1038/ki.2012.451
mid YM (2020) Preventing acute kidney injury and improving 114. KDIGO (2012) KDIGO clinical practice guideline for the man-
outcome in critically ill patients utilizing risk prediction score agement of blood pressure in chronic kidney disease. https​://
(PRAIOC-RISKS) study. A prospective controlled trial of AKI kdigo​.org/wp-conte​nt/uploa​ds/2016/10/KDIGO​-2012-Blood​
prevention. J Nephrol 33(2):325–334. https​://doi.org/10.1007/ -Press​ure-Guide​line-Engli​sh.pdf (online im Internet)
s4062​0-019-00671​-6
110. Zhang WR, Parikh CR (2019) Biomarkers of acute and chronic Publisher’s Note Springer Nature remains neutral with regard to
kidney disease. Annu Rev Physiol 81:309–333. https​://doi. jurisdictional claims in published maps and institutional affiliations.
org/10.1146/annur​ev-physi​ol-02051​8-11460​5
111. Silver SA, Harel Z, Harvey A, Adhikari NK, Slack A, Acedillo
R, Jain AK, Richardson RM, Chan CT, Chertow GM, Bell
CM, Wald R (2015) Improving care after acute kidney injury:

13

You might also like