Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Nephrol Dial Transplant, 2023, 0, 1–11

https://doi.org/10.1093/ndt/gfad215
Advance access publication date: 3 October 2023

Tubular toxicity of proteinuria and the progression of

REVIEW
chronic kidney disease

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


Zhalaliddin Makhammajanov1 , Abduzhappar Gaipov 2 ,3
, Askhat Myngbay4 , Rostislav Bukasov5 , Mohamad Aljofan1
and Mehmet Kanbay 6
1
Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
2
Department of Medicine, School of Medicine, Nazarbayev University, Astana, Kazakhstan
3
Clinical Academic Department of Internal Medicine, CF “University Medical Center”, Astana, Kazakhstan
4
Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
5
Department of Chemistry, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
6
Division of Nephrology, Department of Internal Medicine, Koc University, Istanbul, Turkey
Correspondence to: Abduzhappar Gaipov; E-mail: abduzhappar.gaipov@nu.edu.kz

ABSTRACT
Proteinuria is a well-established biomarker of chronic kidney disease (CKD) and a risk predictor of associated disease outcomes. Pro-
teinuria is also a driver of CKD progression toward end-stage kidney disease. Toxic effects of filtered proteins on proximal tubular
epithelial cells enhance tubular atrophy and interstitial fibrosis. The extent of protein toxicity and the underlying molecular mecha-
nisms responsible for tubular injury during proteinuria remain unclear. Nevertheless, albumin elicits its toxic effects when degraded
and reabsorbed by proximal tubular epithelial cells. Overall, healthy kidneys excrete over 1000 individual proteins, which may be
potentially harmful to proximal tubular epithelial cells when filtered and/or reabsorbed in excess. Proteinuria can cause kidney dam-
age, inflammation and fibrosis by increasing reactive oxygen species, autophagy dysfunction, lysosomal membrane permeabilization,
endoplasmic reticulum stress and complement activation. Here we summarize toxic proteins reported in proteinuria and the current
understanding of molecular mechanisms of toxicity of proteins on proximal tubular epithelial cells leading to CKD progression.

Keywords: albuminuria, chronic kidney disease, proteinuria, renal tubular toxicity, tubulointerstitial fibrosis

INTRODUCTION sion, e.g. via toxic effects of filtered proteins on proximal tubu-
lar epithelial cells (PTECs), leading to tubular cell loss and tubu-
Chronic kidney disease (CKD) is characterized by long-term
lar atrophy followed by tubulointerstitial fibrosis, and eventually
functional decline and structural deterioration of the kidneys [1].
ESKD [7]. The presence of excess proteins in the urine follows at
The estimated prevalence of CKD varies between 11% and 13%
least one of the mechanisms: increased levels of low-molecular-
worldwide [2]. The level of glomerular filtration rate (GFR) and
weight proteins in the systemic circulation (prerenal proteinuria);
the extent of proteinuria/albuminuria define the severity of CKD
impaired permselectivity of the glomerulus (glomerular protein-
[3]. Proteinuria refers to elevated urinary protein levels, while
uria); abnormal reabsorption of low-molecular-weight proteins by
albuminuria specifically indicates abnormal albumin loss in the
tubular cells and tubular cell–expressed protein loss (tubular pro-
urine. GFR and albuminuria levels are the excretory and barrier
teinuria); or protein shedding from cells of the lower urinary tract
dysfunction indicators, respectively [1]. In the general population,
(post-renal proteinuria) [8] (Supplementary data, Table S1).
CKD is commonly observed among people over 65 years of age
Prerenal proteinuria is primarily caused by circulating plasma
as episodes and causes of kidney injury accumulate throughout
proteins released due to progressing diseases located outside
life; hence CKD in the elderly is usually multifactorial [2, 3].
the kidneys (Supplementary data, Table S2). The most common
Unfortunately, mortality due to CKD has been increasing over
causes of prerenal proteinuria are plasma cell neoplasms, rhab-
the past 25 years, and it is expected to grow yearly by about 1%
domyolysis and hemolysis [9–11]. Plasma cell neoplasms are re-
[4]. The primary factors associated with increased mortality have
sponsible for the overproduction of monoclonal immunoglobulin-
been identified as lower estimated GFR and higher proteinuria [5].
free light chains that render cytotoxicity on PTECs via different
These factors were further supported by a recent meta-analysis
mechanisms, leading to progressive CKD [9]. During rhabdomyol-
of clinical trials, which revealed that with treatment-induced
ysis, an excessive amount of myoglobin is released from muscle
reduction of proteinuria, every 30% reduction was associated
breakdown, swiftly filtered by the glomerulus and accumulated
with a 23.7% decreased risk of CKD progression to end-stage
in tubules, causing proximal tubular toxicity and acute kidney in-
kidney disease (ESKD) [6]. Notably, this reduction in proteinuria
jury (AKI) [10]. Likewise, massive hemolysis results in the accu-
was independent of the drug classes in the treatments.
mulation of hemoglobin in the systemic circulation and then in
Evidence shows that proteinuria is a potential biomarker and
the glomerular ultrafiltrate. Excess free hemoglobin and released
a risk predictor for ESKD and CKD-associated complications [7].
heme in PTECs exert significant direct cytotoxicity to cells, pro-
Proteinuria is recognized as a significant driver of CKD progres-
moting AKI and/or CKD [11].

Received: June 16, 2023; Editorial decision: September 4, 2023


© The Author(s) 2023. Published by Oxford University Press on behalf of the ERA. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com
2 | Nephrol Dial Transplant, 2023, Vol. 0, No. 0

Renal proteinuria is the primary proteinuria responsible for PROXIMAL TUBULAR RECEPTORS AND
CKD development [7] (Table 1 and Supplementary data, Table S3). PROTEINURIA
Glomerular proteinuria arises from a dysfunctional glomerular
Proximal tubules are crucial in maintaining low-protein urine
filtration barrier characterized by impaired charge and size selec-
by an enzymatic breakdown and endocytosis of filtered proteins.
tivity. On the other hand, tubular proteinuria is caused by dys-
PTECs have extensive apical endocytic apparatus, where reab-
functional tubules, mainly due to impaired endocytic receptors
sorbed proteins are endocytosed, concentrated in vesicles and un-
such as the megalin–cubilin complex [12]. Unlike renal protein-
dergo hydrolysis in lysosomes. Megalin, cubilin, amnionless and
uria, post-renal proteinuria is a secondary form of proteinuria. It is

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


the neonatal Fc receptor for IgG are the primary receptors that
usually transient, resulting from infectious, inflammatory and/or
maintain the uptake of proteins from the ultrafiltrate [45]. Me-
cancerous conditions of the distal genitourinary tract.
galin is a member of the low-density lipoprotein receptor family
Molecular mechanisms for the progression of CKD into ESKD
and is highly expressed at the apical surface of PTECs [45]. Cubilin
are primarily associated with tubulointerstitial damage due to the
has no transmembrane domain and therefore needs megalin to be
cytotoxicity of filtered urinary proteins to PTECs, regardless of the
associated with the membrane and for cellular endocytosis with
type of glomerular disease, causing proteinuria. In this review, we
bound proteins. Cubilin also needs amnionless, a transmembrane
discuss the effects of proximal tubular toxicity due to filtered pro-
protein, for trafficking to the plasma membrane and correct local-
teins on the progression of CKD in renal proteinuria.
ization [45].
Notably, acute and chronic diseases, particularly those af-
fecting proximal tubular endocytic receptors, can lead to low-
PROTEIN COMPOSITION IN PROTEINURIA molecular-weight proteinuria, which, if left untreated, may
The glomerular ultrafiltrate primarily contains low-molecular- progress to ESKD. Like patients with glomerular proteinuria,
weight proteins with molecular weights <60 kDa because of many patients with low-molecular-weight proteinuria also show
glomerular permselectivity but are efficiently reabsorbed in the progressive kidney function decline. Nonetheless, it remains
proximal tubule [13]. Among serum proteins, albumin is the only unclear whether low-molecular-weight proteinuria is a con-
abundant larger protein that appears in the urine of healthy indi- tributor to tubular damage or solely a biomarker of such
viduals at a concentration <30 mg/24 h out of total protein con- damage [46].
tent of <150 mg/24 h. The intact filtration barrier retains most Despite the knowledge advancement in tubular endocytic
serum albumin because of its molecular weight of 69.367 kDa receptors, little is known about their regulation and roles in
and a net charge of −15. Despite their high molecular weight, proteinuria. Previous in vitro studies have identified megalin as
some albumin molecules can appear in the ultrafiltrate owing a sensor of filtered albumin, determining whether PTECs will be
to their ellipsoid shape and flexibility [12]. Nevertheless, the re- protected or injured by it [47]. At low doses, albumin seems to
nal tubules efficiently reabsorb most of the albumin from the promote PTECs survival by inducing the megalinprotein kinase B
ultrafiltrate. Thus, albumin in the final urine is negligible and interaction and the phosphorylation of BCL2 associated agonist
accounts for approximately 20% of the total protein. However, of cell death (BAD). Conversely, high doses of albumin reduce
albumin levels may increase up to 300-fold in glomerular pro- megalin expression and lead to cell apoptosis. Also, in vitro stud-
teinuria due to filtration barrier damage and the limited capac- ies showed that long-term exposure to albumin decreased the
ity of the proximal tubule to degrade and recover such albumin binding and reabsorption capacity of PTECs, suggesting that per-
from the filtrate [14]. Any disruption of the glomerular filtration sistent exposure to albumin can be toxic to endocytic receptors
barrier that leads to an abnormal loss of albumin causes selec- [48]. A recent study demonstrated that the expression of megalin,
tive glomerular proteinuria (i.e. albuminuria). Additionally, the cubilin and the neonatal Fc receptor for IgG was downregulated
presence of other high-molecular-weight proteins results in non- in PTECs in an experimental model of protein-overload protein-
selective glomerular proteinuria (Supplementary data, Table S1) uria, while the expression of megalin was also markedly reduced
[15]. In clinical practice, urinary albumin concentration between in kidneys of proteinuric patients [49]. Still, these receptors
30 and 300 mg/24 h is called microalbuminuria, and >300 mg/24 h were found in high levels in the urine, indicating a potential
is called proteinuria/albuminuria. shedding from cell surfaces in proteinuria. However, increased
Although albumin is the most abundant serum protein in shedding of megalin in urine samples of patients with IgA and
the urine, some studies reported thousands of different proteins membranous nephropathy was associated with its maintained
as a component of normal urine, which can be detected us- expression [50].
ing different techniques [16]. Marimuthu’s group identified 1823 Nevertheless, it is worth mentioning that albuminuria caused
proteins [17], while Santucci’s group reported 3429 proteins by by receptor-related genetic kidney diseases, particularly genetic
analyzing urine samples of healthy individuals [18]. Generally, mutations of cubilin, has been found to be benign and not to lead
the main protein component of urine consists of 50–100 mg to kidney function decline in patients with chronic albuminuria
of uromodulin secreted from cells of the thick ascending limb [51]. This study also showed that albuminuria was developed due
of Henle, which makes up about 50% of the total protein and to decreased albumin reabsorption by mutated cubilin, thereby
is hence the most abundant in urine [16]. Normal urine also causing less cellular cytotoxicity.
contains low amounts of low-molecular-weight proteins and se- Overall, proteinuria/albuminuria appears to cause damage to
creted proteins from lower urinary tracts, such as immunoglob- endocytic receptors, with or without changes in their expression.
ulin A (IgA) (Supplementary data, Table S4). The most fre- However, whether proteinuria/albuminuria directly affects tubu-
quently reported proteins in proteinuria are listed in Table 1 and lar receptors has not been clarified and thus is a subject for fur-
Supplementary data, Tables S2 and S3, with indications of their ther research.
effects on PTECs.
Table 1: Most frequently reported urinary proteins and protein-bound molecules in renal proteinuria and their effects on proximal tubular cells.

Protein
molecular
Origin of proteinuria Tubular reabsorption Urinary proteins as biomarkers for weight
Protein (causes) (mechanism/receptor) Effects on PTECs (mechanism/cell phenotype) pathologies (kDa)

Albumin Renal: glomerular Reabsorption by – Proinflammatory via ROS accumulation – Glomerular and/or tubulointerstitial 69.367
damage and/or megalin–cubilin – Endothelin-1-mediated tubulointerstitial fibrosis and damage and CKD progression
inadequate complex, fluid-phase AKI–CKD progression (progressor of AKI to CKD) [20] – Detection of early-stage CKD [25]
reabsorption due to endocytosis and FcRn – Profibrotic via NOX activation [21] – Risk for AKI/progressive
tubular damage [19] [13] – Lysosome-mediated damage [22] CKD/CVD/ESKD/mortality in diabetic
– Endoplasmic reticulum stress-mediated damage [23] and nondiabetic patients [26]
– Oxidative damage via cadmium-albumin
complex/NOX activation/dysfunctional
mitochondria/lysosomal membrane
permeabilization [24]

Albumin-bound Renal: glomerular and/or Reabsorption is through – Proinflammatory via ROS accumulation [28] NA
fatty acids (e.g. tubular damage albumin endocytosis – Pro-apoptotic [27]
palmitate and and/or fatty acid – Profibrotic [29]
oleate) transporter-2–
mediated endocytosis
[27, 28]
Transferrin Renal: glomerular and/or Reabsorption is mediated – Endothelin-1-mediated tubulointerstitial fibrosis and – Early endothelial dysfunction in 78
tubular damage via cubilin and AKI–CKD progression (progressor of AKI to CKD) [20] diabetic patients at a very early
transferrin receptor 1 – Proinflammatory renal damage [31]
receptors [13] – Cytotoxicity via C3 deposition [30] – Prediction of onset of DKD [32]

IgG Renal: glomerular FcRn-mediated – Endothelin-1-mediated tubulointerstitial fibrosis and – Glomerular damage in diabetic and 150
damage (FSGS) endocytosis [33] AKI–CKD progression (progressor of AKI to CKD) [20] nondiabetic patients with
proteinuric glomerulopathies [34]
– Prediction of onset of DKD [32]

Complement C3 Renal: filtered and/or NA – Proinflammatory [35] – IgA nephropathy [38] 180
locally expressed in – Reperfusion damage [36]
injured tubular cells – Profibrotic and cytolytic via generation of C3a and
C5a anaphylatoxins and membrane attack complex
[37]

Ceruloplasmin Renal: glomerular NA – Cytotoxic via increased release of copper from – IgA nephropathy [38] 132
damage ceruloplasmin under acidic conditions in tubular – Prediction of onset of DKD [32]
lumen [39]

N-acetyl-β-d- Renal: glomerular Not reabsorbed – Hydrolysis of glycoproteins, glycolipids, or – Tubular cell dysfunction and a 130-140
glucosaminidase damage or locally mucopolysaccharides in cells [40] predictor of outcome in primary
secreted due to tubular glomerulonephritis [40]
damage – Risk of kidney disease and
progression in diabetic patients [41]

Fibrinogen Renal: glomerular NA – Tubulointerstitial fibrosis by enhancing renal – Progression of CKD to ESKD [44] 340
damage and/or locally fibroblast proliferation [43]
secreted by injured
tubular cells [42]

CVD, cardiovascular disease; DKD, diabetic kidney disease; FcRn, the neonatal Fc receptor; FSGS, focal segmental glomerulosclerosis; NA, not applicable; NADPH, nicotinamide adenine dinucleotide phosphate; NOX, nicoti-
namide adenine dinucleotide phosphate oxidase.
Z. Makhammajanov et al. | 3

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


4 | Nephrol Dial Transplant, 2023, Vol. 0, No. 0

Proximal tubular epithelial cell

Cubilin TGF-β1 TβRII FATP2


FFAs
Megalin Proteins (albumin)

ECM synthesis

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


Enzymes ↑

TIMP-1 and TIMP-2 Rac1


Inflammation Enzymes
• NF-κB Amino acids
NOX • eNOS
Mitochondrion • Cytokines Stress/dysfunction
• VEGF ↑ Increase
↑ROS • MAPK
↑ATP ↑ROS Cyt c Induce
↑Cyt c • JNK/SAPK
Inhibit
Movement

Lysosome Caspase-9-mediated
PTEC death
ERK1/2
↑Actin and fibronectin

Tubular interstitial fibrosis ECM synthesis

Figure 1: Intracellular ROS activation and its effect on PTECs in proteinuria/albuminuria. ROS production is one of the major players in the
pathogenesis of tubulointerstitial injury. Reuptake of massive urinary filtered proteins, particularly albumin, leads to Rac1-mediated NOX activation
and ROS synthesis. Reabsorbed albumin also increases ROS accumulation in the cytoplasm by causing mitochondrial dysfunction. Reabsorbed
albumin is transported to lysosomes, and albumin-bound FFAs are transported further to tubular mitochondria after either albumin degradation into
amino acids in lysosomes or receptor-mediated endocytosis. FFA molecules are also reabsorbed from the ultrafiltrate by FATP2 and transported to
mitochondria. In the mitochondria, FFAs accelerate mitochondrial ROS synthesis leading to the loss of mitochondrial membrane potential and the
release of ROS and Cyt c into the cytoplasm. Cyt c activates caspase-9-pathway-mediated PTEC apoptosis. Accumulated ROS induces the activation of
NF-ĸB, eNOS, proinflammatory cytokines, VEGF, MAPK and JNK/SAPK. Most of these are potent proinflammatory factors as well as induce ROS
generation enabling a vicious cycle of tubulointerstitial injury. Furthermore, ROS is involved in TGF-β-mediated ECM synthesis and expansion that
results in interstitial fibrosis, which can also occur independently of TGF-β1 induction. Notably, the accumulating ECM components need to be
degraded and normalized. However, this regulatory mechanism is broken by albumin-induced TIMP-1 and TIMP-2 which block a family of zinc
metal–dependent enzymes that degrade one or more ECM proteins. In addition, FFAs can promote ECM development by enhancing the expression of
alpha–smooth muscle cell actin and inducing the secretion of fibronectin which is present in tubulointerstitial fibrosis. ATP, adenosine triphosphate;
Cyt c, cytochrome complex; eNOS, endothelial nitric oxide synthase; FATP2, fatty acid transport protein-2; JNK/SAPK, Jun N-terminal
kinases/stress-activated protein kinases; MAPK, p38 mitogen-activated protein kinase; NOX, nicotinamide adenine dinucleotide phosphate oxidase;
Rac1, Rho-related C3 botulinum toxin substrate 1; TIMP-1, tissue inhibitor of metalloproteinase 1; TIMP-2, tissue inhibitor of metalloproteinase 2;
VEGF, vascular endothelial growth factor.

TUBULAR TOXICITY OF FILTERED PROTEINS minimal change disease, disease progression is mild, and patients
Proteinuria increases the production of ROS do not develop ESKD with proper treatment and care. This may be
inside proximal tubular cells explained by the substantially low FFAs content of urinary albu-
min in minimal change disease compared with other nephrotic
Previous studies showed that reuptake of massive albumin trig-
syndromes [55].
gers high levels of reactive oxygen species (ROS) in PTECs (Fig. 1)
Serum albumin is the main carrier protein for FFAs (also
[52]. Albumin activates Rac1, a Rho-family small GTPase, by pro-
named non-esterified fatty acids), which, after passing the in-
tein kinase C in PTECs, leading to NOX, nicotinamide adenine din-
jured glomerular filtration barrier, are endocytosed into PTECs,
ucleotide phosphate oxidase activation and ROS production [52].
and transported to cell mitochondria after albumin degradation
Mechanistically, urinary albumin induces ROS generation by trig-
into amino acids by lysosomes [28]. During proteinuria, cellular
gering mitochondrial dysfunction [53]. Importantly, Ruggiero et al.
uptake of FFAs is also mediated by the apical plasma membrane
argue that albumin-bound free fatty acids (FFAs) cause mitochon-
receptor, FATP2, and FFAs are transported into the mitochondria
drial ROS-mediated PTEC demise, but not albumin itself [53]. In-
to trigger lipoapoptosis [27]. Inside mitochondria, FFAs are utilized
deed, it should be noted that multiple in vitro and in vivo exper-
for ATP generation by the electron transport system, tricarboxylic
iments have claimed that delipidated albumin is not cytotoxic,
acid cycle and β-oxidation [56]. Thus, protein overload results in
but rather albumin-bound FFAs (e.g. palmitate and oleate) cause
the accumulation of abundant FFAs in tubular cell mitochondria,
and exacerbate tubulointerstitial injury during protein overload
and they accelerate mitochondrial ROS synthesis leading to the
of PTECs [27, 53, 54]. In some cases of albuminuria, particularly in
loss of mitochondrial membrane potential [28]. Subsequently, this
Z. Makhammajanov et al. | 5

leads to the release of mitochondrial cytochrome-c along with or selectively to target specific cellular components for degrada-
ROS into cytosol and activation of caspase-9-pathway-mediated tion [65]. Examples of selective autophagy processes include mi-
PTEC apoptosis [57], leading to the loss of functional PTECs in the tophagy, which targets damaged or dysfunctional mitochondria,
kidney (Fig. 1). and lysophagy, which removes damaged or dysfunctional lyso-
Accumulated intracellular ROS induce a variety of signaling somes. Autophagy activation has been suggested to play protec-
cascades such as nuclear factor-kappaB (NF-κB), Jun N-terminal tive roles in PTECs early during AKI by increasing the resilience of
kinases/stress-activated protein kinases, p38 mitogen-activated tubular cells to ischemic necroinflammation [66]. Nevertheless,
protein kinase and uncoupling of endothelial nitric oxide syn- numerous studies showed that persistent or impaired autophagy

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


thase, along with increased expression of inflammatory cytokines activation could significantly contribute to AKI–CKD transition
and vascular endothelial growth factor [58–60]. Interestingly, most and CKD progression in general (Fig. 2) [67].
of these signaling pathways will also increase the generation of Proteinuria activates autophagy in PTECs to protect the cells
ROS. For example, a dimerized/coupled endothelial nitric oxide from injury; however, as proteinuria persists, protein overload
synthase produces nitric oxide; however, an uncoupled form of impairs autophagy and renders their demise. Treatment of the
the enzyme will lead to the generation of superoxide anion O2 •– , human proximal tubular HK-2 cell line with urinary proteins or
the main form of ROS [61]. human serum albumin resulted in a dose- and time-dependent
Altogether, these findings indicate that chronic protein over- increase in the synthesis of microtubule-associated protein 1
load in combination with lipid moiety substantially increases cel- light chain 3-II (an autophagosome marker) [68]. This increase
lular synthesis of ROS, thereby triggering and exacerbating tubu- peaked at 8 hours, and effective autophagic flux was completed
lointerstitial necroinflammation that eventually results in ESKD. at 16 hours post-treatment, in comparison to the control cells.
In this study, oxidative stress was reported to mediate urinary
Proteinuria-induced ROS mediate protein-induced autophagy activation. This study also demon-
tubulointerstitial fibrosis development strated autophagy activation induced by proteinuria in rat
protein-overload models, which could be modulated by chloro-
Wolf and colleagues suggest that albumin upregulates the type II
quine and rapamycin. Similarly, other studies also confirmed the
transforming growth factor-beta receptor (TGF-β) in PTECs; this
protective roles of protein-induced autophagy in PTECs [69].
can amplify the autocrine effect of proteinuria-induced TGF-β1
Persistent protein exposure to PTECs is tubulotoxic by sup-
[62]. Bondi et al. claim that nicotinamide adenine dinucleotide
pressing autophagy, allowing the accumulation of toxic intra-
phosphate oxidase–derived ROS is involved in TGF-β1-induced ac-
cellular molecules, such as long-lived proteins, which may be
tivation and transformation of fibroblasts into myofibroblasts, a
cytotoxic on PTECs. In vitro studies demonstrated that treat-
primary responsible source for extracellular matrix (ECM) syn-
ment of HK-2 and mouse primary proximal tubule cells with
thesis and expansion that results in interstitial fibrosis (Fig. 1)
albumin led to a dose-dependent reduction in the formation of
[21]. Overproduced ROS induces α-SMA-positive myofibroblast ac-
microtubule-associated protein light chain 3-II and autophagic
tivation through the ERK1/2 signaling pathway [21]. This ROS-
flux [14]. This effect was observed in the presence of bafilomycin,
mediated fibroblast conversion into myofibroblast can occur in-
an H+ -ATPase inhibitor that hinders microtubule-associated
dependently of TGF-β1 induction [63]. Notably, the accumulat-
protein light chain 3-II degradation. Additionally, these cells
ing ECM components are balanced by proteinases degrading ECM
exhibited a significantly increased half-life of long-lived proteins,
molecules. Still, this counterregulatory mechanism gets impaired
indicating impaired autophagy. Albumin-induced reduction of
by albumin-induced tissue inhibitors of metalloproteinases-1 and
autophagic response was mediated by inducing the activity of
tissue inhibitors of metalloproteinases-2 which block a family of
a potent inhibitor of autophagy, the mammalian target of ra-
zinc metal–dependent enzymes that degrade one or more ECM
pamycin, as shown by highly phosphorylated S6K, a downstream
proteins at neutral pH [63]. Besides, lipid moiety of urinary al-
target of mammalian target of rapamycin, after treatment with
bumin has previously been shown to be responsible for ECM de-
amino acids and proteins in vitro [14], suggesting that albumin
velopment in protein-overloaded rats by upregulating the expres-
impairs autophagy via mammalian target of rapamycin pathway
sion of alpha–smooth muscle cell actin, a marker of myofibroblast
after lysosomal degradation into amino acids.
transformation, and inducing the secretion of ECM proteins such
Chronic proteinuria can also disrupt selective autophagy, accu-
as fibronectin, which is present in tubulointerstitial fibrosis [54].
mulating defective organelles, such as mitochondria, which pro-
Recent work in the unilateral ureteral obstruction kidneys model
mote tubular injury [70, 71]. Recent in vitro and in vivo studies
also demonstrated the roles of receptor-mediated uptake of FFAs
found that chronic albumin exposure of NRK-52E (rat-derived
in kidney fibrogenesis [29]. Moreover, a previous human study of
PTEC cell line) cells and experimental mice led to impaired NIP3-
kidney biopsies of patients with heavy proteinuria reported in-
like protein X-mediated mitophagy, increasing the accumula-
creased production of TGF-β1 in PTECs and increased urine excre-
tion of damaged mitochondria and mitochondrial ROS synthesis
tion [64], signifying the role of TGF-β1 in tubulointerstitial fibrosis.
[70]. In addition, these albumin-treated cells exhibited oxidative
Overall, it seems that either directly or indirectly, albuminuria
stress–dependent cell death [72], and mice exhibited tubular le-
via lipid moiety causes increased accumulation of ECM protein
sions and renal dysfunction [70]. Studies of diabetic kidney dis-
and its decreased degradation, cumulatively enhancing the pro-
ease models showed that defective mitophagy builds up damaged
gression of tubulointerstitial fibrosis.
mitochondria and ROS in PTECs, activating oxidative stress path-
ways and, eventually, tubular cell death [71]. Therefore, the quality
Proteinuria impairs tubular cell stress resilience control of mitochondria is essential for the efficient reabsorption
by compromising autophagy of filtered proteins by PTECs, which require high energy.
Autophagy is a cellular recycling pathway that increases cell re- These data indicate that autophagy may be critical in the early
silience to oxidative stress by retrieving biomolecules from redun- stage of proteinuria to protect cells from injury. However, persis-
dant or injured cellular components [65]. Autophagy carries out tent proteinuria probably has markedly cytotoxic effects on PTECs
recycling either non-selectively to maintain cellular homeostasis by inhibiting basal and selective autophagy activities, allowing
6 | Nephrol Dial Transplant, 2023, Vol. 0, No. 0

FATP2 Cubilin
FFAs
Megalin Proteins (albumin)
Proximal tubular epithelial cell

Autophagosome

? mTOR

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


Rac1 NOX
Lysosome
Mitochondrion

↓ ↓ ↑ROS ?

↑ATP ↑ROS Cyt c
Kidney Inflammation
repair Apoptosis
↑pH ↓Degradation Lysosome
ER
↓Fusion ↓Mitophagy Cyt c ↑Ca
↓ ROS
Autophagosome
Calpain CHOP

↓ mTOR
PTEC death
Inflammation


↑ROS

Tubular interstitial fibrosis ECM synthesis

Cathepsins Unfolded/long-lived proteins ↑ Increase Inhibit


Stress/dysfunction Amino acids Induce Movement

LC3-II

Figure 2: Proteinuria-induced dysfunctional autophagy, lysosomal membrane permeabilization and endoplasmic reticulum stress. Proteinuria initially
activates autophagy in PTECs by increasing ROS generation to protect cells from injury. However, prolonged exposure of filtered proteins to PTECs
starts to render toxic tubular effects by suppressing autophagic activities (e.g. mitophagy, basal autophagy and lysophagy), allowing the accumulation
of defective organelles such as mitochondria and toxic intracellular molecules such as long-lived proteins, which promote progressive tubular injury.
Accumulated defective mitochondria increase intracellular ROS and induce PTEC death, leading to tubulointerstitial inflammation, fibrosis and
nephron loss. Reabsorbed albumin can inhibit basal autophagy by activating mTOR after lysosomal degradation into amino acids, thereby allowing the
accumulation of long-lived proteins in the cytoplasm. Furthermore, massive proteinuria leads to an increase in the number and mass of lysosomes,
but it results in decreased lysosomal enzymatic and degradation activities. Massive proteinuria also overwhelms lysosomal pathways and leads to LMP
allowing the release of lysosomal ROS and cathepsins into the cytoplasm. In the cytoplasm, cathepsins cause PTEC death. In addition, LMP may cause
a decline in autophagosome clearance of defective lysosomes. Moreover, proteinuria leads to ER stress via at least two molecular processes, including
increased ROS and intracellular calcium levels in PTECs. Subsequently, ER stress causes calpain and/or CHOP pathways–mediated PTEC death. ATP,
adenosine triphosphate; CHOP, CCAAT/enhancer-binding protein homologous protein; Cyt c, cytochrome complex; ER, endoplasmic reticulum; FATP2,
fatty acid transport protein-2; LC3-II, microtubule-associated protein light chain 3-II; LMP, lysosomal membrane permeabilization; mTOR, mammalian
target of rapamycin; NOX, nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase; Rac1, Rho-related C3 botulinum toxin substrate 1.

accumulation of toxic intracellular molecules such as long-lived radicals from lysosomal hydrogen peroxide, promoting lysoso-
proteins/protein aggregates and damaged organelles such as mi- mal membrane damage [74]. In addition, massive proteinuria
tochondria, thus promoting progressive tubular injury and CKD. likely overwhelms lysosomal pathways and results in a lysoso-
mal rupture in PTECs, promoting tubular injury (Fig. 2) [22]. In
support of this notion, Liu et al. conducted in vitro and in vivo
Lysosomal membrane permeabilization experiments and found that lysosomal mass and amount were
A lysosome is a cellular membrane-enclosed organelle that car- considerably elevated in HK-2 cells and PTECs of patients with
ries out the degradation of cellular components and macro- nephrotic syndrome in response to protein overload. However,
molecules delivered through either endocytosis or autophagy [73]. lysosomal enzymatic and degradation activities were decreased,
Lysosomes are abundant in free iron because of the degradation as seen by impaired cathepsin-B/cathepsin-L activities and acid-
of iron-containing proteins. This elevates ROS inside lysosomes ification of lysosomes [75]. Additionally, in HK-2 cells, there was
via the iron-mediated generation of strongly reactive hydroxyl evidence of cathepsin-B leakage into the cytoplasm. Moreover, in
Z. Makhammajanov et al. | 7

PTECs from patients with various causes of nephrotic syndrome, Cytotoxicity of complement proteins and
both cathepsin-B and lysosome-associated membrane protein-1 complement activation
showed leakage, indicating lysosomal membrane permeabiliza- Excessive proteinuria with complement activation in the kidney
tion. These effects were induced by oxidative stress following ex- tubules is one of the major drivers of tubulointerstitial damage
posure to urinary proteins [75]. Subsequently, depending on the [81]. PTECs are susceptible to complement-mediated cell dam-
damage and release of its contents, including cathepsins, lyso- age due to negative or low expression of complement suppres-
somal membrane permeabilization triggers either apoptosis or sors such as membrane cofactor protein, complement receptor
necrosis of cells [74].

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


1, decay accelerating factor or CD59 on the apical region of the
In addition, lysosomal membrane permeabilization releases cells [82]. Complement molecules are absent in the glomerular
ROS into the cytoplasm, exacerbating oxidative stress and lead- ultrafiltrate, but they appear during proteinuria and can activate
ing to cell death [74]. Therefore, proteinuria-induced damaged the alternative pathway of the complement system [83]. Recent
lysosomes can be an important contributor to the progression evidence suggests that urinary properdin, a positive regulator of
of CKD. Notably, defective lysosomes must be eliminated by the alternative pathway, together with filtered other complement
lysophagy along with recovery of reduced degradation activity molecules, is responsible for the initiation of complement activa-
via lysosome biogenesis to maintain the survival of PTECs [76]. tion through binding to PTECs and functioning as a docking sta-
However, lysosome depletion in the form of lysosomal membrane tion for the assembly of alternative pathway C3 convertase on the
permeabilization may cause defective lysosome clearance and apical surface of proximal tubules [84].
autophagy flux decline [76], thereby accumulating damaged lyso- Additionally, properdin strongly stabilizes C3 convertase and
somes and increasing apoptotic pathways. These suggest that ef- thus enhances alternative pathway amplification, leading to the
ficient lysosomal function is essential in the physiology of protein generation of C3a and C5a anaphylatoxins and membrane at-
reabsorption; however, protein overload of lysosomes disrupts tack complex (Fig. 3) [37]. Consequently, complement activation
this degradation process and causes lysosomal membrane may contribute to CKD progression with different mechanisms
permeabilization, resulting in lysosome-dependent cell death that eventually lead to interstitial fibrosis. Abbate et al. found that
and impaired autophagic flux, thereby promoting nephron loss, abnormal accumulation of C3 in the tubular lumen by protein
interstitial inflammation and CKD. overload rather than locally expressed C3 triggered proteinuria-
induced tubulointerstitial inflammation and cell damage in a
Endoplasmic reticulum stress murine model of protein-overload proteinuria [35]. An animal
The endoplasmic reticulum is essential in protein synthesis and study conducted on C3a receptor and/or C5a receptor–deficient
folding, lipid synthesis, and calcium storage. However, it can mice with transplanted kidneys from Crry–/– C3–/– mice reported
become stressed when the demand for protein folding exceeds its that the absence of C3a receptor significantly protected kidney
capacity. Studies suggest that proteinuria leads to endoplasmic function via considerable reduction of inflammatory cell infiltra-
reticulum stress via at least two molecular processes, including tion, tubulointerstitial injury and inflammation [85]. In this study,
increased ROS and intracellular calcium levels in PTECs (Fig. 2) complement receptor 1–related gene/protein-y (Crry) is a rodent-
[77]. Endoplasmic reticulum stress is characterized by dysfunc- specific complement inhibitor that was knocked out along with C3
tional endoplasmic reticulum and accumulation of unfolded to prevent embryonic lethality due to uncontrolled complement
proteins in the endoplasmic reticulum lumen [78], which triggers activation [85]. Furthermore, experiments using primary and sec-
an adaptive response called unfolded protein response [79]. If this ondary human cultures demonstrated that C3a induces the ex-
response is insufficient or sustained due to the toxicity of chronic pression of type I collagen and TGF-β1 in PTECs, suggesting that
proteinuria, it fails to overcome endoplasmic reticulum stress and C3a may be an essential player in interstitial fibrosis and CKD pro-
triggers apoptotic pathways to eliminate damaged cells that even- gression [86, 87].
tually exacerbate tubulointerstitial injury [77]. Ohse et al. demon- Moreover, a previous study with C5aR−/− MRL mice supports the
strated, using immortalized rat PTECs and an animal model of hypothesis that C5a can contribute to tubulointerstitial inflam-
massive proteinuria, puromycin aminonucleoside nephropathy, mation and fibrosis development [88]. Similar to C3a, C5a may
that albumin exposure to PETCs markedly increased endoplasmic also facilitate tubulointerstitial fibrosis formation via direct ef-
reticulum stress proteins, including glucose-regulated protein 78 fects on interstitial fibroblasts [89]. Unlike the two anaphylatoxins,
and oxygen-regulated protein 150, which led to calpain-mediated the insertion of membrane attack complex into the PTEC mem-
caspase-12 activation and thus endoplasmic reticulum stress– brane upregulates the synthesis of ROS, tumor necrosis factor–
induced cell apoptosis [23]. Later, it was shown that albumin- alpha and interleukin 6, which altogether cause tubulointerstitial
induced unfolded protein response causes tubular apoptosis and damage and collagen synthesis associated with tubulointerstitial
lesions by Lipocalin-2 modulation via activating transcription fibrosis development [90, 91]. Also, the urinary membrane attack
factor 4 in corporation with CCAAT/enhancer-binding protein ho- complex is a potent inducer of PTEC cytolysis which disrupts the
mologous protein in experimental proteinuric mice [77]. Notably, cellular actin network [91].
the inhibition of endoplasmic reticulum stress by 4-phenylbutyric In addition, other filtered plasma proteins can also facilitate
acid protected kidneys of mice from proteinuria-induced renal complement activation in the tubular lumen. For example, apical
lesions and Lipocalin-2 overexpression, and thus inhibited CKD exposure of urinary transferrin on PTECs upregulates cellular
progression [77]. Furthermore, endoplasmic reticulum stress– biosynthesis of complement C3 and stimulates its secretion
associated pro-apoptotic pathways may enter a vicious cycle of [30]. Besides, urinary albumin assists with properdin-mediated
causing irreversible cell death by inducing mitochondrial calcium assembly and docking of C3 convertase at the surface of PTECs
increase, dysfunction and pro-apoptotic factor secretion [80]. by disturbing cell binding with factor H, a crucial regulator of
Overall, tubular toxicity of proteins leads to severe endoplasmic alternative pathways [92]. Thus, proteinuria-mediated comple-
reticulum stress that substantially contributes to tubular cell ment activation in the tubular lumen is one of the main drivers
loss and tubule atrophy during CKD progression. of tubular injury, interstitial fibrosis and CKD progression.
8 | Nephrol Dial Transplant, 2023, Vol. 0, No. 0

Proteinuria
C3a, C5a, MAC

Proximal tubular
epithelial cell

C5a C3a MAC Properdin


Complement molecules

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


C3 convertase
↑ TGF-β1 Collagen ↑ROS Proteins
Cytolysis
↑TNF-α Factor H
↑IL-6
↑ Increase
Induce
Inhibit
Fibrosis Inflammation
Movement

Figure 3: The roles of proteinuria-mediated activated complement products in PTECs. Complement molecules are not filtered from the glomerulus but
during proteinuria, these molecules appear in the tubular lumen and can activate the alternative pathway of the complement system. Properdin along
with filtered other complement molecules leads to complement activation through binding to PTECs and functioning as a docking station for the
assembly of alternative pathway C3 convertase. Properdin stabilizes C3 convertase and enhances AP amplification leading to the generation of C3a
and C5a anaphylatoxins and MAC. C3a promotes tubulointerstitial fibrosis by inducing the synthesis of collagen and TGF-β1 in PTECs. C5a promotes
tubulointerstitial fibrosis by TGF-β1 induction. MAC can contribute to the development of tubulointerstitial fibrosis by inducing collagen synthesis. In
addition, MAC induces ROS, TNF-α and IL-6, as well as promoting tubulointerstitial injury and cytolysis of PTECs. Furthermore, filtered plasma
proteins can facilitate tubular complement activation except for filtered complement components. For example, urinary transferrin induces C3
expression and biosynthesis in PTECs, which is secreted into the tubular lumen. Besides, urinary albumin assists with properdin-mediated assembly
and docking of C3 convertase at the surface of PTECs by inhibiting tubular cell binding with factor H. IL-6, interleukin 6; MAC, membrane attack
complex; TNF-α, tumor necrosis factor-alpha.

CONCLUSION AUTHORS’ CONTRIBUTIONS


Proteinuria is a sensitive biomarker of CKD progression. Protein- All authors contributed to researching data for the article, dis-
uria can be nephrotoxic directly due to toxicity to PTECs and in- cussing the article’s content, writing the article, and reviewing or
directly by increasing metabolic stress. Therefore, identifying and editing the manuscript before submission.
establishing a list of toxic proteins with their state of toxicity on
PTECs can pave the way for identifying reliable specific biomark-
ers in the early stages of CKD and improves our understanding of DATA AVAILABILITY STATEMENT
disease progression. The most reported molecular mechanisms No new data were generated or analyzed in support of this
in proteinuria that promote kidney tubular damage, interstitial research.
inflammation and fibrosis include the increase of intracellular
ROS, autophagy dysfunction, lysosomal membrane permeabiliza-
tion, endoplasmic reticulum stress and complement activation. CONFLICT OF INTEREST STATEMENT
Of note, these mechanisms are consequences of increased reab-
The authors declare no conflict of interests.
sorption of filtered proteins and/or FFAs bound to albumin. Thus,
further research is needed to elucidate the effects of reducing
the metabolic workload of PTECs by blocking endocytic receptors
REFERENCES
and whether blocking protein reabsorption suppresses protein cy-
totoxicity and halts CKD progression. In addition, it remains to 1. Zoccali C, Vanholder R, Massy ZA et al. The systemic nature
be studied whether blocking protein uptake into PTECs can elicit of CKD. Nat Rev Nephrol 2017;13:344–58. https://doi.org/10.1038/
similar nephroprotective effects as blocking uptake of sodium and nrneph.2017.52
glucose with sodium-glucose cotransporter 2 inhibitors, e.g. with 2. Hill NR, Fatoba ST, Oke JL et al. Global prevalence of
cubulin antagonists. chronic kidney disease – a systematic review and meta-
analysis. PLoS One 2016;11:e0158765. https://doi.org/10.1371/
journal.pone.0158765
3. Romagnani P, Remuzzi G, Glassock R et al. Chronic kidney dis-
SUPPLEMENTARY DATA ease. Nat Rev Dis Primers 2017;3:17088. https://doi.org/10.1038/
Supplementary data are available at ndt online. nrdp.2017.88
4. Thomas B, Matsushita K, Abate KH et al. Global cardiovas-
cular and renal outcomes of reduced GFR. J Am Soc Nephrol
2017;28:2167–79. https://doi.org/10.1681/ASN.2016050562
FUNDING 5. Nitsch D, Grams M, Sang Y et al. Associations of estimated
The authors of this review acknowledge funding from the glomerular filtration rate and albuminuria with mortality and
Nazarbayev University Collaborative Research Program (CRP) for renal failure by sex: a meta-analysis. BMJ 2013;346:f324. https:
2020–2022 (Funder Project Reference: 091019CRP2105). //doi.org/10.1136/bmj.f324
Z. Makhammajanov et al. | 9

6. Heerspink HJL, Kröpelin TF, Hoekman J et al. Drug-induced re- nal proximal tubule. Int J Mol Sci 2019;20:2379. https://doi.org/10.
duction in albuminuria is associated with subsequent renopro- 3390/ijms20102379
tection: a meta-analysis. J Am Soc Nephrol 2015;26:2055. https: 25. Fassett RG, Venuthurupalli SK, Gobe GC et al. Biomarkers in
//doi.org/10.1681/ASN.2014070688 chronic kidney disease: a review. Kidney Int 2011;80:806–21.
7. Liu D, Lv LL. New understanding on the role of proteinuria https://doi.org/10.1038/ki.2011.198
in progression of chronic kidney disease. Adv Exp Med Biol 26. Gansevoort RT, Matsushita K, van der Velde M et al. Lower es-
2019;1165:487–500. https://doi.org/10.1007/978- 981- 13- 8871- 2_ timated GFR and higher albuminuria are associated with ad-
24 verse kidney outcomes. A collaborative meta-analysis of gen-

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


8. Gaipov A, Makhammajanov Z, Dauyey Z et al. Urinary protein eral and high-risk population cohorts. Kidney Int 2011;80:93–104.
profiling for potential biomarkers of chronic kidney disease: https://doi.org/10.1038/ki.2010.531
a pilot study. Diagnostics 2022;12:2583. https://doi.org/10.3390/ 27. Khan S, Cabral PD, Schilling WP et al. Kidney proximal tubule
diagnostics12112583 lipoapoptosis is regulated by fatty acid transporter-2 (FATP2).
9. Sirac C, Batuman V, Sanders PW. The proximal tubule toxicity J Am Soc Nephrol 2018;29:81–91. https://doi.org/10.1681/ASN.
of immunoglobulin light chains. Kidney Int Rep 2021;6:1225–31. 2017030314
https://doi.org/10.1016/j.ekir.2021.02.026 28. Nishi Y, Satoh M, Nagasu H et al. Selective estrogen receptor
10. Bosch X, Poch E, Grau JM. Rhabdomyolysis and acute kid- modulation attenuates proteinuria-induced renal tubular dam-
ney injury. N Engl J Med 2009;361:62–72. https://doi.org/10.1056/ age by modulating mitochondrial oxidative status. Kidney Int
NEJMra0801327 2013;83:662–73. https://doi.org/10.1038/ki.2012.475
11. Van Avondt K, Nur E, Zeerleder S. Mechanisms of haemolysis- 29. Chen Y, Yan Q, Lv M et al. Involvement of FATP2-mediated tubu-
induced kidney injury. Nat Rev Nephrol 2019;15:671–92. lar lipid metabolic reprogramming in renal fibrogenesis. Cell
12. Tojo A, Kinugasa S. Mechanisms of glomerular albumin filtra- Death Dis 2020;11:994.
tion and tubular reabsorption. Int J Nephrol 2012;2012:1–9. https: 30. Tang S, Lai KN, Chan TM et al. Transferrin but not albumin me-
//doi.org/10.1155/2012/481520 diates stimulation of complement C3 biosynthesis in human
13. Christensen EI, Birn H, Storm T et al. Endocytic receptors proximal tubular epithelial cells. Am J Kidney Dis 2001;37:94–103.
in the renal proximal tubule. Physiology (Bethesda) 2012;27: https://doi.org/10.1053/ajkd.2001.20593
223–36. 31. Sanchez-Hidalgo JJ, Suarez-Cuenca JA, Lozano-Nuevo JJ et al.
14. Nolin AC, Mulhern RM, Panchenko MV et al. Proteinuria causes Urine transferrin as an early endothelial dysfunction marker
dysfunctional autophagy in the proximal tubule. Am J Physiol in type 2 diabetic patients without nephropathy: a case control
Renal Physiol 2016;311:F1271–9. https://doi.org/10.1152/ajprenal. study. Diabetol Metab Syndr 2021;13:128. https://doi.org/10.1186/
00125.2016 s13098- 021- 00745- 1
15. Tojo A, Kinugasa S. Mechanisms of glomerular albumin filtration 32. Hellemons ME, Kerschbaum J, Bakker SJ et al. Validity of
and tubular reabsorption. Int J Nephrol 2012;2012:481520. https: biomarkers predicting onset or progression of nephropathy
//doi.org/10.1155/2012/481520 in patients with type 2 diabetes: a systematic review. Diabet
16. Aitekenov S, Gaipov A, Bukasov R. Review: detection and quan- Med 2012;29:567–77. https://doi.org/10.1111/j.1464-5491.2011.
tification of proteins in human urine. Talanta 2021;223:121718. 03437.x
https://doi.org/10.1016/j.talanta.2020.121718 33. Kobayashi N, Suzuki Y, Tsuge T et al. FcRn-mediated transcyto-
17. Marimuthu A, O’Meally RN, Chaerkady R et al. A compre- sis of immunoglobulin G in human renal proximal tubular ep-
hensive map of the human urinary proteome. J Proteome Res ithelial cells. Am J Physiol Renal Physiol 2002;282:F358–65. https:
2011;10:2734–43. https://doi.org/10.1021/pr2003038 //doi.org/10.1152/ajprenal.0164.2001
18. Santucci L, Candiano G, Petretto A et al. From hundreds 34. Tencer J, Bakoush O, Torffvit O. Diagnostic and prognostic sig-
to thousands: widening the normal human urinome. J Pro- nificance of proteinuria selectivity index in glomerular dis-
teomics 2015;112:53–62. https://doi.org/10.1016/j.jprot.2014.07 eases. Clin Chim Acta 2000;297:73–83. https://doi.org/10.1016/
.021 S0009- 8981(00)00235- 7
19. Benzing T, Salant D. Insights into glomerular filtration and albu- 35. Abbate M, Zoja C, Corna D et al. Complement-mediated dysfunc-
minuria. N Engl J Med 2021;384:1437–46. https://doi.org/10.1056/ tion of glomerular filtration barrier accelerates progressive renal
NEJMra1808786 injury. J Am Soc Nephrol 2008;19:1158–67. https://doi.org/10.1681/
20. Liu BC, Tang TT, Lv LL et al. Renal tubule injury: a driving force ASN.2007060686
toward chronic kidney disease. Kidney Int 2018;93:568–79. https: 36. Farrar CA, Zhou W, Lin T et al. Local extravascular pool of C3
//doi.org/10.1016/j.kint.2017.09.033 is a determinant of postischemic acute renal failure. FASEB J
21. Bondi CD, Manickam N, Lee DY et al. NAD(P)H oxidase me- 2006;20:217–26. https://doi.org/10.1096/fj.05-4747com
diates TGF-beta1-induced activation of kidney myofibroblasts. 37. Chen SF, Chen M. Complement activation in progression of
J Am Soc Nephrol 2010;21:93–102. https://doi.org/10.1681/ASN. chronic kidney disease. Adv Exp Med Biol 2019;1165:423–41.
2009020146 https://doi.org/10.1007/978- 981- 13- 8871- 2_20
22. Toblli JE, Bevione P, Di Gennaro F et al. Understanding the mech- 38. Kalantari S, Rutishauser D, Samavat S et al. Urinary prognos-
anisms of proteinuria: therapeutic implications. Int J Nephrol tic biomarkers and classification of IgA nephropathy by high
2012;2012:546039. https://doi.org/10.1155/2012/546039 resolution mass spectrometry coupled with liquid chromatog-
23. Ohse T, Inagi R, Tanaka T et al. Albumin induces endoplas- raphy. PLoS One 2013;8:e80830. https://doi.org/10.1371/journal.
mic reticulum stress and apoptosis in renal proximal tubular pone.0080830
cells. Kidney Int 2006;70:1447–55. https://doi.org/10.1038/sj.ki. 39. Ito S, Fujita H, Narita T et al. Urinary copper excretion in type
5001704 2 diabetic patients with nephropathy. Nephron 2001;88:307–12.
24. Fels J, Scharner B, Zarbock R et al. Cadmium complexed with https://doi.org/10.1159/000046013
beta2-microglubulin, albumin and lipocalin-2 rather than met- 40. Bazzi C, Petrini C, Rizza V et al. Urinary N-acetyl-beta-
allothionein cause megalin:cubilin dependent toxicity of the re- glucosaminidase excretion is a marker of tubular cell
10 | Nephrol Dial Transplant, 2023, Vol. 0, No. 0

dysfunction and a predictor of outcome in primary binding proteins. Biochim Biophys Acta 1991;1081:1–24. https://
glomerulonephritis. Nephrol Dial Transplant 2002;17:1890–6. doi.org/10.1016/0005- 2760(91)90244- C
https://doi.org/10.1093/ndt/17.11.1890 57. Erkan E, Devarajan P, Schwartz GJ. Mitochondria are the major
41. Kern EF, Erhard P, Sun W et al. Early urinary markers of dia- targets in albumin-induced apoptosis in proximal tubule cells.
betic kidney disease: a nested case-control study from the Di- J Am Soc Nephrol 2007;18:1199–208. https://doi.org/10.1681/ASN.
abetes Control and Complications Trial (DCCT). Am J Kidney Dis 2006040407
2010;55:824–34. https://doi.org/10.1053/j.ajkd.2009.11.009 58. Jha JC, Thallas-Bonke V, Banal C et al. Podocyte-specific Nox4
42. Wang Y, Zheng C, Xu F et al. Urinary fibrinogen and renal tubu- deletion affords renoprotection in a mouse model of dia-

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


lointerstitial fibrinogen deposition: discriminating between pri- betic nephropathy. Diabetologia 2016;59:379–89. https://doi.org/
mary FSGS and minimal change disease. Biochem Biophys Res 10.1007/s00125- 015- 3796- 0
Commun 2016;478:1147–52. https://doi.org/10.1016/j.bbrc.2016. 59. Papadimitriou A, Peixoto EB, Silva KC et al. Increase in AMPK
08.083 brought about by cocoa is renoprotective in experimental di-
43. Sorensen I, Susnik N, Inhester T et al. Fibrinogen, acting as a mi- abetes mellitus by reducing NOX4/TGFbeta-1 signaling. J Nutr
togen for tubulointerstitial fibroblasts, promotes renal fibrosis. Biochem 2014;25:773–84. https://doi.org/10.1016/j.jnutbio.2014.
Kidney Int 2011;80:1035–44. https://doi.org/10.1038/ki.2011.214 03.010
44. Wang H, Zheng C, Lu Y et al. Urinary fibrinogen as a predictor of 60. Jha JC, Gray SP, Barit D et al. Genetic targeting or pharmacologic
progression of CKD. Clin J Am Soc Nephrol 2017;12:1922–9. https: inhibition of NADPH oxidase nox4 provides renoprotection in
//doi.org/10.2215/CJN.01360217 long-term diabetic nephropathy. J Am Soc Nephrol 2014;25:1237–
45. Nielsen R, Christensen EI, Birn H. Megalin and cubilin in prox- 54. https://doi.org/10.1681/ASN.2013070810
imal tubule protein reabsorption: from experimental models 61. Aljofan M, Ding H. High glucose increases expression of
to human disease. Kidney Int 2016;89:58–67. https://doi.org/10. cyclooxygenase-2, increases oxidative stress and decreases the
1016/j.kint.2015.11.007 generation of nitric oxide in mouse microvessel endothelial
46. Vinge L, Lees GE, Nielsen R et al. The effect of progressive cells. J Cell Physiol 2010;222:669–75.
glomerular disease on megalin-mediated endocytosis in the kid- 62. Wolf G, Schroeder R, Ziyadeh FN et al. Albumin up-regulates
ney. Nephrol Dial Transplant 2010;25:2458–67. https://doi.org/10. the type II transforming growth factor-beta receptor in cultured
1093/ndt/gfq044 proximal tubular cells. Kidney Int 2004;66:1849–58. https://doi.
47. Caruso-Neves C, Pinheiro AAS, Cai H et al. PKB and megalin de- org/10.1111/j.1523-1755.2004.00958.x
termine the survival or death of renal proximal tubule cells. 63. Stephan JP, Mao W, Filvaroff E et al. Albumin stimulates the
Proc Natl Acad Sci USA 2006;103:18810–5. https://doi.org/10.1073/ accumulation of extracellular matrix in renal tubular epithe-
pnas.0605029103 lial cells. Am J Nephrol 2004;24:14–9. https://doi.org/10.1159/
48. Gekle M, Mildenberger S, Freudinger R et al. Long-term pro- 000075347
tein exposure reduces albumin binding and uptake in prox- 64. Goumenos DS, Tsakas S, El Nahas AM et al. Transforming growth
imal tubule-derived opossum kidney cells. J Am Soc Nephrol factor-β1 in the kidney and urine of patients with glomerular
1998;9:960–8. https://doi.org/10.1681/ASN.V96960 disease and proteinuria. Nephrol Dial Transplant 2002;17:2145–52.
49. Fatah H, Benfaed N, Chana RS et al. Reduced proximal tubular https://doi.org/10.1093/ndt/17.12.2145
expression of protein endocytic receptors in proteinuria is as- 65. Boya P, Reggiori F, Codogno P. Emerging regulation and func-
sociated with urinary receptor shedding. Nephrol Dial Transplant tions of autophagy. Nat Cell Biol 2013;15:713–20. https://doi.org/
2018;33:934–43. https://doi.org/10.1093/ndt/gfx321 10.1038/ncb2788
50. Seki T, Asanuma K, Asao R et al. Significance of urinary full- 66. Gong L, Pan Q, Yang N. Autophagy and inflammation regulation
length megalin in patients with IgA nephropathy. PLoS One in acute kidney injury. Front Physiol 2020;11:576463. https://doi.
2014;9:e114400. https://doi.org/10.1371/journal.pone.0114400 org/10.3389/fphys.2020.576463
51. Bedin M, Boyer O, Servais A et al. Human C-terminal CUBN 67. Tang C, Livingston MJ, Liu Z et al. Autophagy in kidney homeosta-
variants associate with chronic proteinuria and normal renal sis and disease. Nat Rev Nephrol 2020;16:489–508. https://doi.org/
function. J Clin Invest 2020;130:335–44. https://doi.org/10.1172/ 10.1038/s41581- 020- 0309- 2
JCI129937 68. Liu WJ, Luo MN, Tan J et al. Autophagy activation reduces
52. Morigi M, Macconi D, Zoja C et al. Protein overload-induced renal tubular injury induced by urinary proteins. Autophagy
NF-kappaB activation in proximal tubular cells requires 2014;10:243–56. https://doi.org/10.4161/auto.27004
H(2)O(2) through a PKC-dependent pathway. J Am Soc Nephrol 69. Tan J, Wang M, Song S et al. Autophagy activation promotes
2002;13:1179–89. removal of damaged mitochondria and protects against renal
53. Ruggiero C, Elks CM, Kruger C et al. Albumin-bound fatty acids tubular injury induced by albumin overload. Histol Histopathol
but not albumin itself alter redox balance in tubular epithelial 2018;33:681–90.
cells and induce a peroxide-mediated redox-sensitive apoptosis. 70. Xu D, Chen P, Wang B et al. NIX-mediated mitophagy protects
Am J Physiol Renal Physiol 2014;306:F896–906. https://doi.org/10. against proteinuria-induced tubular cell apoptosis and renal in-
1152/ajprenal.00484.2013 jury. Am J Physiol Renal Physiol 2019;316:F382–95. https://doi.org/
54. van Timmeren MM, Bakker SJ, Stegeman CA et al. Addition of 10.1152/ajprenal.00360.2018
oleic acid to delipidated bovine serum albumin aggravates re- 71. Zhan M, Usman IM, Sun L et al. Disruption of renal tubular mi-
nal damage in experimental protein-overload nephrosis. Nephrol tochondrial quality control by myo-inositol oxygenase in dia-
Dial Transplant 2005;20:2349–57. betic kidney disease. J Am Soc Nephrol 2015;26:1304–21. https:
55. Ghiggeri GM, Ginevri F, Candiano G et al. Characterization of //doi.org/10.1681/ASN.2014050457
cationic albumin in minimal change nephropathy. Kidney Int 72. Galluzzi L, Vitale I, Aaronson SA et al. Molecular mechanisms of
1987;32:547–53. https://doi.org/10.1038/ki.1987.243 cell death: recommendations of the Nomenclature Committee
56. Veerkamp JH, Peeters RA, Maatman RG. Structural and func- on Cell Death 2018. Cell Death Differ 2018;25:486–541. https://doi.
tional features of different types of cytoplasmic fatty acid- org/10.1038/s41418- 017- 0012- 4
Z. Makhammajanov et al. | 11

73. Saftig P, Klumperman J. Lysosome biogenesis and lysosomal munol 2007;179:2600–8. https://doi.org/10.4049/jimmunol.179.4.
membrane proteins: trafficking meets function. Nat Rev Mol Cell 2600
Biol 2009;10:623–35. https://doi.org/10.1038/nrm2745 84. Lammerts RGM, Talsma DT, Dam WA et al. Properdin pat-
74. Wang F, Gómez-Sintes R, Boya P. Lysosomal membrane perme- tern recognition on proximal tubular cells is heparan
abilization and cell death. Traffic 2018;19:918–31. https://doi.org/ sulfate/syndecan-1 but not C3b dependent and can be
10.1111/tra.12613 blocked by tick protein Salp20. Front Immunol 2020;11:1643.
75. Liu WJ, Xu BH, Ye L et al. Urinary proteins induce lysoso- https://doi.org/10.3389/fimmu.2020.01643
mal membrane permeabilization and lysosomal dysfunction 85. Bao L, Wang Y, Haas M et al. Distinct roles for C3a and

Downloaded from https://academic.oup.com/ndt/advance-article/doi/10.1093/ndt/gfad215/7288664 by Universidad del Tolima user on 30 October 2023


in renal tubular epithelial cells. Am J Physiol Renal Physiol C5a in complement-induced tubulointerstitial injury. Kidney Int
2015;308:F639–49. https://doi.org/10.1152/ajprenal.00383. 2011;80:524–34. https://doi.org/10.1038/ki.2011.158
2014 86. Braun MC, Reins RY, Li TB et al. Renal expression of the C3a re-
76. Chen XC, Li ZH, Yang C et al. Lysosome depletion-triggered au- ceptor and functional responses of primary human proximal
tophagy impairment in progressive kidney injury. Kidney Dis tubular epithelial cells. J Immunol 2004;173:4190–6. https://doi.
(Basel) 2021;7:254–67. https://doi.org/10.1159/000515035 org/10.4049/jimmunol.173.6.4190
77. El Karoui K, Viau A, Dellis O et al. Endoplasmic reticulum stress 87. Peake PW, O’Grady S, Pussell BA et al. C3a is made by proxi-
drives proteinuria-induced kidney lesions via Lipocalin 2. Nat mal tubular HK-2 cells and activates them via the C3a receptor.
Commun 2016;7:10330. Kidney Int 1999;56:1729–36. https://doi.org/10.1046/j.1523-1755.
78. Kaufman RJ. Stress signaling from the lumen of the endoplas- 1999.00722.x
mic reticulum: coordination of gene transcriptional and trans- 88. Wenderfer SE, Ke B, Hollmann TJ et al. C5a receptor deficiency
lational controls. Genes Dev 1999;13:1211–33. https://doi.org/10. attenuates T cell function and renal disease in MRLlpr mice.
1101/gad.13.10.1211 J Am Soc Nephrol 2005;16:3572–82. https://doi.org/10.1681/ASN.
79. Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and func- 2005040373
tions of the unfolded protein response. Nat Rev Mol Cell Biol 89. Boor P, Konieczny A, Villa L et al. Complement C5 medi-
2020;21:421–38. https://doi.org/10.1038/s41580- 020- 0250- z ates experimental tubulointerstitial fibrosis. J Am Soc Nephrol
80. Bravo-Sagua R, Rodriguez AE, Kuzmicic J et al. Cell death 2007;18:1508–15. https://doi.org/10.1681/ASN.2006121343
and survival through the endoplasmic reticulum-mitochondrial 90. Abe K, Li K, Sacks SH et al. The membrane attack complex, C5b-9,
axis. Curr Mol Med 2013;13:317–29. https://doi.org/10.2174/ up regulates collagen gene expression in renal tubular epithelial
156652413804810781 cells. Clin Exp Immunol 2004;136:60–6. https://doi.org/10.1111/j.
81. Matsuo S, Morita Y, Mizuno M et al. Proteinuria and damage to 1365-2249.2004.02411.x
tubular cells—is complement a culprit? Nephrol Dial Transplant 91. Biancone L, David S, Della Pietra V et al. Alternative path-
1998;13:2723–6. https://doi.org/10.1093/ndt/13.11.2723 way activation of complement by cultured human proximal
82. Zhou W, Marsh JE, Sacks SH. Intrarenal synthesis of com- tubular epithelial cells. Kidney Int 1994;45:451–60. https://doi.
plement. Kidney Int 2001;59:1227–35. https://doi.org/10.1046/j. org/10.1038/ki.1994.59
1523-1755.2001.0590041227.x 92. Zaferani A, Vives RR, van der Pol P et al. Factor h and properdin
83. Spitzer D, Mitchell LM, Atkinson JP et al. Properdin can initi- recognize different epitopes on renal tubular epithelial heparan
ate complement activation by binding specific target surfaces sulfate. J Biol Chem 2012;287:31471–81. https://doi.org/10.1074/
and providing a platform for de novo convertase assembly. J Im- jbc.M112.380386

Received: June 16, 2023; Editorial decision: September 4, 2023


© The Author(s) 2023. Published by Oxford University Press on behalf of the ERA. All rights reserved. For permissions, please e-mail:
journals.permissions@oup.com

You might also like