Sato 2020

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Pathophysiology of AKI to CKD progression

Yuki Sato, MD, PhD *,† Masahiro Takahashi, MD † and Motoko Yanagita, MD, PhD†,‡

Summary: Acute kidney injury (AKI), defined as a rapid decrease in glomerular filtration rate, is a common and dev-
astating pathologic condition. AKI is associated with significant morbidity and subsequent chronic kidney disease
(CKD) development. Regardless of the initial insult, CKD progression after AKI involves multiple types of cells,
including proximal tubular cells, fibroblasts, and immune cells. Although the mechanisms underlying this AKI to
CKD progression have been investigated extensively over the past decade, therapeutic strategies still are lacking.
One of the reasons for this stems from the fact that AKI and its progression toward CKD is multifactorial and variable
because it is dependent on patient background. In this review, we describe the current understanding of AKI and its
maladaptive repair with a focus on proximal tubules and resident fibroblasts. Subsequently, we discuss the unique
pathophysiology of AKI in the elderly, highlighting our recent finding of age-dependent tertiary lymphoid tissues.
Semin Nephrol 40:206−215 Ó 2020 Elsevier Inc. All rights reserved.
Keywords: Acute kidney injury, proximal tubule, fibroblast, tertiary lymphoid tissues, chronic inflammation

A
cute kidney injury (AKI) is a common patho- (CKD) or end-stage renal disease (ESRD). Bucaloiu
logic condition characterized by a steep decrease et al4 reported that approximately 50% of hospital-asso-
in glomerular filtration.1 AKI is caused by vari- ciated AKI patients who had recovered successfully
ous common pathologic conditions, including dehydra- from AKI and were discharged were newly diagnosed
tion and sepsis. The incidence of AKI has increased over with CKD during the median follow-up period of
past decades.2 One study reported that the prevalence of 3.3 years. In another cohort from North America, the rel-
non−dialysis-requiring AKI was more than 5,000 cases ative hazard risk of chronic dialysis in patients who had
per million per year.3 Moreover, a significant proportion recovered from dialysis-requiring AKI was 3.23 com-
of AKI survivors progressed to chronic kidney disease pared with controls. Thus, AKI now is recognized as a
significant risk factor for CKD and ESRD.5
Although a number of molecules and pathways have
*Medical Innovation Center, TMK Project, Kyoto University, Kyoto, been identified as involved in AKI and its progression by
Japan animal studies over past decades, the development of
yDepartment of Nephrology, Graduate School of Medicine, Kyoto effective treatment strategies for this AKI to CKD transi-
University, Kyoto, Japan
tion in human beings still is lacking. One of the reasons
zInstitute for the Advanced Study of Human Biology, Kyoto Univer-
sity, Kyoto, Japan for this is the heterogeneity of AKI. AKI occurs in
Financial support: Supported by the Japan Agency for Medical patients from heterogeneous backgrounds with age,
Research and Development under grants 19gm1210009 (Agency for baseline renal function, and accompanying comorbid-
Medical Research and Development−Core Research for Evolutional ities. This may modify the common pathophysiology of
Science and Technology), JP19gm5010002, and JP19gm0610011
AKI and contribute to its heterogeneity. Importantly, an
(M.Y.); by grants from the TMK Project, Grant-in-Aids for Scientific
Research B (26293202, 17H04187), a Grant in Aid for Scientific increasing number of epidemiologic studies have shown
Research on Innovative Areas “Stem Cell Aging and Disease” from that aging and CKD significantly increases the risk of
the Ministry of Education, Culture, Sports, Science and Technology AKI development and its severity.6-8
of Japan (17H05642), a Grant-in-Aid for Young Scientists (B) from In this review, we discuss the pathophysiology of AKI
the Japan Society for the Promotion of Science, the Translational
and its progression to CKD, focusing on the mechanisms
Research program, Strategic Promotion for practical application of
INnovative medical Technology from the Agency for Medical underlying maladaptive repair after renal tubule injury.
Research and Development, grants from the Uehara Memorial Foun- We also discuss the role of tertiary lymphoid tissues
dation, Takeda Science Foundation, and the Sumitomo Foundation. (TLT) after AKI and their potential influences on mal-
This work also was supported in part by the World Premier Interna- adaptive repair.
tional Research Center Initiative, Ministry of Education, Culture,
Sports, Science and Technology, Japan.
Conflict of interest statement: Yuki Sato is employed by the TMK Proj-
ect; and Motoko Yanagita has received research grants from Astel- PROXIMAL TUBULES AS A PRIMARY TARGET OF
las, Chugai, Daiichi Sankyo, Fujiyakuhin, Kyowa Hakko Kirin, AKI
Mitsubishi Tanabe, MSD, Nippon Boehringer Ingelheim, and Torii.
Address reprint requests to Motoko Yanagita, MD, Department of The kidney is composed of multiple types of differenti-
Nephrology, Graduate School of Medicine, Kyoto University, 54 ated cells with unique characteristics and is responsible
Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan. E-mail: for the maintenance of internal homeostasis. Proximal
motoy@kuhp.kyoto-u.ac.jp
0270-9295/ - see front matter tubules are the most popular cell type in the kidney,
© 2020 Elsevier Inc. All rights reserved. occupying almost 90% of the renal cortex. Proximal
https://doi.org/10.1016/j.semnephrol.2020.01.011 tubular cells express various transporters and channels

206 Seminars in Nephrology, Vol 40, No 2, March 2020, pp 206−215


AKI progression to CKD 207

on the apical surface and reabsorb various vital substan- with limitations.17,18 A precise understanding of the cel-
ces, including sodium, bicarbonate, glucose, and amino lular and molecular mechanisms for intrinsic repair is
acids in the glomerular filtrate. In addition, proximal critically important because strategies to harness intrinsic
tubular cells reabsorb intrinsic and extrinsic macromole- renal repair have the potential to become novel therapeu-
cules through receptor-mediated endocytosis and metab- tic options for AKI to CKD progression.
olize various substances.9 Interestingly, proximal tubules After injury, tubular cells dedifferentiate, proliferate,
mostly depend on fatty acids as their energy source and and redifferentiate, leading to the repair of nephrons.
generate adenosine triphosphate (ATP) by mitochondrial Many investigators have examined the origin of the
b-oxidation. Proximal tubules therefore are rich in the tubular cells that replenish the epithelial cells after AKI
mitochondria used to perform the task described earlier. in murine models. Humphreys et al19 used Six2-Cre
Although renal proximal tubules are essential for mice to genetically label almost all of the epithelial cells
maintaining internal homeostasis, they also are suscepti- of nephrons. They found that despite extensive cell pro-
ble to various kinds of cellular stress and are considered liferation after ischemic-reperfusion injury, no dilution
a main target of AKI.10 For instance, proximal tubular of genetic labels was observed after the repair phase.20
cells are vulnerable to ischemic stress. In the outer These results indicated that surviving epithelial cells
medulla of the kidney, S3 segments of proximal tubules largely contribute to the repair of proximal tubules after
are particularly susceptible to ischemic AKI because of AKI. Subsequently, using Slc34a1-CreERT2, which
their huge burden of sodium reabsorption and the physio- labels proximal tubular cells more specifically, the same
logically hypoxic microenvironment. Importantly, proxi- group further confirmed that injured proximal tubules
mal tubules have limited glycolytic activity, which also were regenerated mainly through their own prolifera-
renders them highly susceptible to ischemic insult.10 In tion.21 The Ndrg1-CreERT2 mouse line, which also
addition, proximal tubules are highly susceptible to labels proximal tubule cells, showed consistent results.13
nephrotoxic substances such as anticancer drugs. For Interestingly, we found that proximal tubules became
instance, cisplatin, a platinum-based alkylating antineo- shorter after AKI than before, indicating the limitation
plastic agent, is taken up by proximal tubules through of the regenerative capacity of proximal tubules.13 These
both via passive diffusion and active transport, which is repair processes were exemplified elegantly by a recent
mediated mainly by organic cation transporter 2 on the study using the intravital imaging technique. In response
basolateral side of proximal tubules. Cisplatin is concen- to laser-induced tubular injury, intact adjacent proximal
trated within the proximal cells and induces proximal tubules proliferate and replace the lost cells. Interest-
tubule injury in a dose-dependent manner.11 ingly, during this process, platelet-derived growth factor
Interestingly, genetic proximal tubule−specific injury (PDGF)-receptor b−positive resident fibroblasts migrate
shows several CKD features. By using the inducible to the injured site and promote tubular regeneration in a
diphtheria toxin receptor system, which induces injury in PDGFb-dependent manner22 because inhibition of
the cells expressing Cre recombinase by the administra- PDGFb hindered the fibroblast migration and tubular
tion of diphtheria toxin, Grgic et al12 showed that Six2- regeneration. These results showed that resident fibro-
Cre lineage renal epithelial cell injury resulted in intersti- blasts are motile and contribute to tubular regeneration
tial fibrosis and glomerulosclerosis. By using by acting as a mechanical stabilization of the denuded
Ndrg1CreERT2 mice,13 which label proximal tubules, tubular basement membrane. We also recently reported
we also showed that proximal tubule−specific injury that although a-smooth muscle actin (aSMA)-positive
leads to not only renal fibrosis and reduced Epo produc- myofibroblasts generally are accepted as a driver of renal
tion, but also glomerulosclerosis and atubular glomer- fibrosis, in the early phase of kidney injury, myofibro-
uli.14 These studies showed that proximal tubular injury blasts acquire retinoic acid−producing ability and are
is both a primary trigger of AKI as well as a potential actively involved in the repair process of tubular cells,
determinant of later disease progression, and that the possibly by promoting tubular cell proliferation.23
protection of proximal tubules is essential to preventing
the development of AKI and its progression to CKD.15
Do All Proximal Tubular Cells Have a Similar
Capacity for Regeneration?
INTRINSIC REGENERATIVE POTENTIAL OF
Although the elegant lineage tracing studies described
PROXIMAL TUBULE CELLS earlier showed that surviving proximal tubular cells are
Although proximal tubules are susceptible to various cel- the origin of renewal proximal tubular cells after injury,
lular stresses, they also have an intrinsic repair capacity, this did not exclude the possibility of an intratubular
which minimizes damage and circumvents serious renal stem cell. It remains controversial whether there are any
dysfunction. A recent clinical study showed that among distinct subpopulations of tubular cells responsible for
2,922 patients with a single episode of dialysis-requiring regeneration after tubular injury, or if regeneration is a
AKI, 13.6% had normalized renal function after 1 year,16 stochastic process that surviving tubular cells participate
indicating that the kidney has the potential to repair itself, in equally.
208 Y. Sato et al.

Recently, Romagnani and colleagues24 addressed this c-Jun N-terminal kinase signaling, resulting in interstitial
question using a lineage tracing technique with Confetti fibrosis.28 Therefore, targeting injured tubules at G2/M
mice. They showed that Pax2-Cre lineage-labeled cells, cell-cycle arrest can be a novel therapeutic target and sev-
which originally reside in the S3 segment in the proximal eral interventions have been documented. For instance,
tubules and distal tubules and are recognized as a puta- the pharmacologic inhibition of p53, a key cell-cycle reg-
tive intratubular progenitor population, were resistant to ulator, as well as histone deacetylase inhibitors, bypassed
cell injury and clonally expand after injury, suggesting G2/M arrest in the injured tubular cells and reduced inter-
the presence of intratubular progenitors. Interestingly, stitial fibrosis.28-30 Other groups have shown that specific
they also found that in response to injury some tubular inhibitors of cyclin-dependent kinases 4/6, a key mediator
cells showed endocycle-mediated hypertrophy, a state in of cell-cycle checkpoint progression from the G1 to S
which cells undergo DNA synthesis without cell divi- phase, also are effective to optimize cell-cycle stages of
sion. In contrast, using sequential labeling with thymi- injured tubules.31,32 In addition, the macrophage migra-
dine analogs, Humphreys et al25 previously addressed tion factor in renal tubular cells also has been shown to
the same question and showed that after ischemic reper- limit inflammation and fibrosis by abrogating cell-cycle
fusion injury, intrinsic repair occurred mainly through arrest in renal tubular cells.33
the self-duplication of surviving tubular epithelial cells,
not by intratubular progenitor cells. Although the reasons
Aberrant Reactivation of the Developmental
for the discrepancy among these studies remain unclear,
the reliability of cell proliferation markers such as Ki67 Pathway Promotes Fibrosis
and proliferating cell nuclear antigen in the context of Reactivation of some developmental signaling pathways,
AKI is questioned and considered one of the reasons.24 such as Wnt, Hedgehog, and Notch, have been shown in
To minimize the risk of renal dysfunction, it is reason- animal models of AKI and renal fibrosis. These activa-
able to think that kidneys have multiple mechanisms for tions likely are induced as part of tubular regeneration
tissue repair that can vary depending on several factors, after injury and involved in maladaptive repair.18 For
such as disease severity and the initial trigger. Further example, injured tubules express sonic hedgehog, the
studies in various contexts therefore are required to under- most studied ligand of the hedgehog pathway, in response
stand the details of mechanisms for intrinsic renal repair. to injury, and they secrete sonic hedgehog, which binds to
its receptors on interstitial fibroblasts and up-regulates
their expression of profibrotic genes, such as fibronectin
INJURED PROXIMAL TUBULE AS A DRIVER OF AKI and collagen.34 The Wnt/b-catenin pathway, another
TO CKD PROGRESSION important developmental pathway, also is reactivated in
Although the kidney has several intrinsic repair capaci- tubular cells during the acute phase of kidney injury,
ties, as described in the previous section, in case of its which potentially is protective because the proximal
failure (eg, chronic kidney injury), these normally effi- tubule−specific deletion of b-catenin showed aggravated
cient repair mechanisms become impaired and several apoptosis and tubular injury in this phase.35 The sustained
additional mechanisms trigger maladaptive repair expression of Wnt ligands, however, induced myofibro-
responses, which lead to CKD progression. Substantial blast transformation and resulted in fibrosis. These studies
evidence has shown that injured proximal tubular cells show the involvement of the reactivation of developmen-
contribute to AKI to CKD progression by promoting tal signaling pathways in repair processes after injury,
inflammation and fibrosis.8,26,27 Here, we describe sev- which eventually can result in maladaptive repair.
eral pathologic roles of injured proximal tubules that
underlie the AKI to CKD progression.
Mitochondrial Dysfunction in Injured Proximal
Tubular Cells
Injured Proximal Tubules in G2/M Cell-Cycle Arrest Mitochondrial dysfunction increasingly has been recog-
Promote Renal Inflammation and Fibrosis nized as a critical contributor of AKI to CKD progres-
Over the past decade, various studies have shown the sion.36 Although mitochondria are necessary for several
humoral communications between injured tubular cells cellular functions, such as ATP synthesis under physio-
and interstitial cells, and their roles in regeneration and logical conditions, during injury, they also hold several
fibrosis after injury. Bonventre and colleagues reported detrimental roles for cells, such as the production of
that severe AKI results in tubular cell-cycle arrest at the reactive oxygen species and the induction of apoptosis.37
G2/M phase of the cell cycle, which secrete various profi- Recent studies have shown that most AKIs include vari-
brotic and proinflammatory factors.26,28 Transforming ous degrees of mitochondria dysfunction in tubular epi-
growth factor-b and connective tissue growth factor are thelial cells.
well-investigated profibrotic and proinflammatory factors Mitochondrial injury occurs early in the course of
known to be secreted by injured proximal tubular cells in AKI, especially ischemia-reperfusion injury, and is asso-
G2/M cell-cycle arrest,26 at least partially mediated by ciated with cellular injury. Under ischemic conditions,
AKI progression to CKD 209

hypoxia disrupts oxidative phosphorylation in mitochon-


dria and causes excessive reactive oxygen species pro-
duction and ATP deficiency, which further damages
various molecules, including DNAs, proteins, and lipids,
resulting in cell death and inflammation. Damaged mito-
chondria also release several danger molecules, such as
DNA and cardiolipin, leading to NOD-, LRR- and pyrin
domain-containing protein 3 inflammasome activation.
Therefore, protecting mitochondrial function is essential
for treating AKI with different etiologies.37,38 It is of
note that mitochondria-specific antioxidant molecules,
such as mitoubiquinone, MitoTEMPO, and SkQR1, also
showed protective effects in experimental models of
ischemic and septic AKI in association with reduced oxi-
dative stress.39-42 Mitochondrial dysfunction also is Figure 1. Renal fibrosis and inflammatory cell infiltration: two com-
involved in CKD progression after AKI. Szeto et al43 mon pathologic features in injured kidney. aSMA-positive myofibro-
blast accumulation and inflammatory cell infiltration in renal
reported that mitochondrial dysfunction was observed in interstitial space are common pathologic findings in murine injured
rat proximal tubules 9 months after ischemic AKI and kidney. Immunofluorescence of CD45 (green) and aSMA (red)
that the late administration of mitochondrial-protective 10 days after unilateral ureteral obstruction. Scale bar: 50 mm.
molecule SS-31 promoted recovery from CKD.
To maintain a healthy cellular state, mitochondria
constantly are being renewed. Damaged mitochondria diverse etiologies and pathophysiology of AKI, patho-
are selectively removed through the process of mitoph- logic changes in the kidney converge on several common
agy, the autophagic degradation of damaged mitochon- CKD features, including interstitial fibrosis, peritubular
dria, and the loss of mitochondrial mass is replenished capillary loss, and renal anemia.47-50 Interstitial fibrosis
by mitochondrial biogenesis. Peroxisome proliferator is a hallmark of CKD and is defined as the accumulation
activated receptor gamma coactivator 1 alpha (PGC-1a) of extracellular matrix (ECM) and is produced mainly
is a master regulator of mitochondria biogenesis and is by aSMA-positive myofibroblasts (Fig. 1). Abnormal
highly expressed in the cortex and outer stripe of the ECM accumulation in the interstitial spaces distorts
medulla where the mitochondrial activity is relatively organ architecture, disturbs blood supply, and decreases
high.44 Interestingly, overexpression of PGC-1a in tubu- renal oxygenation. Peritubular capillary loss and renal
lar cells increased mitochondrial mass and also showed anemia also contribute to chronic hypoxia, the final com-
renoprotection, not cell death, after ischemic and inflam- mon pathway to ESRD.51 Interestingly, interstitial fibro-
matory kidney injury,45 whereas global PGC-1a knock- sis, renal anemia, and peritubular capillary loss are
out mice showed severe renal dysfunction in a model of linked mechanistically, and resident fibroblasts are a cen-
septic AKI.46 Given that the level of PGC-1a has been tral player in these conditions.
shown to decrease after AKI and increase with repair, Resident fibroblasts are spindle-shaped mesenchymal
these results showed that PGC-1a is protective in AKI cells residing in the interstitial space.52 Interestingly, res-
and is necessary for post-AKI repair. Furthermore, inves- ident fibroblasts in the kidney have the ability to sense
tigations into the downstream mechanisms of PGC-1a hypoxia and produce erythropoietin (EPO), thereby play-
have shown that de novo NAD+ biosynthesis mediates ing a crucial role in maintaining internal homeostasis.53
renal protection.45 NAD+ is an essential energy carrier During injury, however, the phenotype of fibroblasts
for fatty acid oxidation in mitochondria; b-hydroxybuty- changes dramatically. In our previous study, we showed
rate, a breakdown product of fatty acid oxidation, indu- that resident fibroblasts in the cortex and the outer
ces prostaglandin E2, which maintains renal function.45 medulla of the kidney are lineage-labeled with P0-Cre,
which labels the migrating neural crest cells. In response
to injury, P0-Cre lineage−labeled fibroblasts transdiffer-
FIBROBLAST DYSFUNCTION IS A CENTRAL entiate into aSMA-positive myofibroblasts, proliferate
dramatically, and execute fibrosis by producing a large
PATHOGENIC PROCESS IN CKD PROGRESSION
amount of ECMs (Fig. 2).48 Recently, Humphreys and
AFTER AKI colleagues showed that Gli1-Cre lineage−labeled cells
Emerging evidence also has shown that a significant are progenitors of myofibroblasts and can be targeted
overlap exists between AKI and CKD pathophysiology, therapeutically.54 They showed that the ablation of Gli1-
in both of which resident fibroblasts play crucial roles, Cre lineage−labeled cells resulted in a more than 50%
suggesting the potentially important contribution of reduction of renal fibrosis in a murine unilateral ureteral
fibroblasts in the AKI to CKD sequence.47 Despite the obstruction model.54
210 Y. Sato et al.

Figure 2. Heterogeneous fibroblasts involved in AKI to CKD progression. Resident fibroblasts in the kidney locally
transdifferentiate into several distinct phenotypic fibroblasts in response to their microenvironmental cues. In response
to injury, resident fibroblasts in the kidney transdifferentiate aSMA-positive myofibroblasts at the cost of EPO-produc-
ing ability and execute renal fibrosis. Some of the myofibroblasts acquire retinoic acid (RA)-producing ability. Although
RA derived from myofibroblasts supports tubular regeneration, in aged injured kidney, RA additionally induces p75NTR
expression in fibroblasts and promotes the transdifferentiation of fibroblasts into TLT-associated fibroblasts. RALDH2,
retinaldehyde dehydrogenase 2. Reprinted with permission from Sato and Yanagita,7,53 Nangaku,51 and Sato et al.76

Importantly, during this phenotypic transition into PHENOTYPIC PLASTICITY AND HETEROGENEITY
myofibroblasts, fibroblasts lose the ability to produce OF IMMUNE CELLS
EPO in response to hypoxia (Fig. 2).48 This loss of EPO-
In the pathophysiology of kidney injury and repair,
producing ability, however, is reversible by several
immune cells have a significant influence on the fate of the
agents, including selective estrogen-receptor modula-
injured kidney.7,59,60 Several types of immune cells, such
tors.48 This is consistent with human clinical data. Broo-
as monocytes and lymphocytes, are recruited to the site of
khart et al55 reported that hemodialysis patients living at
injury and can play important roles in both tissue destruc-
high altitudes had a higher hematocrit with a lower dose
tion and tissue repair. Although inflammation is originally
of recombinant EPO treatment than hemodialysis
a biological response that is necessary for eradicating
patients at lower altitudes, suggesting that hypoxia-
pathogens and promoting tissue repair after injury, excess
driven EPO production remains operative to some
and nonresolving inflammation can lead to tissue damage
extent, even in patients with ESRD. Another example is
and fibrosis. Therefore, the balance between proinflamma-
that the pharmacologic inhibition of prolyl hydroxylase,
tory and anti-inflammatory immune responses has a signif-
which regulates EPO production by degrading hypoxia-
icant impact on the renal outcome after AKI. Here, we
inducible factor 1a in an oxygen-dependent manner,
described the role of immune cells in the pathophysiology
prominently increased intrinsic EPO production in
of AKI to CKD progression.
hemodialysis patients, suggesting the reserve capacity of
fibroblasts for EPO production.56 These clinical studies
indicate that EPO-producing cells still exist, even in
ESRD patients, and that the EPO-producing ability is Roles of Immune Cells in Kidney Injury and Repair
reversible. During the acute phase of AKI, innate immune cells such
In parallel to CKD progression, peritubular capillary as monocytes and neutrophils are recruited in response to
loss also becomes detectable more commonly.57 Although damage-associated molecular patterns released from
pericytes wrap peritubular capillary vessels with their necrotic cells. These recruited cells are activated and
multiple projections and structurally stabilize them under release proinflammatory cytokines, such as interleukin
normal conditions, during injury, pericytes are detached (IL)6 and tumor necrosis factor a, propagating inflamma-
from capillary vessels and instead wrap injured tubules, tory responses. Ischemic insult causes an increase in vas-
which make the vessels structurally unstable and leads to cular permeability and the aberrant activation of adhesion
capillary regression and rarefaction.47,50,58 molecules, both of which also accelerate the infiltration of
AKI progression to CKD 211

immune cells. Lymphocytes also are involved in the patho- experimental study showed that a pre-existing reduction
physiology. The initially activated tissue-resident mononu- in functional nephrons is associated with CKD progres-
clear phagocytes take up antigen and move to the draining sion after AKI.70 Second, the age-dependent decrease in
lymph nodes, where they present antigen to T cells.61 Sub- regenerative capacity may contribute to maladaptive
sequently, activated T cells clonally expand with the effec- repair in the aged kidney. As described in the previous
tor phenotype and migrate to the kidney,62 where they section, surviving proximal tubular cells proliferate after
release proinflammatory cytokines, such as interferon g, injury and repair themselves. With aging, however, the
and promote inflammation. proliferative capacity of the tubular cells is reduced and
In contrast to the detrimental roles of immune cells in several causative factors have been identified. The
the acute phase of injury described earlier, substantial increased expression of zinc-a-glycoprotein, an inhibitor
evidence also has shown the reparative roles of immune of epithelial cell proliferation, in proximal tubules in
cells in the injured kidneys. For instance, while in the aged mice has been shown to be involved in this reduced
early phase of AKI, macrophages show the proinflamma- proliferative capacity.71 Third, age-dependent chronic
tory M1 phenotype and exacerbate kidney injury as inflammation also hinders intrinsic cellular repair after
described earlier, in the recovery phase of AKI, M1 mac- injury and promotes organ damage.72 Aging results in
rophages switch to the anti-inflammatory M2 phenotype, chronic low-grade inflammation in the murine and
which modulates inflammatory responses and promotes human kidney.72 One of the reasons for this is the accu-
tissue repair.63 Similarly, anti-inflammatory T cells in mulation of senescent cells. Senescent cells are defined
the kidney after AKI also have been reported. The most as being in a permanent state of cell-cycle arrest induced
studied T cell involved in renal repair is the by cellular stress; they develop a senescence-associated
CD4+CD25+FoxP3+ regulatory T cell.64 Regulatory T secretory phenotype that entails the release of numerous
cells counteract proinflammatory responses by contact- biologically active molecules, such as IL6 and matrix
dependent mechanisms and by producing anti-inflamma- metalloproteinases.73 Senescent cells contribute to age-
tory IL10, and therefore limit excess inflammation to dependent inflammation by senescence-associated secre-
promote a reparative process. CD4-CD8- ab T cells also tory phenotype,74,75 and also limit the potential for tubu-
were suggested to release anti-inflammatory cytokines lar regeneration after injury because they are unable to
and promote kidney recovery.65 proliferate.

UNIQUE PATHOPHYSIOLOGY OF AKI IN THE Tertiary Lymphoid Tissues as a Cause for Sustained
ELDERLY Inflammation After AKI in the Elderly
AKI and its progression to CKD or ESRD are more Despite the clinical relevance of AKI in the elderly, the
likely to occur in the elderly. In a retrospective cohort mechanism underlying the poor prognosis largely was
study that followed up hospitalized patients older than elusive. To investigate the difference in injury responses
67 years of age, patients with both AKI and CKD had a between young and aged kidneys, we induced several
41-fold increase in the development of ESRD compared kidney injury models in young and aged mice. Consis-
with those with no history of either disease.6 Given that tent with the clinical evidence, although young kidneys
the aging population is growing worldwide and the inci- repair themselves after injury, aged kidneys show renal
dence of AKI also is increasing, it is important to under- fibrosis and sustained inflammation. Unexpectedly, we
stand precisely what occurs in the aged kidney after found that aged kidneys, but not young kidneys, showed
AKI. multiple inflammatory cell aggregates around the
artery.76 The size of aggregates was associated with
impaired renal function and increased expression of
Multiple Factors for Increased Susceptibility and proinflammatory cytokines, such as interferon g and
Severity of AKI in the Elderly tumor necrosis factor a, suggesting a possible contribu-
Multiple factors have been identified as contributors to tion to sustained inflammation after injury in the aged
the increased susceptibility of AKI and its CKD progres- kidney.76 These aggregates are composed mainly of T
sion in the aged kidney.66,67 First, the presence of age- and B lymphocytes, both of which proliferate within the
dependent structural and functional changes may aggregates. This indicates that these inflammatory cell
increase the susceptibility to AKI. Wiggins and col- aggregates are functional and work as tertiary lymphoid
leagues showed that glomerular aging correlates well tissues (TLTs) (Fig. 3).76
with a progressive linear reduction in podocyte density, TLTs are ectopic lymphoid tissues and function as
which accelerates global glomerulosclerosis.68 A recent local sites of lymphocyte activation.77 They can propa-
study showed that the number of nephrons decreases gate local antigen-specific immune responses, and their
with aging.69 It is reasonable to think that nephron loss roles can be either beneficial or detrimental depending
directly confers a higher susceptibility to AKI, and an on the context. For example, in case of infection, TLTs
212 Y. Sato et al.

tubular basement membrane.81 This study also showed


the clonal expansion and ongoing somatic hypermutation
of B cells in TLTs of lupus nephritis.81

Heterogeneous Fibroblasts Orchestrate TLT


Formation
Although TLTs are composed mainly of lymphocytes,
stromal cells (especially fibroblasts) play crucial roles in
the induction and maintenance. In aged injured kidneys,
several fibroblasts with distinct phenotypes are involved
in TLT formation. Most TLTs harbor homeostatic che-
mokines, such as C-X-C motif chemokine ligand 13
(CXCL13) and C-C motif chemokine ligand 19
Figure 3. TLT in aged injured kidney. TLT is composed mainly of T
and B lymphocytes and formed around the artery. Immunofluores- (CCL19), whose transgenic ectopic expression of these
cence of CD3e (green) and B220 (red). Scale bar: 50 mm. chemokines in nonlymphoid tissue induce the develop-
ment of functional TLTs.83,84 We found that fibroblasts
can play protective roles for the host by generating inside TLTs in aged injured kidneys exclusively produce
immune responses to pathogens. In contrast, in the case CXCL13 and CCL19, and attract T cells and B cells,
of autoimmunity, TLTs contribute to sustained inflam- resulting in TLT formation.76 Interestingly, the pheno-
mation and local antibody production,77,78 and are detri- types of fibroblasts inside and outside TLTs totally dif-
mental for the host organ.77 They also are considered to fer. In addition to the ability to produce CXCL13 and
function as niches for tumor progenitor cells78,79 and CCL19, fibroblasts inside TLTs strongly express the p75
pathogenic memory T cells.80 TLTs also are induced in neurotrophin receptor (p75NTR), a neural crest marker.
several CKDs, such as lupus nephritis81 and chronic Interestingly, retinoic acid−producing fibroblasts sur-
rejected transplanted kidneys,82 which are not age- round TLTs in the early phase of TLT formation and
dependent. In an analysis of lupus nephritis, TLTs were induce strong expression of p75NTR in fibroblasts inside
present in more than half of patients, and the existence TLTs.76 Some of the p75NTR-positive fibroblasts inside
of TLTs was associated with the severity of interstitial TLTs lose their p75NTR expression in the later phase of
inflammation and immune complex deposition in the TLT formation and instead express complement receptor

Figure 4. TLT in murine and human kidney. In murine renal TLT, fibroblasts positive for retinaldehyde dehydrogenase
2 (RALDH2) surround TLTs. In contrast, fibroblasts within TLTs are negative for RALDH2 and instead are strongly
positive for p75NTR, a neural crest marker. Some of the p75NTR+ fibroblasts inside TLT acquire the ability to produce
homeostatic chemokine CXCL13 and CCL19, and drive and maintain TLTs. CD21+/p75NTR- follicular dendritic cells
(FDCs) emerge as an integral part of the fibroblast network within TLT, and peripheral lymph node addressin-positive
high endothelial venule (HEV) also develop within TLTs. Although cellular and molecular components of TLT in human
kidneys are similar to those in murine TLT, p75NTR colocalizes with CD21, and RALDH2+ stromal cells surround
CD21+ FDCs in human renal TLTs. Abbreviations: RA, retinoic acid. Reprinted with permission from Sato and Yana-
gita,7,53 Nangaku,51 and Sato et al.76
AKI progression to CKD 213

CD21, a marker for follicular dendritic cells. Follicular research into this field is required, especially in the
dendritic cells are predominantly stromal cells and form elderly, because the world population is aging and the
B-cell follicles, which support germinal center responses pathophysiology of AKI in the elderly is unique. A better
in lymphoid tissues.85 McMahon and colleagues recently understanding of the pathophysiology and heterogeneity
confirmed B cell clonal expansion and autoantibody pro- of AKI to CKD progression in human beings is
duction in aged murine renal TLTs.86,87 Interestingly, necessary and will lead to the development of novel thera-
we performed a lineage-tracing study to determine the peutics to enhance intrinsic repair and suppress maladap-
origin of these distinct phenotypic fibroblasts using P0- tive responses.
Cre mice, and showed that most were lineage-labeled
with P0-Cre, indicating that fibroblasts from a single
ACKNOWLEDGMENTS
lineage locally diversify into these distinct phenotypic
fibroblasts in response to their microenvironmental cues Y.S. and M.T. contributed equally.
(Fig. 2).76
REFERENCES
TLTs as a Novel Therapeutic Target for Kidney
1. Lameire NH, Bagga A, Cruz D, De Maeseneer J, Endre Z, Kellum
Diseases JA, et al. Acute kidney injury: an increasing global concern. Lan-
Importantly, although not specific to TLTs, anti-CD4 cet. 2013;382:170-9.
2. Hsu RK, McCulloch CE, Dudley RA, Lo LJ, Hsu CY. Temporal
monoclonal antibody treatment abolished TLTs and changes in incidence of dialysis-requiring AKI. J Am Soc Neph-
ameliorated renal fibrosis as well as inflammation, sup- rol. 2013;24:37-42.
porting the concept that targeting TLTs is a promising 3. Hsu CY, McCulloch CE, Fan D, Ordo~ nez JD, Chertow GM, Go
therapeutic target for AKI to CKD progression in the AS. Community-based incidence of acute renal failure. Kidney
elderly.76 Consistently, in autoimmunity, the genetic Int. 2007;72:208-12.
4. Bucaloiu ID, Kirchner HL, Norfolk ER, Hartle JE, Perkins
depletion of TLT-associated fibroblasts also impairs RM. Increased risk of death and de novo chronic kidney dis-
TLT formation and reduces lymphocyte activation with ease following reversible acute kidney injury. Kidney Int.
improvement in tissue pathology.88 Age-dependent TLT 2012;81:477-85.
formation also is detectable in the human kidney, and 5. Wald R, Quinn RR, Luo J, Li P, Scales DC, Mamdani MM, et al.
the cellular and molecular components are similar to Chronic dialysis and death among survivors of acute kidney injury
requiring dialysis. JAMA. 2009;302:1179-85.
those in mice (Fig. 4), suggesting that age-dependent 6. Ishani A, Xue JL, Himmelfarb J, Eggers PW, Kimmel PL, Molito-
TLT formation is conserved across species. This similar- ris BA, et al. Acute kidney injury increases risk of ESRD among
ity is critically important because the discrepancy elderly. J Am Soc Nephrol. 2009;20:223-8.
between human and rodent kidney has been suspected as 7. Sato Y, Yanagita M. Immune cells and inflammation in AKI
a reason why numerous agents that had shown promising to CKD progression. Am J Physiol Renal Physiol. 2018;315:
F1501-12.
results in experimental AKI failed to show efficacy in 8. He L, Wei Q, Liu J, Yi M, Liu Y, Liu H, et al. AKI on CKD:
human patients.20,89 heightened injury, suppressed repair, and the underlying mecha-
Although renal TLTs are induced in human kidneys nisms. Kidney Int. 2017;92:1071-83.
by several factors, such as aging and autoimmunity, sev- 9. Nielsen R, Christensen EI, Birn H. Megalin and cubilin in proxi-
eral studies have yielded inconsistent results and the mal tubule protein reabsorption: from experimental models to
human disease. Kidney Int. 2016;89:58-67.
clinical relevance remains to be elucidated. One of the 10. Chevalier RL. The proximal tubule is the primary target of injury
reasons for this is the lack of an established assessment and progression of kidney disease: role of the glomerulotubular
method for TLTs in human kidneys. Further studies are junction. Am J Physiol Renal Physiol. 2016;311:F145-61.
required to understand the nature of TLTs in human kid- 11. Kitai Y, Matsubara T, Yanagita M. Onco-nephrology: current
neys and to develop a TLT assessment method for deter- concepts and future perspectives. Jpn J Clin Oncol. 2015;45:
617-28.
mining the clinical relevance of TLTs in various 12. Grgic I, Campanholle G, Bijol V, Wang C, Sabbisetti VS, Ichi-
pathologic conditions. mura T, et al. Targeted proximal tubule injury triggers interstitial
fibrosis and glomerulosclerosis. Kidney Int. 2012;82:172-83.
13. Endo T, Nakamura J, Sato Y, Asada M, Yamada R, Takase M,
CONCLUSIONS et al. Exploring the origin and limitations of kidney regeneration.
J Pathol. 2015;236:251-63.
Our expanding knowledge on the AKI to CKD progres- 14. Takaori K, Nakamura J, Yamamoto S, Nakata H, Sato Y, Takase
sion from murine models has improved our understanding M, et al. Severity and frequency of proximal tubule injury deter-
of this pathologic sequence and is providing opportunities mines renal prognosis. J Am Soc Nephrol. 2016;27:2393-406.
for the development of novel therapeutics. Nonetheless, 15. Liu BC, Tang TT, Lv LL, Lan HY. Renal tubule injury: a
no therapeutic interventions have been shown to prevent driving force toward chronic kidney disease. Kidney Int. 2018;
93:568-79.
CKD progression after AKI, suggesting a gap between 16. Stoumpos S, Mark PB, McQuarrie EP, Traynor JP, Geddes
murine and human AKI. Given that the incidence of AKI CC. Continued monitoring of acute kidney injury survivors
is increasing and its burden is tremendous, further might not be necessary in those regaining an estimated
214 Y. Sato et al.

glomerular filtration rate >60 mL/min at 1 year. Nephrol Dial 36. Galvan DL, Green NH, Danesh FR. The hallmarks of mitochon-
Transplant. 2017;32:81-8. drial dysfunction in chronic kidney disease. Kidney Int. 2017;92:
17. Humphreys BD, Cantaluppi V, Portilla D, Singbartl K, Yang L, 1051-7.
Rosner MH, et al. Targeting endogenous repair pathways after 37. Bhargava P, Schnellmann RG. Mitochondrial energetics in the
AKI. J Am Soc Nephrol. 2016;27:990-8. kidney. Nat Rev Nephrol. 2017;13:629-46.
18. Little MH, Kairath P. Does renal repair recapitulate kidney devel- 38. Szeto HH. Pharmacologic approaches to improve mitochondrial
opment? J Am Soc Nephrol. 2017;28:34-46. function in AKI and CKD. J Am Soc Nephrol. 2017;28:2856-65.
19. Humphreys BD, Valerius MT, Kobayashi A, Mugford JW, 39. Patil NK, Parajuli N, MacMillan-Crow LA, Mayeux PR. Inactiva-
Soeung S, Duffield JS, et al. Intrinsic epithelial cells repair the tion of renal mitochondrial respiratory complexes and manganese
kidney after injury. Cell Stem Cell. 2008;2:284-91. superoxide dismutase during sepsis: mitochondria-targeted anti-
20. de Caestecker M, Humphreys BD, Liu KD, Fissell WH, Cerda J, oxidant mitigates injury. Am J Physiol Renal Physiol. 2014;306:
Nolin TD, et al. Bridging translation by improving preclinical F734-43.
study design in AKI. J Am Soc Nephrol. 2015;26:2905-16. 40. Dare AJ, Bolton EA, Pettigrew GJ, Bradley JA, Saeb-Parsy K,
21. Kusaba T, Lalli M, Kramann R, Kobayashi A, Humphreys BD. Murphy MP. Protection against renal ischemia-reperfusion injury
Differentiated kidney epithelial cells repair injured proximal in vivo by the mitochondria targeted antioxidant MitoQ. Redox
tubule. Proc Natl Acad Sci U S A. 2014;111:1527-32. Biol. 2015;5:163-8.
22. Schiessl IM, Grill A, Fremter K, Steppan D, Hellmuth MK, Cas- 41. Plotnikov EY, Chupyrkina AA, Jankauskas SS, Pevzner IB, Sila-
trop H. Renal interstitial platelet-derived growth factor receptor-b chev DN, Skulachev VP, et al. Mechanisms of nephroprotective
cells support proximal tubular regeneration. J Am Soc Nephrol. effect of mitochondria-targeted antioxidants under rhabdomyoly-
2018;29:1383-96. sis and ischemia/reperfusion. Biochim Biophys Acta. 2011;1812:
23. Nakamura J, Sato Y, Kitai Y, Wajima S, Yamamoto S, Oguchi A, 77-86.
et al. Myofibroblasts acquire retinoic acid-producing ability dur- 42. Plotnikov EY, Pevzner IB, Zorova LD, Chernikov VP, Prusov
ing fibroblast-to-myofibroblast transition following kidney injury. AN, Kireev II, et al. Mitochondrial damage and mitochondria-tar-
Kidney Int. 2019;95:526-39. geted antioxidant protection in LPS-induced acute kidney injury.
24. Lazzeri E, Angelotti ML, Peired A, Conte C, Marschner JA, Antioxidants (Basel). 2019;8:176.
Maggi L, et al. Endocycle-related tubular cell hypertrophy and 43. Szeto HH, Liu S, Soong Y, Seshan SV, Cohen-Gould L, Manichev
progenitor proliferation recover renal function after acute kidney V, et al. Mitochondria protection after acute ischemia prevents pro-
injury. Nat Commun. 2018;9:1344. longed upregulation of IL-1. J Am Soc Nephrol. 2017;28:1437-49.
25. Humphreys BD, Czerniak S, DiRocco DP, Hasnain W, Cheema R, 44. Lynch MR, Tran MT, Parikh SM. PGC1a in the kidney. Am J
Bonventre JV. Repair of injured proximal tubule does not involve Physiol Renal Physiol. 2018;314:F1-8.
specialized progenitors. Proc Natl Acad Sci U S A. 2011; 45. Tran MT, Zsengeller ZK, Berg AH, Khankin EV, Bhasin MK,
108:9226-31. Kim W, et al. PGC1a drives NAD biosynthesis linking oxidative
26. Ferenbach DA, Bonventre JV. Mechanisms of maladaptive repair metabolism to renal protection. Nature. 2016;531:528-32.
after AKI leading to accelerated kidney ageing and CKD. Nat Rev 46. Tran M, Tam D, Bardia A, Bhasin M, Rowe GC, Kher A, et al.
Nephrol. 2015;11:264-76. PGC-1a promotes recovery after acute kidney injury during sys-
27. Bonventre JV, Yang L. Cellular pathophysiology of ischemic temic inflammation in mice. J Clin Invest. 2011;121:4003-14.
acute kidney injury. J Clin Invest. 2011;121:4210-21. 47. Mack M, Yanagita M. Origin of myofibroblasts and cellular
28. Yang L, Besschetnova TY, Brooks CR, Shah JV, Bonventre JV. events triggering fibrosis. Kidney Int. 2015;87:297-307.
Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after 48. Asada N, Takase M, Nakamura J, Oguchi A, Asada M, Suzuki N,
injury. Nat Med. 2010;16:535-43. et al. Dysfunction of fibroblasts of extrarenal origin underlies
29. Cianciolo Cosentino C, Skrypnyk NI, Brilli LL, Chiba T, Novit- renal fibrosis and renal anemia in mice. J Clin Invest. 2011;
skaya T, Woods C, et al. Histone deacetylase inhibitor enhances 121:3981-90.
recovery after AKI. J Am Soc Nephrol. 2013;24:943-53. 49. Duffield JS. Cellular and molecular mechanisms in kidney fibro-
30. Novitskaya T, McDermott L, Zhang KX, Chiba T, Paueksakon sis. J Clin Invest. 2014;124:2299-306.
P, Hukriede NA, et al. A PTBA small molecule enhances 50. Sato Y, Yanagita M. Functional heterogeneity of resident fibro-
recovery and reduces postinjury fibrosis after aristolochic blasts in the kidney. Proc Jpn Acad Ser B. 2019;95:468-78.
acid-induced kidney injury. Am J Physiol Renal Physiol. 51. Nangaku M. Chronic hypoxia and tubulointerstitial injury: a final
2014;306:F496-504. common pathway to end-stage renal failure. J Am Soc Nephrol.
31. DiRocco DP, Bisi J, Roberts P, Strum J, Wong KK, Sharpless N, 2006;17:17-25.
et al. CDK4/6 inhibition induces epithelial cell cycle arrest and 52. Sato Y, Yanagita M. Resident fibroblasts in the kidney: a major
ameliorates acute kidney injury. Am J Physiol Renal Physiol. driver of fibrosis and inflammation. Inflamm Regen. 2017;37:17.
2014;306:F379-88. 53. Sato Y, Yanagita M. Renal anemia: from incurable to curable. Am
32. Pabla N, Gibson AA, Buege M, Ong SS, Li L, Hu S, et al. Mitiga- J Physiol Renal Physiol. 2013;305:F1239-48.
tion of acute kidney injury by cell-cycle inhibitors that suppress 54. Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S,
both CDK4/6 and OCT2 functions. Proc Natl Acad Sci U S A. Bondzie PA, et al. Perivascular Gli1+ progenitors are key contrib-
2015;112:5231-6. utors to injury-induced organ fibrosis. Cell Stem Cell. 2015;
33. Djudjaj S, Martin IV, Buhl EM, Nothofer NJ, Leng L, Piecychna, 16:51-66.
et al. Macrophage migration inhibitory factor limits renal inflam- 55. Brookhart MA, Schneeweiss S, Avorn J, Bradbury BD, Rothman
mation and fibrosis by counteracting tubular cell cycle arrest. J KJ, Fischer M, et al. The effect of altitude on dosing and response
Am Soc Nephrol. 2017;28:3590-604. to erythropoietin in ESRD. J Am Soc Nephrol. 2008;19:1389-95.
34. Ding H, Zhou D, Hao S, Zhou L, He W, Nie J, et al. Sonic hedge- 56. Bernhardt WM, Wiesener MS, Scigalla P, Chou J, Schmieder
hog signaling mediates epithelial-mesenchymal communication RE, G€ unzler V, et al. Inhibition of prolyl hydroxylases
and promotes renal fibrosis. J Am Soc Nephrol. 2012;23:801-13. increases erythropoietin production in ESRD. J Am Soc Neph-
35. Zhou D, Li Y, Lin L, Zhou L, Igarashi P, Liu Y. Tubule-specific rol. 2010;21:2151-6.
ablation of endogenous b-catenin aggravates acute kidney injury 57. Ohashi R, Shimizu A, Masuda Y, Kitamura H, Ishizaki M, Sugi-
in mice. Kidney Int. 2012;82:537-47. saki Y, et al. Peritubular capillary regression during the
AKI progression to CKD 215

progression of experimental obstructive nephropathy. J Am Soc 76. Sato Y, Mii A, Hamazaki Y, Fujita H, Nakata H, Masuda K, et al.
Nephrol. 2002;13:1795-805. Heterogeneous fibroblasts underlie age-dependent tertiary lym-
58. Campanholle G, Ligresti G, Gharib SA, Duffield JS. Cellular phoid tissues in the kidney. JCI Insight. 2016;1:e87680.
mechanisms of tissue fibrosis. 3. Novel mechanisms of kidney 77. Pipi E, Nayar S, Gardner DH, Colafrancesco S, Smith C, Barone
fibrosis. Am J Physiol Cell Physiol. 2013;304:C591-603. F. Tertiary lymphoid structures: autoimmunity goes local. Front
59. Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Ros- Immunol. 2018;9:1952.
ner MH, et al. Inflammation in AKI: current understanding, key 78. Lehmann-Horn K, Wang SZ, Sagan SA, Zamvil SS, von B€ udingen
questions, and knowledge gaps. J Am Soc Nephrol. 2016;27:371-9. HC. B cell repertoire expansion occurs in meningeal ectopic lym-
60. D’Alessio FR, Kurzhagen JT, Rabb H. Reparative T lymphocytes phoid tissue. JCI Insight. 2016;1:e87234.
in organ injury. J Clin Invest. 2019;129:2608-18. 79. Finkin S, Yuan D, Stein I, Taniguchi K, Weber A, Unger K, et al.
61. Dong X, Swaminathan S, Bachman LA, Croatt AJ, Nath KA, Ectopic lymphoid structures function as microniches for tumor
Griffin MD. Antigen presentation by dendritic cells in renal lymph progenitor cells in hepatocellular carcinoma. Nat Immunol. 2015;
nodes is linked to systemic and local injury to the kidney. Kidney 16:1235-44.
Int. 2005;68:1096-108. 80. Shinoda K, Hirahara K, Iinuma T, Ichikawa T, Suzuki AS, Sugaya
62. Maarouf OH, Uehara M, Kasinath V, Solhjou Z, Banouni N, Bah- K, et al. Thy1+IL-7+ lymphatic endothelial cells in iBALT pro-
mani B, et al. Repetitive ischemic injuries to the kidneys result in vide a survival niche for memory T-helper cells in allergic airway
lymph node fibrosis and impaired healing. JCI Insight. 2018;3. inflammation. Proc Natl Acad Sci U S A. 2016;113:E2842-51.
63. Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi BS, et al. Dis- 81. Chang A, Henderson SG, Brandt D, Liu N, Guttikonda R, Hsieh
tinct macrophage phenotypes contribute to kidney injury and C, et al. In situ B cell-mediated immune responses and tubuloin-
repair. J Am Soc Nephrol. 2011;22:317-26. terstitial inflammation in human lupus nephritis. J Immunol. 2011;
64. Sharma R, Kinsey GR. Regulatory T cells in acute and chronic 186:1849-60.
kidney diseases. Am J Physiol Renal Physiol. 2018;314:F679-98. 82. Thaunat O, Field AC, Dai J, Louedec L, Patey N, Bloch MF, et al.
65. Martina MN, Noel S, Saxena A, Bandapalle S, Majithia R, Jie C, Lymphoid neogenesis in chronic rejection: evidence for a local
et al. Double-negative ab T cells are early responders to AKI and humoral alloimmune response. Proc Natl Acad Sci U S A. 2005;
are found in human kidney. J Am Soc Nephrol. 2016;27:1113-23. 102:14723-8.
66. O’Sullivan ED, Hughes J, Ferenbach DA. Renal aging: causes and 83. Luther SA, Lopez T, Bai W, Hanahan D, Cyster JG. BLC expres-
consequences. J Am Soc Nephrol. 2017;28:407-20. sion in pancreatic islets causes B cell recruitment and lymphotoxin-
67. Schmitt R, Melk A. Molecular mechanisms of renal aging. Kidney dependent lymphoid neogenesis. Immunity. 2000;12:471-81.
Int. 2017;92:569-79. 84. Luther SA, Bidgol A, Hargreaves DC, Schmidt A, Xu Y, Pan-
68. Naik AS, Afshinnia F, Cibrik D, Hodgin JB, Wu F, Zhang M, iyadi J, et al. Differing activities of homeostatic chemokines
et al. Quantitative podocyte parameters predict human native kid- CCL19, CCL21, and CXCL12 in lymphocyte and dendritic cell
ney and allograft half-lives. JCI Insight. 2016;1(7):e86943. recruitment and lymphoid neogenesis. J Immunol. 2002;169:
69. Denic A, Lieske JC, Chakkera HA, Poggio ED, Alexander MP, 424-33.
Singh P, et al. The substantial loss of nephrons in healthy human 85. Heesters BA, Myers RC, Carroll MC. Follicular dendritic cells:
kidneys with aging. J Am Soc Nephrol. 2017;28:313-20. dynamic antigen libraries. Nat Rev Immunol. 2014;14:495-504.
70. Polichnowski AJ, Lan R, Geng H, Griffin KA, Venkatachalam MA, 86. Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, et al.
Bidani AK. Severe renal mass reduction impairs recovery and pro- Molecular characterization of the transition from acute to chronic
motes fibrosis after AKI. J Am Soc Nephrol. 2014;25:1496-507. kidney injury following ischemia/reperfusion. JCI Insight. 2017;2:
71. Schmitt R, Marlier A, Cantley LG. Zag expression during aging e94716.
suppresses proliferation after kidney injury. J Am Soc Nephrol. 87. Cippa PE, Liu J, Sun B, Kumar S, Naesens M, McMahon AP. A
2008;19:2375-83. late B lymphocyte action in dysfunctional tissue repair follow-
72. Sato Y, Yanagita M. Immunology of the ageing kidney. Nat Rev ing kidney injury and transplantation. Nat Commun. 2019;10:
Nephrol. 2019;15:625-40. 1157.
73. Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular 88. Nayar S, Campos J, Smith CG, Iannizzotto V, Gardner DH, Mour-
senescence. Trends Cell Biol. 2018;28:436-53. cin F, et al. Immunofibroblasts are pivotal drivers of tertiary lym-
74. Hashimoto M, Asai A, Kawagishi H, Mikawa R, Iwashita Y, phoid structure formation and local pathology. Proc Natl Acad Sci
Kanayama K, et al. Elimination of p19ARF-expressing cells enhan- U S A. 2019;116:13490-7.
ces pulmonary function in mice. JCI Insight. 2016;1:e87732. 89. Okusa MD, Rosner MH, Kellum JA, Ronco C, Acute Dialysis
75. Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Quality Initiative XIII Workgroup. Therapeutic targets of human
et al. Naturally occurring p16(Ink4a)-positive cells shorten AKI: harmonizing human and animal AKI. J Am Soc Nephrol.
healthy lifespan. Nature. 2016;530:184-9. 2016;27:44-8.

You might also like