You are on page 1of 22

REVIEWS

Endothelium structure and function


in kidney health and disease
Noemie Jourde-​Chiche1,2*, Fadi Fakhouri3, Laetitia Dou2, Jeremy Bellien4,
Stéphane Burtey1,2, Marie Frimat5,6, Pierre-​André Jarrot2,7, Gilles Kaplanski2,7,
Moglie Le Quintrec8,9, Vincent Pernin8,9, Claire Rigothier10,11, Marion Sallée1,2,
Veronique Fremeaux-​Bacchi12,13, Dominique Guerrot 14 and Lubka T. Roumenina 13,15,16
*
Abstract | The kidney harbours different types of endothelia, each with specific structural and
functional characteristics. The glomerular endothelium, which is highly fenestrated and covered
by a rich glycocalyx, participates in the sieving properties of the glomerular filtration barrier
and in the maintenance of podocyte structure. The microvascular endothelium in peritubular
capillaries, which is also fenestrated, transports reabsorbed components and participates in
epithelial cell function. The endothelium of large and small vessels supports the renal vasculature.
These renal endothelia are protected by regulators of thrombosis, inflammation and complement,
but endothelial injury (for example, induced by toxins, antibodies, immune cells or inflammatory
cytokines) or defects in factors that provide endothelial protection (for example, regulators
of complement or angiogenesis) can lead to acute or chronic renal injury. Moreover, renal
endothelial cells can transition towards a mesenchymal phenotype, favouring renal fibrosis and
the development of chronic kidney disease. Thus, the renal endothelium is both a target and a
driver of kidney and systemic cardiovascular complications. Emerging therapeutic strategies
that target the renal endothelium may lead to improved outcomes for both rare and common
renal diseases.

The endothelium constitutes the inner monolayer of context of common diseases such as diabetes melli-
cells that lines blood and lymphatic vessels. It was tradi- tus and hypertension, in rare diseases such as atypical
tionally considered to act as a simple conduit for trans- haemolytic uraemic syndrome (aHUS) and glomer-
porting blood, as a barrier to separate blood from the ulonephritides related to systemic diseases2 and in
vessel wall and in the regulation of vessel permeability situations associated with acute kidney injury (AKI))3.
and diapedesis of immune cells at sites of inflammation. In addition, the accumulation of uraemic toxins that
However, we now understand that the endothelium is occurs with kidney dysfunction contributes to endothe-
a dynamic organ. Different populations of endothelial lial dysfunction and the cardiovascular burden of
cells (ECs) exist, each with different architectures and patients with chronic kidney disease (CKD)4,5. The
functions that together promote an antithrombotic transition of renal ECs towards a mesenchymal phe-
and anti-​inflammatory environment and maintain tissue notype in a process called endothelial-​to-mesenchymal
perfusion and vascular tone (Figs. 1,2). transition (EndMT) promotes renal fibrosis and the
Uraemic toxins The kidney is a highly vascularized organ, character- development of CKD. In addition, systemic endothe-
Blood compounds that are
ized by a remarkable diversity of EC populations1 (Fig. 3). lial dysfunction and accelerated atherosclerosis result-
normally eliminated in the
urine but accumulate in ECs from the microvasculature, in particular, regulate ing from CKD might itself accelerate decline in renal
patients with chronic kidney blood flow to local tissue beds and modulate coagula- function. Thus, endothelial damage in the context of
disease or acute kidney injury tion, inflammation and vascular permeability. Beyond kidney dysfunction can drive further kidney injury and
and exert deleterious effects. these functions, the ECs of the glomerulus contribute systemic complications.
to the glomerular filtration barrier and support podo- Improved understanding of the role of endothe-
cyte structure, whereas ECs of the microvasculature lial injury in renal disease has paved the way for new
*e-​mail: noemie.jourde@
in kidney tubules contribute to tubular reabsorption. therapeutic strategies (for example, complement block-
ap-hm.fr; lubka.roumenina@
crc.jussieu.fr These varied functions of ECs make them key driv- ade in aHUS) that target the endothelium. The fur-
https://doi.org/10.1038/ ers and targets of inflammatory and thrombotic pro- ther development of strategies to protect EC structure
s41581-018-0098-z cesses that can damage the kidney (for example, in the and function has the potential to reverse coagulation and

Nature Reviews | Nephrology


Reviews

Key points Renal endothelia


The environment of renal ECs
• The kidney contains diverse populations of endothelial cells, including the The phenotype and properties of ECs vary between vas-
glomerular endothelium, microvascular endothelium in peritubular capillaries and cular beds according to their location and microenviron-
the endothelium of large and small vessels, and each of these populations has specific
ment. In vivo, the vascular endothelium is continuously
characteristics and functions.
subjected to the oscillating mechanical shear stress of
• Homeostasis of renal endothelial cells is crucial for the preservation of glomerular blood flow, the extent of which depends on the type
structure and function, the preservation of an anti-​inflammatory and an
of blood vessel and its geometry6. This shear stress is a
antithrombotic environment and the prevention of renal fibrosis.
major determinant of EC morphology, function, gene
• Glomerular endothelial cells, in particular, are susceptible to injury in typical and
expression and behaviour6–8.
atypical haemolytic uraemic syndrome, lupus nephritis, antineutrophil cytoplasmic
antibody vasculitides and antibody-​mediated rejection as well as in situations of
The kidney harbours a wide variety of ECs, includ-
vascular endothelial growth factor (VEGF) depletion. ing glomerular ECs (GECs), microvascular ECs within
• Common forms of chronic kidney disease (CKD) — diabetic kidney disease and
peritubular capillaries and ECs within larger venous and
arteriolar nephrosclerosis — are also characterized by renal endothelial dysfunction. arterial blood vessels (Fig. 3). These populations of ECs
• Alterations in endothelial repair capacity, endothelial-​to-mesenchymal transition
are exposed to different environments within the kidney
and capillary rarefaction contribute to the fibrogenic processes that lead to CKD. (for example, different oxygen pressures and osmolalities
• Therapeutic strategies aimed at preserving and/or restoring the integrity of the
within the kidney). Importantly, renal ECs contribute
endothelial glycocalyx, reversing the procoagulant and pro-​inflammatory to different transport activities: GECs are involved in
phenotype of injured endothelial cells and slowing renal fibrosis hold promise for glomerular filtration whereas ECs of peritubular capil-
the treatment of renal disease. laries are involved in tubular secretion and reabsorption.
Both GECs and peritubular ECs are fenestrated,
although ultrastructural differences exist between them9.
inflammatory processes in other glomerulonephritides This fenestration is critical for the permselectivity of the
and to slow the progression of CKD in patients with dia- glomerular filtration barrier and for the efficient passage
betic kidney disease (DKD) or arteriolar nephrosclero- of high volumes of fluids and the formation of urine.
sis. Here, we describe the unique features of the healthy GECs are lined by a particularly thick filamentous
renal endothelium and the pathological mechanisms glycocalyx that is enriched in negatively charged pro-
leading to endothelial damage in emblematic renal teoglycans (mostly heparan and chondroitin sulfate)
diseases. We discuss the processes and consequences of that form a network with glycosaminoglycans (mostly
renal EndMT and the effects of CKD and uraemic toxins hyaluronic acid). The glycocalyx contributes to the reg-
on the systemic endothelium. We also describe the role ulation of vascular permeability and fluidic balance and
of the renal endothelium in the initiation and progres- repels blood cells away from the vascular wall10. Densely
sion of the two most frequent kidney diseases — DKD packed hyaluronic acid anchors the glycocalyx to the glo-
and arteriolar nephrosclerosis. merular basement membrane (GBM) and fills endothe-
lial fenestrae, preventing the passage of albumin11. The
thickness of the glycocalyx exceeds the dimensions
Author addresses of cellular adhesion molecules such as intercellular
1
Aix-​Marseille University, Centre de Nephrologie et Transplantation Renale, AP-​HM adhesion molecule 1 (ICAM1), vascular cell adhesion
Hopital de la Conception, Marseille, France. protein 1 (VCAM1), P-​selectins and E-selectins, which
2
Aix-​Marseille University, C2VN, INSERM 1263, Institut National de la Recherche are expressed by ECs, thus hampering cell adhesion to
Agronomique (INRA) 1260, Faculte de Pharmacie, Marseille, France. the endothelium.
3
Centre de Recherche en Transplantation et Immunologie, INSERM, Université de ECs of peritubular capillaries have a thin stroma.
Nantes and Department of Nephrology, Centre Hospitalier Universitaire de Nantes, As seen with electron microscopy, EC fenestrations are
Nantes, France. covered by a thin diaphragm, which is composed of gly-
4
Department of Pharmacology, Rouen University Hospital and INSERM, Normandy coproteins organized in radial fibrils and modulates the
University, Université de Rouen Normandie, Rouen, France. sieving properties of these ECs12,13. These ECs lie on a
5
Université de Lille, INSERM, Centre Hospitalier Universitaire de Lille, U995, Lille
basement membrane, on the opposite side of which are
Inflammation Research International Center (LIRIC), Lille, France.
6
Nephrology Department, Centre Hospitalier Universitaire de Lille, Lille, France. pericytes. These pericytes14 drive capillary constriction
7
Assistance Publique-​Hôpitaux de Marseille, Service de Médecine Interne et to regulate medullary and cortical blood flow; they also
d’Immunologie Clinique, Hôpital de La Conception, Marseille, France. regulate capillary permeability, participate in angiogene-
8
Centre Hospitalier Universitaire de Lapeyronie, Département de Néphrologie Dialyse et sis and are a source of myofibroblasts in renal fibrogene-
Transplantation Rénale, Montpellier, France. sis15. Peritubular microvascular ECs also provide a niche
9
Institute for Regenerative Medicine and Biotherapy (IRMB), Montpellier, France. for resident organ stem cells, which reside in close prox-
10
Tissue Bioengineering, Université de Bordeaux, Bordeaux, France. imity and benefit from the paracrine effects of the angi-
11
Service de Néphrologie Transplantation, Dialyse et Aphérèse, Centre Hospitalier ocrine factors they release. This angiocrine signalling is
Universitaire de Bordeaux, Bordeaux, France. organ-​specific and has a crucial role in tissue repair and
12
Assistance Publique-​Hôpitaux de Paris, Service d’Immunologie Biologique, Hôpital
in preventing scarring and fibrosis16.
Européen Georges Pompidou, Paris, France.
13
INSERM, UMR_S 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France.
14
Normandie Université, Université de Rouen Normandie, Rouen University Hospital, Response of renal ECs to stress
Department of Nephrology, Rouen, France. In addition to acting as a barrier, renal ECs are a source
15
Sorbonne Universités, Paris, France. and target of circulating factors. They express vasoac-
16
Université Paris Descartes, Sorbonne Paris Cité, Paris, France. tive factors (such as endothelin 1 (ET1), prostacyclin

www.nature.com/nrneph
Reviews

Leukocyte trafficking Inflammation inflammatory receptors2. Activation of ECs under sit-


• ICAM1 • MCP1 uations of stress can either be rapid and independent
• VCAM • Complement activation
• E-selectin • Oxidative stress of the transcription of new genes (a process known
as type I activation) or slower and dependent on the
Metabolism
transcription of new genes (a process known as type II
Angiogenesis
• VEGF–VEGFR • Glycolysis activation)17. For example, ECs exposed to stress stimuli
• Angiopoietin 2–TIE2 • FAO (such as cytokines, histamine, thrombin, ADP, anaphyl-
• HIF activation • Glutamine and
• CD146 asparagine metabolism atoxins of the complement system and the complement
• Glycolysis • mTOR membrane attack complex C5b-9, haem, reactive oxygen
• FAO • HIF species (ROS) and bacterial lipopolysaccharide (LPS))
• Glutamine and Endothelial
asparagine metabolism cell respond within minutes with exocytosis of intracellular
• vWF granules known as Weibel–Palade bodies, which con-
• Jagged 1–NOTCH-1 Vascular permeability
and glomerular tain components such as the prothrombotic glycoprotein
filtration vWF, P-​selectin, IL-8 and ET1. Longer-​term exposure of
Control of VSMC proliferation • Endothelial ECs to inflammatory cytokines (such as TNF, IFNγ and
• Microparticles fenestrations
• Jagged 1–NOTCH-1 • Glycocalyx IL-1), pathogen-​associated molecular patterns (PAMPs;
• miR-126 • VEGF such as LPS and Shiga toxin (Stx)) or haemolysis-​derived
• NO • CD146 products (such as haem and haemoglobin) induces sig-
nalling within ECs to increase the expression and synthe-
Haemostasis and coagulation Vascular tone sis of adhesion molecules (such as E-​selectin, VCAM1
• TM • vWF Vasodilators: Vasoconstrictors:
• Glycocalyx • TF • NO • Endothelin and ICAM1) while decreasing the production of protec-
• tPA – • TXA2 + • H2S – • TXA2 + tive molecules such as thrombomodulin18. The above-​
• TFPI • PAI1 • PGI2 • PGH2 mentioned inflammatory cytokines, as well as histamine,
• PGI2
thrombin, bradykinin, complement component C5a,
Fig. 1 | role of the endothelium in health and disease. The endothelium has a crucial vascular endothelial growth factor (VEGF) and haem,
role in the regulation of multiple processes, including angiogenesis (through the actions increase endothelial permeability and impair EC barrier
of vascular endothelial growth factor (VEGF)–VEGF receptor (VEGFR), angiopoietin function. P-​selectin released from Weibel–Palade bod-
2–TIE2 and hypoxia-​inducible factor (HIF) activation; CD146; glycolysis; fatty acid ies stimulates recruitment of leukocytes to the activated
oxidation (FAO); glutamine and asparagine metabolism; von Willebrand factor (vWF); endothelium, whereas E-​selectin promotes the rolling
and jagged 1–NOTCH1 signalling); haemostasis and coagulation (through the protective and adherence of monocytes and activated lymphocytes
role of the glycocalyx and the synthesis of procoagulant factors, such as vWF, tissue to ECs, and VCAM1 and ICAM1 facilitate the arrest of
factor (TF), thromboxane A2 (TXA2) and plasminogen activator inhibitor 1 (PAI1), and
leukocytes allowing their diapedesis. Moreover, in addi-
anticoagulant factors, such as thrombomodulin (TM), tissue plasminogen activator (tPA),
TF pathway inhibitor (TFPI) and prostaglandin I2 (PGI2)); vascular tone (through the
tion to increasing the expression of adhesion molecules,
synthesis of vasodilators such as nitric oxide (NO), hydrogen sulfide (H2S) and PGI2 and pro-​inflammatory mediators also induce shedding of
vasoconstrictors such as endothelin, TXA2 and prostaglandin H2 (PGH2) and through the the glycocalyx, resulting in increased exposure of the
control of vascular smooth muscle cell (VSMC) proliferation); vascular permeability and adhesion molecules and leukocyte recruitments.
glomerular filtration (through the functions of endothelial fenestrations, the glycocalyx, Inflammation and coagulation are closely linked
VEGF and CD146); inflammation (through the synthesis of intercellular adhesion in the endothelial response to stress. Protease-​activated
molecule 1 (ICAM1), vascular cell adhesion protein 1 (VCAM1), E-​selectin and membrane receptors 1–4 (PAR1–PAR4)19 are G protein-​coupled
cofactor protein 1 (MCP1); regulation of complement activation and oxidative stress; receptors (GPCRs) that are expressed on the surface
and the recruitment and trafficking of leukocytes); control of VSMC proliferation of ECs. They are specifically activated by coagulation
(through the functions of microparticles, jagged 1–NOTCH1 signalling, miR-126 and NO);
proteases and modulate the inflammatory and throm-
and metabolism (through the functions of glycolysis, FAO, glutamine and asparagine
metabolism as well as mTOR and HIF).
botic phenotype of ECs. For example, the activation of
PAR1 by thrombin upregulates the endothelial expres-
sion of inflammatory cytokines (such as IL-1, IL-6 and
and nitric oxide (NO)); thrombotic regulators (such as TNF) and cell adhesion molecules (such as E-​selectin,
von Willebrand factor (vWF), tissue factor (TF), plas- P-selectin, ICAM1 and VCAM1)19, promotes EC apop-
minogen activators and plasminogen activator inhibi- tosis and increases endothelial permeability19. The acti-
tor 1 (PAI1)); intercellular adhesion molecules (such vation of PAR2 by activated factor X (FXa) participates in
as platelet EC adhesion molecule (PECAM1), ICAM1, tissue remodelling, mesangial proliferation and kidney
ICAM2 and VCAM1); and inflammatory modulators, damage in animal models of ischaemia–reperfusion
including chemokines2, and thus contribute actively injury20, mesangioproliferative and crescentic glomer-
to the microenvironment. Under physiological condi- ulonephritides21,22 and renal fibrosis23. Indeed, FXa and
tions, ECs exhibit antithrombotic properties through the activation of PARs induces the expression of inflam-
activation of protein C by expressing the thrombin matory mediators, such as monocyte chemoattractant
regulator thrombomodulin, and specific proteoglycans protein 1 (MCP1; also known as CCL2), IL-1β and TNF
and by releasing tissue plasminogen activator (tPA)1. in many cell types, including glomerular macrophages,
Upon activation (for example, by inflammation, oxida- ECs and podocytes24. Thus, PARs have an important role
tive stress, injury, uraemic toxins or in the context of in the interplay between coagulation and inflammation.
infection), ECs undergo phenotypic changes, charac- The peptide vasoconstrictor ET1, which is produced
terized by the downregulation of protective regulators locally in the kidney not only by ECs but also by other
and the upregulation of pro-​adhesive molecules and cells, including podocytes, mesangial cells, parietal

Nature Reviews | Nephrology


Reviews

epithelial cells and tubular cells, acts in an autocrine or development of CKD28. As mentioned above, ET1 is
paracrine manner to regulate renal haemodynamics25. released from Weibel–Palade bodies by exocytosis
For example, binding of ET1 to the ETA receptor from ECs under conditions of endothelial stress. The
increases renal vasculature resistance and reduces renin–angiotensin–aldosterone system (RAAS), which
glomerular filtration rate (GFR), whereas binding of is involved in hypertension, renal damage and CKD pro-
ET1 to the ETB receptor increases tubular water and gression29, interacts closely with the endothelin system29.
sodium reabsorption26. Activation of the endothelin Angiotensin II (ANGII), which induces vasoconstriction
system is observed in various renal diseases and pro- of glomerular efferent arterioles, increases intraglomer-
motes mitochondrial stress in ECs, leading to disrup- ular pressure and favours the development of glomeru-
tion of the podocyte cytoskeleton27 as well as mesangial losclerosis, also increases the production of ET1 in the
cell proliferation, renal inflammation, fibrosis and the kidney, the expression of ETA and the binding of ET1
to ETA, further increasing renal vasculature resistance.
a Anti-inflammatory and anticoagulant phenotype
Key features of renal ECs
Although renal ECs behave largely like other ECs
in terms of their function as a barrier, a conduit for
EC transporting blood and a source of circulating factors,
Repair
by EPCs differences exist in the composition of their microenvi-
ronment, in their proteome and in the extent of their
Shear-stress-induced Glycocalyx Repair by response to various stimuli (Supplementary Table 1).
NO synthesis mature ECs Differences in the glycocalyx composition, such as the
abundance of sialic acid and the sulfation level of hepa-
ran sulfate30, determine the pattern of interaction of ECs
with plasma factors, such as complement factor H (FH).
This regulatory factor controls the activation of comple-
ment by recognizing and binding glycosaminoglycan on
host tissue. One glycosaminoglycan-​binding region of
FH — CCP6–CCP8 — anchors FH to Bruch's membrane
b Pro-inflammatory, procoagulant and complement-activating phenotype in the eye, whereas another, CCP19–20, anchors FH to
Activation Lesion Repair GECs30. Genetic variations in the CCP6–8 and CCP19–
20 domains therefore contribute differently to the dam-
age of eyes or kidneys in diseases such as age-related
TF Adhesion molecules macular degeneration and aHUS, respectively.
C3a Compared with cultured human umbilical vein ECs
Reduced mobilization (HUVECs), which are considered the classic model of
C5a
Coagulation
Monocyte and survival of EPCs ECs, GECs produce more fibrinolytic factors at resting
state. Moreover, the fibrinolytic properties of GECs are
potentiated upon activation with TNF or LPS, whereas
ROS Release Reduced
generation of EMPs proliferation Glycocalyx HUVECs become more pro-​thrombotic31. These differ-
breakdown ences suggest that the glomerular endothelium is par-
Detachment ticularly resistant to thrombus formation, even under
of CECs
conditions of EC stress. In contrast to differences in the
expression of thrombotic factors, only weak differences
exist between GECs and HUVECs in their expression
Reduced NO availability levels of adhesion molecules, complement factors, com-
plement activation capacity and permeability at resting
state and after activation with TNF, IFNγ, IL-1 or brief
Fig. 2 | endothelium dynamics in health and disease. a | Under physiological
conditions, the endothelium handles shear-​stress-induced synthesis of nitric oxide (NO) exposure to haem18,32–34. Nevertheless, GECs have a dif-
and maintains an anticoagulant and anti-​inflammatory environment through the ferent metabolic profile than HUVECs at resting state
production of regulatory factors and through the actions of the intact protective and after exposure to Stx and/or TNF35. Exposure of
glycocalyx. Endothelial repair is achieved by the proliferation of adjacent mature GECs to Stx and TNF strongly reduced NAD metabo-
endothelial cells (ECs) or by the incorporation of endothelial progenitor cells (EPCs), lism, leading to depletion of the energy substrate acetyl
which also promote angiogenesis through the synthesis of pro-​angiogenic factors. CoA and the antioxidant glutathione. By contrast, these
b | Situations of stress or injury lead to activation of the endothelium, characterized by effects were less pronounced in HUVECs, in which cellu­
the release of reactive oxygen species (ROS) and endothelial microparticles (EMPs) and lar acetyl CoA content even increased, which may partly
activation of complement and other pro-​inflammatory mediators. Disruption of the explain the particular susceptibility of GECs to Stx-
glycocalyx exposes adhesion molecules on the endothelium surface, leading to
induced injury35. These findings are in line with a study
leukocyte recruitment and inflammation; exposure of tissue factor (TF) induces
activation of the coagulation cascade. A reduction in NO release as a consequence of showing that oxidative stress, membrane lipid peroxi-
endothelial dysfunction induces impairment of flow-​mediated vasodilation. Apoptotic dation and depletion of reduced glutathione contrib-
or damaged ECs become detached and form circulating ECs (CECs), exposing the uted to renal injury in a mouse model of Stx-​associated
extracellular matrix with its procoagulant TF to components of the coagulation cascade. HUS36. These findings suggest that Stx and TNF render
Endothelium repair by adjacent mature cells or by EPCs is also compromised. GECs particularly susceptible to oxidative stress and lead

www.nature.com/nrneph
Reviews

Endothelium in medium and large vessels Podocyte

Endothelial
cell
Endothelial cell Glycocalyx
Mesangial
cell
Smooth Glomerular
muscle cell endothelium

Peritubular capillary

Endothelial Tubular
cell epithelial
cell

Fig. 3 | Diverse populations of endothelial cells in the kidney. The kidney harbours three main populations of
endothelial cells with distinct architectures and functions. Endothelial cells within medium and large vessels are
continuous, confluent, connected by intercellular junctions and elongated in the direction of blood flow. Glomerular
endothelial cells are highly fenestrated and are covered on their luminal surface by a thick glycocalyx, which participates in
the sieving properties of the glomerular filtration barrier. Crosstalk between glomerular endothelial cells, mesangial cells
and podocytes is crucial for the maintenance of glomerular structure and function. Similar to glomerular endothelial
cells, endothelial cells of the peritubular capillaries are also fenestrated, and their fenestrae are covered by a thin
diaphragm composed of glycoproteins. This endothelial population facilitates the reabsorption and secretion of fluids
and molecules from the adjacent tubular epithelial cells.

us to speculate that pro-​oxidative haemolysis-​derived context of tissue ischaemia45, complement activation


products, such as haemoglobin, haem and erythrocyte and inflammatory cytokine stimulation).
microvesicles37,38, are particularly noxious for kidney Renal EndMT is a specific feature of endothelial
microvessels and will amplify the cell damage, comple- dysfunction and can contribute to renal microvas-
ment activation and thrombosis in the context of renal cular rarefaction 45, fibrosis and the progression of
thrombotic microangiopathy (TMA)2,39,40. CKD46. This process has been investigated in experi-
mental models of renal injury, including ischaemia–
Endothelial-​to-mesenchymal transition reperfusion injury, unilateral ureteral obstruction
ECs exhibit remarkable plasticity during both embry- (UUO)46 and streptozotocin-​induced DKD42. Use of
ogenesis and in adulthood 41. Maintenance of the endothelial-​lineage tracing demonstrated the presence
endothelial phenotype is a physiologically active process, of αSMA-​positive myofibroblasts of endothelial ori-
requiring energy and signalling input in ECs41. Some gin in the interstitium of fibrotic kidneys from mice
forms of endothelial injury (for example, in the setting with streptozotocin-​induced DKD42, suggesting that
of AKI resulting from sepsis or ischaemia–reperfusion EndMT contributes to renal fibrosis. A small number of
injury) induce ECs to undergo EndMT, leading to vascu- myofibroblasts of endothelial origin were also observed
lar rarefaction, organ fibrosis and dysfunction41. EndMT in the afferent and efferent glomerular arterioles of
involves a partial or complete phenotypic switch of ECs these mice, suggesting that EndMT may participate
to a mesenchymal cell fate. In this process, which can in glomerulosclerosis.
be driven by activation of TGFβ, AKT and/or mTOR As mentioned above, excessive TGFβ signalling
pathways42, ECs lose markers typical of the endothelial probably contributes to the activation of EndMT.
lineage (for example, VE-​cadherin and CD31), their TGFβ-knockout mice are non-viable; however, mice with
polarization and primary cilia43 and acquire myofi- heterozygous endothelium-specific deletion of the
broblastic characteristics, including the expression TGFβ receptor are viable and display enhanced angio-
of α-smooth muscle actin (αSMA) and N-cadherin41. genic potential under basal conditions47. These mice are
EndMT has a central role in cardiac development also protected from tubulo-​interstitial fibrosis and renal
during embryogenesis44. After birth, EndMT occurs microvasculature rarefaction, with preserved renal blood
in several pathologic conditions (for example, in the flow and low levels of tissue hypoxia after induction of

Nature Reviews | Nephrology


Reviews

folic-​acid-associated nephropathy or UUO47, implicating membrane surface, thereby facilitating thrombus forma-
a role for TGFβ signalling and EndMT in the regulation tion53. EMPs derived from patients with CKD have higher
of angiogenic and fibrotic responses to kidney injury. levels of exposed phosphatidylserine than those obtained
In humans, EndMT contributes to renal damage from control individuals63, and exposure of cultured ECs
in sepsis-​induced AKI48,49. In kidney transplant recip- to uraemic toxins promotes the release of EMPs with
ients, EndMT can result from ischaemia–reperfusion TF-​dependent procoagulant activity64. High numbers
injury and from antibody-​mediated rejection (ABMR). of EMPs are also observed in the blood of patients with
Markers of EndMT are expressed in peritubular cap- antiphospholipid antibodies (aPLs)65, but only plasma
illaries of patients with ABMR50 and are predictive of from patients with a history of thrombosis stimulates
long-term graft dysfunction and proteinuria. cultured HUVECs to release procoagulant EMPs66, high-
lighting the fact that both the number and the properties
The circulating endothelial compartment of EMPs are of interest as biomarkers in this setting.
The circulating endothelial compartment consists of cir- EMP activation is probably closely linked to comple-
culating ECs (CECs), endothelial-​derived microvesicles ment activation. The deposition of complement on cell
or microparticles (EMPs) and endothelial progenitor surfaces and the formation of the membrane attack com-
cells (EPCs). The balance between endothelial injury, plex C5b−9 can induce the release of EMPs67. Moreover,
as reflected by the presence of CECs and EMPs, and treatment of mice with ciclosporin induces activation
the capacity for endothelial repair, as reflected by the of the alternative pathway of complement within the
recruitment of functional EPCs, is crucial for maintain- kidneys, leading to glomerular deposition of C3 (ref.68)
ing endothelial homeostasis51. Although it is currently and the release of EMPs carrying C3 deposited on their
not possible to determine the organ of origin of CECs surface. These C3-carrying EMPs interact poorly with
and EMPs, these cells and cell-​derived packages are the regulatory FH, resulting in further activation of the
of interest as potential biomarkers of acute or chronic alternative pathway68. This process might amplify intra-
endothelial dysfunction in renal diseases (Table 1). CECs renal endothelial damage and favour the development
are mature ECs that have been shed from the injured of TMA. The involvement of EMPs in this amplifying
vessel wall (for example, after myocardial infarction loop of endothelial injury might be of interest in the
or arterial angioplasty)52. EMPs are submicrometric context of haemolytic uraemic syndrome (HUS) and in
vesicles that are shed from stressed or damaged ECs53 organ transplantation.
upon remodelling of the plasma membrane, with exter- Endothelial lesions can also result from a defect in
nalization of phosphatidylserine53. Numbers of CECs repair capacity51. EPCs were first described in 1997
and EMPs, which are most commonly measured in the (ref.69) as a population of cells from the haematopoietic
blood by CD146-based immunomagnetic separation54 lineage that incorporated into sites of angiogenesis. They
and flow cytometry53, are increased in diseases involv- were subsequently shown to originate either from the
ing EC activation and/or injury52,53 and typically corre- bone marrow or from tissue-​specific resident progenitor
late with disease severity and clinical outcome52. Patients cells16 and to promote angiogenesis and microvascular
with systemic lupus erythematosus (SLE) have increased endothelial repair through the production of angiocrine
numbers of CECs, especially those with lupus nephri- factors such as VEGFA, FGF2 and angiopoietins16,51.
tis and renal TMA, reflecting microvascular injury55. Studies in animal models of endothelial kidney injury,
Numbers of CECs are also increased in patients on hae- such as experimental glomerulonephritis or CKD
modialysis, especially those with active cardiovascular resulting from 5/6 nephrectomy, indicate that endothe-
disease4 in whom it predicts the occurrence of future lial regeneration occurs predominantly via EPCs of
cardiovascular events56, suggesting that the number of renal origin, with ECs of extrarenal origin contributing
CECs reflects endothelial damage and is a biomarker 0–10% of ECs following regeneration70–72. Intravenous
of cardiovascular complications. Levels of EMPs are also infusion of autologous cultured EPCs obtained from
elevated in patients with CKD57,58 and correlate with bone marrow to mice following 5/6 nephrectomy atten-
clinical markers of vascular damage58. uated damage to the remaining renal tissue, probably
Flow-​mediated dilation
Noninvasive, ultrasonography-​ Circulating EMPs and CECs are not only markers through the release of paracrine factors such as VEGF73.
based test of endothelial of vascular damage but also mediate endothelial dys- Homing of these EPCs to glomerular and peritubular
function that measures function59. Apoptotic and/or necrotic CECs can interact capillaries, as well as to liver and spleen, was observed
brachial arterial diameter in with healthy endothelium and induce pro-​inflammatory after 4 weeks73. In larger vessels, resident EPCs from the
response to a substantial
increase in arterial flow after
signals60. Both apoptotic and necrotic CECs are bound vessel wall are most likely the main drivers of vascular
the release of brachial and engulfed by healthy ECs, and incubation of cultured repair, as demonstrated by the minimal contribution of
constriction with a cuff. Flow-​ ECs with apoptotic CECs results in increased expression bone-​marrow-derived EPCs to endothelial replacement
mediated dilation reflects the of chemokines and increased binding of leukocytes to in a model of transplant atherosclerosis13,74.
ability of endothelial cells to
ECs60. In patients with systemic diseases, such as vasculitis
secrete nitric oxide (NO) and
induce arterial vasodilation in or SLE, EMPs can contribute to a pro-​inflammatory envi- Evaluating EC dysfunction and damage
response to shear stress. ronment by transporting inflammatory mediators from Several approaches are used to evaluate endothelial func-
Alterations in flow-​mediated the site of injury into the circulation61. In patients with tion in the clinic (Table 1). These include functional tests
dilation represent an early antineutrophil cytoplasmic antibody (ANCA)-associated (mainly measurement of flow-​mediated dilation); evalu-
marker of endothelial
dysfunction and can be
vasculitides (AAV), numbers of EMPs in plasma corre- ation of markers indicative of a pro-​inflammatory or
reversed with endothelial late with disease activity62. EMPs express procoagula- prothrombotic endothelium phenotype; dysregulation
recovery. tion factors, such as phosphatidylserine and TF, on their of NO metabolism or disruption of the glycocalyx4,75;

www.nature.com/nrneph
Reviews

Table 1 | Noninvasive approaches to evaluate endothelial function in humans


Type of exploration Markers Consequence of endothelial dysfunction
Functional tests
FMD NA Reduced
FMD with administration of an NO NA Unaltered
donor
Forearm blood flow NA Decreased
Cutaneous circulation NA Decreased
Plasmatic markers
Adhesion molecules sVCAM1 Increased
sICAM1 Increased
Soluble E-​selectin Increased
Pro-​inflammatory markers Inflammatory cytokines Increased
Oxidized LDL Increased
Endocan Increased
Thrombotic mediators Thrombomodulin Increased
von Willebrand factor Increased
Tissue factor Increased
Markers of NO dysregulation NO metabolites Modified
Asymmetric dimethylarginine Increased
Symmetric dimethylarginine Increased
Glycocalyx disruption Heparan sulfate Increased
Circulating endothelial compartment
Markers of activation or lesion Circulating endothelial cells Increased
Endothelial microparticles Increased
Markers of repair capacity Endothelial progenitor cells Decreased numbers in the blood and
altered functions in vitro
Circulating angiogenic cells Decreased numbers in the blood and
altered functions in vitro
FMD, flow-​mediated vasodilation; NA , not applicable; NO, nitric oxide; sICAM1, soluble intercellular adhesion molecule; sVCAM1,
soluble vascular cell adhesion protein 1.

and evaluation of circulating endothelial compartments, with endocapillary and/or extracapillary proliferation
indicative of an imbalance between endothelial damage of glomerular cells, with or without immune complex
and repair76. All of the available markers that are used deposition. Peritubular capillary rarefaction, vascular
to evaluate endothelial function in humans reflect sys- fibrosis and interstitial fibrosis (resulting in part from
temic endothelial dysfunction rather than organ-​specific EndMT) can also be seen in renal biopsy samples.
Tubulo-​reticular inclusions endothelial dysfunction. Ultrastructurally, endothelial damage can be observed
Cytoplasmic clusters of
Histological examination of renal biopsy samples is in glomeruli, peritubular capillaries and the vasa
tubule-like structures arising
from the membrane of the required to assess endothelial damage specifically in the recta, evidenced by reduced fenestrations, increased
endoplasmic reticulum, kidney and typically demonstrates capillary or small-​ endothelial vacuoles, thickened basement membranes
thought to result from vessel thrombosis with fibrin deposition. Renal TMA and fibrin-​like intraluminal deposits78,79. In virus-​
activation of type I interferons is also evidenced by the finding of swollen and turgid associated nephropathies and lupus nephritis, typical
in endothelial cells and
ECs, obstructing blood flow in the glomerular capil- tubuloreticular inclusions can be observed in GECs80.
lymphocytes.
lary or small arteries and inducing their detachment Intravital multiphoton microscopy (IVM) has also been
Intravital multiphoton from the GBM or vascular wall, with the formation of used81–83 to study renal blood flow, leukocyte rolling and
microscopy ‘double contours’ (whereby the GBM seems duplicated recruitment, cell death and cell shedding and vascular
(IVM). A form of microscopy
on light microscopy, reflecting the synthesis of GBM-​ permeability in the glomerular and peritubular vascula-
that enables the observation of
biological processes in living like material, which is possibly initiated to fill a void ture of the living kidney in rodents. IVM enables inves-
animals (owing to its low between non-​adherent GECs and the GBM) and clear tigation of endothelial and microvascular alterations
energy and phototoxicity, subendothelial spaces (that is, empty spaces between the in the living animal, showing, for example, the reduced
which allow the prolonged GECs and GBM on light microscopy, reflecting a par- peritubular capillary blood flow in sepsis-​induced AKI
exposure of tissues) with high
resolution owing to the deep
tial detachment of GECs from the GBM)77. Glomerular or after ischaemia–reperfusion injury or increased vas-
penetration of tissues by fibrinoid necrosis is also encountered in biopsy samples cular permeability of ischaemic areas of renal tissue.
photons. from patients with renal vasculitides or lupus nephritis, At the cellular level, IVM has been used to observe how

Nature Reviews | Nephrology


Reviews

alterations in the endothelial glycocalyx might favour Typical Shiga toxin-​associated haemolytic uraemic
albuminuria and increase vascular permeability in syndrome. TMA is characterized by vascular damage,
living rats84. with the formation of fibrin and platelet thrombi90.
TMA translates clinically into a triad of peripheral
Future strategies to study EC populations thrombocytopenia, mechanical haemolytic anaemia
Our understanding of EC diversity among different vas- and acute or chronic organ damage predominantly in
cular beds is limited. Microarray studies of cultured ECs kidney and brain. The most common type of kidney-​
have demonstrated differences in the gene expression predominant TMA is typical HUS caused by an infec-
profiles of ECs isolated from different vascular beds8. tion with Stx-​secreting bacteria, including some strains
However, the use of classic 2D culture conditions inevi- of Escherichia coli (Fig. 4a). Animal studies of typical
tably modifies the behaviour and gene expression profile HUS have been hampered by the finding that conven-
of ECs, including renal ECs, limiting the utility of this tional mice infected with enterohemorrhagic E. coli do
system. The construction of 3D renal vessels to enable not develop key features of the disease, including renal
exposure of ECs to physiological mechanical stresses and lesions. One study showed that induction of the full
surrounding cells, such as pericytes and podocytes, is a renal TMA phenotype required co-​injection of Stx and
key goal for the study of the endothelium in renal dis- LPS into mice91. Another study engineered Citrobacter
eases15. Although progress has been made in the develop- rodentium to secrete Stx, which reproduced the human
ment of vascular scaffolds85–87, further studies are needed phenotype in mice92. Mechanistic studies have shown
to more closely mimic the in vivo environment88. Of note, that Stx causes glomerular injury through binding of
engraftment of renal organoids below the kidney capsule its B-​subunit of globotriaosylceramide (GB3)93. This
of living rats89 resulted in the formation of vascularized receptor is expressed at particularly high levels by
glomeruli with differentiated capillary walls and basic GECs (~50-fold higher than in HUVECs), explaining
physiologic functions89, providing a model with which the susceptibility of the glomerular endothelium to this
to study renal EC function. The development of ex vivo toxin94. Once bound to its receptor, the Stx undergoes
perfused kidneys-​on-a-​chip that recapitulate compo- endocytosis and inhibits protein synthesis within the
nents of the glomerular barrier, such as GECs, the GBM EC by cleaving an adenine base from the 28S ribosomal
and podocytes, and enable glomerular fluid shear stress RNA (rRNA) of the 60S ribosomal subunit95. Moreover,
conditions would also advance studies of EC function. Stx modulates microvascular EC signalling pathways
and ribosomal activity, inducing endothelial permea-
Diseases with endothelial damage bility and cytotoxic changes in ECs by stimulating the
As mentioned earlier, the renal endothelium can be CXCR4–CXCR7–SDF1 pathway96. Following infection,
activated or injured by a variety of factors, including Stx is also transported in the bloodstream bound to
toxins, hyperglycaemia, complement activation frag- platelets, neutrophils, monocytes and red blood cells;
ments, autoantibodies or alloantibodies, immune cells these cells, together with ECs, release Stx-​containing
and cytokines. Defects in mechanisms of endothelium complement-​coated microvesicles that enable GB3-
protection (for example, through dysregulation of com- independent endocytosis of Stx into GECs and peritu-
plement regulatory factors such as FH as occurs in aHUS bular capillary ECs and through basement membranes
or through an imbalance between pro-​angiogenic and into podocytes and tubular cells67. In GECs, Stx induces
anti-​angiogenic factors as occurs in pre-​eclampsia) can activation of complement, mobilization of the Weibel–
also induce glomerular endothelial damage. Injury of Palade bodies and cell surface expression of the adhe-
peritubular capillary ECs in settings such as ischaemia– sion molecule P-​selectin97, which provides a platform
reperfusion can also induce AKI and vascular rarefac- for assembly of the alternative pathway C3 convertase
tion, which can lead to renal fibrosis and subsequent (C3bBb) and further cleavage of C3 into C3a and C3b97.
CKD in part owing to EndMT. Some of these processes The release of C3a further potentiates the expression of
are specific for particular kidney diseases (such as aHUS) P-​selectin and stimulates tPA-​dependent thrombomod-
whereas others contribute more broadly to the mecha- ulin shedding and loss of thromboresistance, leading to
nisms of renal dysfunction seen in common complex a prothrombotic state and thrombus formation. Stx also
diseases, such as hypertension-​attributed nephropathy binds to the ultra-​large multimers of the glycoprotein
and DKD. Irrespective of its aetiology, CKD is associ- vWF, reducing their cleavage by ADAMTS13 and fur-
ated with an accumulation of uraemic toxins, which ther stimulating thrombosis98. Acceleration of the TMA
contribute to systemic endothelial dysfunction and car- process through these pathways eventually leads to AKI.
diovascular disease. The below discussion describes the
mechanisms of renal endothelial damage in renal dis- Lupus nephritis and APS nephropathy. The pathogene-
eases and explains how CKD can, in turn, exacerbate sis of SLE is multifactorial, involving a loss of tolerance
systemic endothelial dysfunction. to self-​antigens and the production of a wide range of
autoantibodies, with formation of immune complexes
Direct endothelial injury or activation and culminating in the damage of tissues, such as the
The most emblematic renal diseases resulting from kidney in lupus nephritis99. Autoantibodies that react
direct endothelial injury are typical HUS, lupus nephri- specifically with the EC surface (anti-​EC antibodies
tis and antiphospholipid syndrome (APS) nephrop- (AECAs)) have been found in patients with SLE100,101,
athy, ABMR in renal transplant recipients, AAV and in whom their presence is associated with markers of
sepsis-induced AKI. endothelial damage102, and with the occurrence and

www.nature.com/nrneph
Reviews

pathological activity of lupus nephritis101,103,104. The TMA lesions are reduced in mice lacking the C5a
specific antigenic targets of these AECAs are yet to be receptor (C5aR)119, highlighting the role of complement
identified; however, their binding to native or modified activation in APS and APS nephropathy. Moreover, the
endothelial antigens might promote a pro-​inflammatory monoclonal anti-​C5 antibody eculizumab has been
and procoagulant EC phenotype, stimulating oxidative successfully used in patients with post-​transplantation
stress and EC apoptosis100 and promoting the develop- aPL-​induced TMA or refractory catastrophic APS110,120.
ment of lupus nephritis, renal vasculopathy and accel- Activation of the proliferation and survival AKT–mTOR
erated systemic atherosclerosis105 (Fig. 4b). Apoptosis pathway by aPLs is also involved in APS nephropa-
of cultured HUVECs is increased following exposure thy, especially in the chronic vasculopathy character-
to purified immunoglobulin G (IgG) from patients ized by EC proliferation and intimal hyperplasia110,115.
with lupus nephritis101. Moreover, anti-​complement C3 Heterogeneous vascular lesions involving endothelial
autoantibodies from patients with lupus nephritis stim- activation or damage are therefore involved in both the
ulated complement activation in cultured HUVECs, systemic and renal manifestations of SLE and APS.
contributing indirectly to endothelial injury106.
SLE is also associated with an imbalance between Antibody-​mediated rejection in kidney transplantation.
endothelial lesion and repair. Patients with SLE — The renal endothelium is particularly prone to injury in
particularly patients with lupus nephritis and renal TMA kidney transplant recipients as ECs from the transplant,
— have increased numbers of CECs55 and decreased which bear the HLA antigens of the donor, are directly
numbers and functions of EPCs and circulating angio- exposed to the recipient’s immune system. The main
genic cells (CACs)107, which are susceptible to apoptosis, cause of graft loss in kidney transplant recipients is
synthesize few pro-​angiogenic molecules, incorporate ABMR121, defined by the presence of donor-​specific
poorly into vascular structures and differentiate poorly antibodies (DSAs) directed against donor HLA alloan-
into mature ECs107. The functional defects in EPCs tigens, microvascular inflammation (glomerulitis and
and CACs in patients with SLE are mediated by type I peritubular capillaritis) and variable C4d deposition
interferons107, which repress the pro-​angiogenic factors in peritubular capillaries121. Of kidney transplant recipi-
IL-1β and VEGF and activate the inflammasome, and ents, 24% develop de novo DSAs within 10 years of trans-
the pro-inflammatory cytokine IL-18 (ref.108). plantation, increasing the risk of impaired long-​term
In association with SLE or in primary APS, aPLs graft survival122,123, whereas preformed DSAs are associ-
can also contribute to renal endothelial damage. APS ated with a high risk of ABMR and early graft failure124.
is defined as the coexistence of thrombotic or obstetric DSAs cause endothelial damage through direct activation
events in the presence of persisting specific aPLs. The of ECs, through activation of the complement system and
renal manifestations of APS are heterogeneous109, com- through recruitment of inflammatory cells such as natu-
prising both large renal vessel manifestations (throm- ral killer (NK) cells via their crystallizable fragment (Fc)
bosis, stenosis or infarction) and microvascular lesions receptors, inducing antibody-​dependent cell cytotoxicity
(TMA or fibrous intimal hyperplasia), which can induce (ADCC)125 (Fig. 4c). In vitro studies show that anti-​HLA
progressive or acute renal dysfunction — a condition antibodies bind to ECs within the transplant, leading to
known as APS nephropathy110. The diversity of renal activation of the mTOR pathway, EC proliferation and
manifestations implies that aPLs exert various, poten- intimal hyperplasia with subsequent ischaemia126,127.
tially overlapping effects on the vasculature, including The Fc regions of IgM and IgG DSAs bind C1q, acti-
promotion of coagulation or impairment of fibrinolysis vating the classical complement pathway and resulting
by binding proteins of the coagulation pathway; direct in endothelial injury. Moreover, ischaemia–reperfusion
cytotoxic effects on EC membrane phospholipids; and injury that can occur with kidney transplantation pro-
the recruitment of pro-​inflammatory, pro-​oxidative motes local activation of complement via lectin and alter-
and prothrombotic factors through increased inter- native pathways, independent of DSAs128,129. Deposition
actions of the circulating β2-glycoprotein 1b (β2GPI) of sublytic levels of C5b-9 causes EC activation, with
with different receptors, such as Toll-​like receptor 2 recruitment and activation of T cells contributing to
(TLR2), TLR4, annexin A2 and the α-​chain of platelet transplant vasculopathy 130, although complement-​
glycoprotein Ib (GPIbα) on ECs or blood cells111 (Fig. 4b). induced lysis of ECs is hampered by the high consti-
Most studies of the endothelial effects of aPLs have tutive expression of protective complement regulators
used either monoclonal murine antibodies, human such as membrane cofactor protein (MCP; also known as
affinity-​purified antibodies (mostly anti-​glycoprotein 1 CD46), CD55 (also known as decay accelerating factor,
(GP1) antibodies) or patient-​derived IgG antibodies112. DAF) or CD59. Inflammation triggered by the anaphyl-
Sequence homology between human and mouse atoxins C3a and C5a can also contribute to ABMR131.
GP1 enables use of patient-​derived aPLs in murine In allograft biopsy samples, the presence of glomerulitis
models113,114. Injection of human aPLs into mice does and peritubular capillaritis indicates ongoing interaction
not induce direct thrombosis in the absence of under- of antibodies with the endothelium130.
lying endothelial dysfunction and/or precipitating Complement-​independent pathways, in particu-
thrombotic events, but it does induce a dose-​dependent lar the NK-​cell-mediated ADCC, are also involved in
increase in thrombus size once thrombosis is initiated, ABMR132. NK cells interact with antibodies via their
which is indicative of a ‘two-​hit’ concept115,116. Thrombus FcγR and can induce rapid apoptosis or lysis of target
formation is decreased in mice deficient in complement ECs through the release of perforin and granzyme and
components C3 (ref.117) or C5 (ref.118), and glomerular the production of IFNγ and other pro-​inflammatory

Nature Reviews | Nephrology


Reviews

a Typical haemolytic uraemic syndrome C3a


Platelet
aggregation
Stx-containing Soluble TM
microvesicles C3b
TM
Stx C3
convertase Factor B
vWF
tPA

GB3 P-selectin C3aR

Endocytic
vesicle GEC
• Inhibition of
protein synthesis Weibel–Palade body
• Cytotoxic changes

b Lupus nephritis APS nephropathy


Podocyte EC Direct Complement
β2GPI activation
membrane
toxicity

Cellular
Stabilization receptor

EC activation
EC activation
Recruitment of and proliferation
inflammatory cells
Immune
complexes Anti-C3b Antiphospholipid
EPC and CAC
dysfunction antibody antibody
Subepithelial Subendothelial Endothelial Anti-endothelial
deposits deposits antigen antibody

c Antibody-mediated rejection
Classical complement C3a
pathway activation C5a
NK
cell
C1q C3 convertase Thrombus formation
DSA
Donor HLA C4d

NK cytotoxic Membrane attack


enzymes complex (C5b-9)
ADCC Complement-dependent lesion

d ANCA-associated vasculitis
MPO or PR3

Priming ANCA
antibody ROS

Neutrophil
Neutrophil
serine Leukocyte
proteases Integrin recruitment
Complement
alternative
NET Adhesion
pathway activation
molecule

NETosis
EC activation
EC lysis

Tissue damage and Neutrophil


fibrinoid necrosis diapedesis

www.nature.com/nrneph
Reviews

◀ Fig. 4 | renal diseases associated with endothelial injury in kidney diseases. ischaemia–reperfusion injury, ECs increase their expres-
a | In typical haemolytic uraemic syndrome, Shiga toxin (Stx) enters glomerular endothelial sion of HLA-​DR, which facilitates the presentation of
cells (GECs) by binding the B-​subunit of globotriaosylceramide (GB3), which induces antigens to T cells and is itself an important target for
Stx endocytosis, leading to inhibition of protein synthesis and cytotoxic damage. The allogeneic T cells139,140. Under inflammatory conditions,
release of Stx-​containing complement-​coated microvesicles enables entry of Stx into
ECs of the graft also modify the local allogeneic response,
other cell populations. Stx induces complement activation, mobilization of Weibel–Palade
bodies, P-​selectin expression and secretion of von Willebrand factor (vWF) multimers, promoting either an IL-6-dependent T helper 17 (TH17)
stimulating platelet aggregation; P-​selectin stimulates C3 convertase activity and tissue cell pro-​inflammatory response or a contact-​CD54-
plasminogen activator (tPA)-dependent shedding of thrombomodulin to generate dependent regulatory T cell response, depending on the
soluble thrombomodulin (TM), further promoting thrombosis. Released C3a will bind to cytokine environment, thus contributing to or regulating
its receptor, C3aR, and will further stimulate the GECs to mobilize the Weibel–Palade the rejection process141.
bodies, perpetuating the process. b | Lupus nephritis is characterized by the glomerular
deposition of circulating immune complexes. GECs can also be activated by ANCA-​ a ssociated vasculitides. Renal involvement
anti-endothelial antibodies; moreover, anti-​complement C3 autoantibodies stimulate in AAV — a group of small-​vessel vasculitides with a
complement activation and recruitment of inflammatory cells, exacerbating glomerular broad spectrum of clinical manifestations — is char-
damage. Lupus nephritis is also characterized by an imbalance between endothelial
acterized by a necrotizing crescentic glomerulone-
damage and repair, with decreased numbers and functions of circulating angiogenic
cells (CACs) and endothelial progenitor cells (EPCs). Antiphospholipid antibodies phritis with absent or very few immune deposits142.
associated with systemic lupus erythematosus or in primary antiphospholipid syndrome ANCA autoantibodies and neutrophils contribute to
(APS) have direct cytotoxic effects on endothelial cell (EC) membrane phospholipids; they the injury of ECs and small-​vessel walls by stimulating
can bind and stabilize β2-glycoprotein 1b (β2GPI) and induce complement activation; oxidative burst, the release of proteolytic enzymes and
their interaction with cellular receptors (for example, Toll-​like receptor 2 (TLR2), TLR4, the formation of neutrophil extracellular traps (NETs)143
annexin A2 and α-​chain of platelet glycoprotein Ib (GPIbα)) can induce EC activation and (Fig. 4d). Activation of neutrophils by ANCAs requires
proliferation. c | In kidney transplant recipients with antibody-​mediated rejection, cell priming, which is mediated by pro-​inflammatory
donor-specific antibodies (DSAs) directed against donor HLAs can directly activate ECs. cytokines, and induces migration of the enzymes pro-
DSAs can also trigger complement activation by binding C1q, activating the classical teinase 3 (PR3) or myeloperoxidase (MPO), which are
complement pathway and inducing thrombus formation. Inflammatory cells, such as
contained in neutrophil azurophilic granules142, to the
natural killer (NK) cells, can be recruited via their crystallizable fragment (Fc) receptor,
leading to the generation of NK cytotoxic enzymes and antibody-​dependent cell neutrophil membrane144,145. Binding of ANCAs to MPO
cytotoxicity (ADCC). d | In antineutrophil cytoplasmic antibody (ANCA)-associated or PR3 on the neutrophil membrane then promotes
vasculitis, activation of neutrophils by ANCAs follows neutrophil cell priming and the activation of neutrophil β2 integrin, which mediates
migration of proteinase 3 (PR3) or myeloperoxidase (MPO) to the cell surface. Neutrophil adhesion of leukocytes to the endothelium and favours
activation induces the formation of neutrophil extracellular traps (NETs), and stimulates microvascular permeability and neutrophil diapedesis.
oxidative burst, with the production of reactive oxygen species (ROS), which together In vitro, ANCA-​stimulated neutrophils activate ECs in
cause EC damage and fibrinoid necrosis. Binding of neutrophils to adhesion molecules a process mediated by NF-κB, which favours neutrophil
expressed by activated ECs promotes neutrophil recruitment and diapedesis, adhesion. Use of targeted small interfering RNA (siRNA)
contributing further to tissue damage. to block NF-​κB specifically in the endothelium attenu-
ated the development of crescentic glomerulonephritis
chemokines that contribute to recruitment of cells of in a mouse model of AAV, demonstrating an important
the adaptive immune system and EC activation133. High role for EC in the induction of AAV-​associated glo-
levels of transcripts corresponding to NK cells were merulonephritis146. ANCAs also induce the release of
found in kidney and heart allograft biopsy samples from enzymes contained in neutrophil toxic granules, such as
patients with DSAs and ABMR134,135, and the contribu- neutrophil serine proteases147, which induce EC injury
tion of NK cells to chronic allograft vasculopathy was and small-​vessel fibrinoid necrosis148. In addition, neu-
confirmed in a murine model of heart transplantation132. trophil activation induces the release of ROS, mediated
In ex vivo samples from kidney transplant recipients, by NADPH oxidase 1 (NOX2). Surprisingly, one study
ADCC reactivity of NK cells against allogeneic target showed that transplantation with NOX2-deficient bone
cells was predictive of a decline in transplant function136. marrow induced a more severe phenotype in a mouse
Moreover, in vitro cytotoxicity of patient-​derived DSAs model of AAV than transplantation with wild-​type bone
was predictive of ABMR136. marrow149. The more severe phenotype was associated
Oxidative burst In addition to DSAs, non-​HLA preformed autoan- with increased renal concentrations of IL-1β, suggest-
Also known as respiratory tibodies directed against cryptic epitopes exposed after ing that NOX2 interferes with inflammasome-​induced
burst, oxidative burst is the tissue damage (such as ischaemia–reperfusion injury, IL-1β processing and ANCA-​induced inflammation149.
rapid release of reactive
oxygen species (ROS) from
vascular injury and/or allograft rejection) might par- Activated neutrophils also exert pathogenic effects
phagocytes (such as ticipate in renal transplant dysfunction137. For example, through the excessive generation of NETs143. ANCA-​
neutrophils or macrophages) to LG3, a fragment of the proteoglycan perlecan normally induced NETs containing MPO and PR3 have been
degrade internalized particles present in the vasculature, is a neoantigen that is released found in the kidneys of patients with AAV-​induced glo-
or pathogens within the
by injured or apoptotic ECs. Patients with early vascular merulonephritis and might induce EC death through
phagosome. Primed
neutrophils can also rejection have high levels of anti-​LG3 antibodies, and the histone-dependent cytotoxicity — a mechanism that is
degranulate and undergo an pre-​existence of anti-​LG3 antibodies in patients awaiting thought to trigger adaptive immunity by exposing extra-
oxidative burst in the absence kidney transplantation is associated with an increased cellular autoantigens to antigen-​presenting cells146,150. The
of pathogens, as in risk of vascular rejection137. alternative pathway of complement also contributes to
antineutrophil cytoplasmic
antibody (ANCA)-associated
Finally, ECs from the transplant not only are a passive AAV pathogenesis. In a mouse model of anti-​MPO vas-
vasculitis, leading to target of alloimmunization but also participate in this culitis, deficiency of C5 or factor B protected mice from
surrounding tissue damage. process138. Upon exposure to hypoxia in the context of disease151. Activation of complement and the generation

Nature Reviews | Nephrology


Reviews

Neutrophil extracellular of C5a might result from interaction with infectious trig- damage2,90,154. Genetic abnormalities or the presence
traps gers such as bacterial infections, neutrophil membranes, of autoantibodies that affect the quantity or function of
(NETs). NETs are composed of plasma circulating microparticles from different cell types FH are the most common causes of alternative pathway
DNA in association with or NETs, inducing a pro-​inflammatory amplification dysregulation156–158, followed by mutations in MCP159
histones and granular proteins
such as neutrophil elastase and
loop involving NETs and the complement cascade152. and factor I160. Overactivation of the alternative path-
myeloperoxidase (MPO). They way due to mutations in the C3 convertase components
are released by neutrophils in a Sepsis-​induced acute kidney injury. Sepsis is caused by C3 (refs161,162) and factor B163,164 are also associated with
process called NETosis, which an overwhelming host response to infection and results aHUS (Fig. 5a). Genetic abnormalities independent of
allows pathogens to be
in life-​threatening dysfunction of organs, including the complement have also been described, notably in dia-
captured in a bactericidal net
and destroyed. NETs can also
kidneys. AKI develops in approximately 50% of patients cylglycerol kinase-​ε (DGKE)165 — a gene involved in
cause tissue damage, such as with sepsis who are admitted to the intensive care unit the protein kinase C (PKC) signalling pathway — or in the
in ANCA-​associated vasculitis, (ICU) and is associated with 30–50% mortality in this gene that encodes thrombomodulin166. DGKE deficiency
or increase the exposure of setting153. Systemic hypotension and ischaemia–reper- induces a permanent procoagulant phenotype in ECs167,
autoantigens to the immune
system, such as in lupus.
fusion injury are not the only culprits in sepsis-​induced whereas decreased expression of thrombomodulin pro-
AKI, with studies over the past decade highlighting a motes thrombi formation and inhibits the inactivation
Cell priming central role of endothelial dysfunction153. ECs are both of C3a, C5a and C3b166.
Cell priming of neutrophils is targets and facilitators of kidney damage in sepsis1. ECs Despite differences in the complement system
the transition from a steady
are exposed to bacterial components (such as LPS), pro-​ between mice and humans, mouse models of aHUS
state with limited antimicrobial
activity to an activated state,
inflammatory cytokines (including TNF and IL-1) and have been developed168. Mice with complete deficiency
allowing the rapid enhancement immune cells, promoting their activation or apopto- in FH develop spontaneous C3 glomerulopathy but not
of phagocytic activity and sis. Together with pericytes, ECs also contribute to the aHUS169 owing to a lack of complement haemolytic acti­
oxidative burst upon a second increased renal vascular resistance and reduced renal vity as a result of increased C3 and C5 consumption170.
stimulation. Neutrophils can be
primed by microbial products,
blood flow typical of sepsis-​induced AKI by promot- However, mice with hepatocyte-​s pecific FH defi-
chemo-​attractants and ing vasoconstriction and tissue oedema as a result of ciency, which exhibit plasma FH levels of 20% of nor-
inflammatory cytokines. fluid leakage and by facilitating leukocyte adhesion and mal, develop severe renal TMA, although this effect is
microthrombus formation1. Multiple factors (includ- not spontaneous and requires concurrent induction of
Azurophilic granules
ing increased generation of thrombin resulting from serum nephrotoxic nephritis171. Transgenic mice with
Azurophilic granules of
neutrophils are
the heightened exposure of TF as a consequence of EC a truncated FH that is functionally equivalent to that
intracytoplasmic vesicles apoptosis, dysfunction of antithrombin and the throm- of FH mutants in humans with aHUS have also been
containing peptides with bomodulin–protein C anticoagulant system observed in developed172. The presence of a point mutation in FH
antimicrobial activity, such as sepsis and impaired fibrinolysis induced by increased that impairs interaction of FH with host cells without
MPO, proteinase 3 (PR3), α-​
defensins, elastase, cathepsin G
PAI1 expression) result in the endothelial dysfunction reducing plasma FH levels also induces renal TMA and
and bactericidal/permeability that contributes to coagulopathy in sepsis153. Levels systemic thrombosis in mice173. These models are val-
increasing protein (BPI). of soluble thrombomodulin, which reflect the extent of uable for the study of aHUS disease mechanisms and
thrombomodulin detachment from ECs, independently therapeutic strategies. Indeed, blockade of C5 (ref.171)
Histone-​dependent
predicted development of AKI in a large cohort of or properdin, which stabilizes the alternative pathway
cytotoxicity
Pro-​inflammatory and
patients with sepsis admitted to the ICU153. C3 convertase174, showed therapeutic efficacy in animal
cytotoxic signalling induced by models of aHUS. The reason why the kidney micro-
extracellular histones. Histones Defective endothelium protection vasculature is particularly susceptible to complement-​
and DNA normally reside in The renal endothelium is vulnerable, and in addition mediated damage is unclear, but a new study showing
the nucleus and form
nucleosomes. Extracellular
to direct injury, it can be damaged through the loss of that renin can cleave C3 suggests a possible explanation
histones are damage-​ protective factors, such as regulators of the complement for this renal tropism175. Of note, the penetrance of aHUS
associated molecular patterns alternative pathway as occurs in aHUS, through an among carriers of mutations in any of the susceptibi­
(DAMPs), which elicit pro-​ imbalance between pro-​angiogenic and anti-​angiogenic lity genes is incomplete176, and a triggering event (such
inflammatory signalling
factors as occurs in pre-​eclampsia or with use of VEGF as infection or pregnancy) and additional genetic pre-
through Toll-​like receptors
(TLRs) and promote cell death.
blockade therapies in oncology. disposing factors (such as risk haplotypes in FH and
MCP) are needed to induce the disease32,177. Treatment
Opsonization Atypical haemolytic uraemic syndrome. Complement of cultured ECs that had been activated with cytokines,
Tagging of a pathogen, a cell or is a component of the innate immune defence system haem or ADP with serum from patients with aHUS
an apoptotic body, with
opsonins, which increases their
and is critical for protecting the host against path- induced greater complement deposition than treatment
interaction with phagocytes ogens154. The alternative pathway of complement is with serum from healthy donors32,39,178,179. Haemolysis,
and natural killer (NK) cells. constitutively active at low levels as a sentinel against which accompanies early disease stages, can exacerbate
These opsonins can be pathogens; however, owing to its powerful lytic capac- aHUS owing to the complement-​activating capacity of
antibodies (immunoglobulin G
ity, dysregulation or overactivation of this pathway can released haem38–40.
(IgG) or IgE) or complement
fractions (C3b or C4b).
trigger host tissue damage155. A healthy endothelium is
protected against complement activity by the presence Pre-​eclampsia and anti-​VEGF therapy. VEGF, con-
of multiple regulatory molecules, such as FH, factor I, trolling the proliferation, differentiation, migration and
MCP, CD55 and CD59. aHUS is associated with dys- permeability of ECs, is the most potent regulator of angi-
regulation of the alternative pathway, resulting in mas- ogenesis180. Several isoforms of VEGF (predominantly
sive EC opsonization, release of the anaphylatoxins C3a VEGF121, VEGF165 and VEGF189) bind to three trans-
and C5a and formation of sublytic membrane attack membrane VEGF receptors (VEGFRs): VEGFR1 (also
complex C5b-9 on GECs, culminating in endothelial known as FLT1), VEGFR2 (also known as KDR) and

www.nature.com/nrneph
Reviews

a Atypical haemolytic uraemic syndrome and organization of the actin cytoskeleton. Podocyte-​
C3a Spontaneous Alternative specific deletion of sFLT1 leads to massive proteinuria
FI hydrolysis pathway
C3b or trigger regulators in mice, highlighting the role of sFLT1 in preserving the
integrity of the glomerular filtration barrier184. sFLT1
Alternative
pathway auto- released by podocytes might also contribute to the regula-
amplification tion of VEGF function by binding VEGF and inducing its
FH loop
antibody C5a uptake by podocytes184. Furthermore, sFLT1 acts on peri­
Factor B
cytes to modulate the differentiation and proliferation
FH of ECs; selective deletion of FLT1 in vascular pericytes
MCP peripheral to the kidney leads to increased vascularization
C3 convertase and excessive neoangiogenesis within the kidney184.
GEC C5b-9 The importance of this balance between VEGF and
sFLT1 within glomeruli is highlighted by two conditions:
pre-​eclampsia and following the use of anti-​VEGF ther-
b VEGF inhibition apies (Fig. 5b). Pre-​eclampsia is a complication of preg-
nancy characterized by hypertension and proteinuria
Podocyte
occurring after 20 weeks of gestation. It is associated
with a dramatic increase in the release of sFLT1 and sol-
uble endoglin by placental cells, leading to an imbalance
between anti-​angiogenic factors (sFLT1 and endoglin)
Placental and pro-​angiogenic factors (VEGF and placental growth
sFLT1
VEGF factor)185,186. The relative deficiency in VEGF damages
the glomerular filtration barrier, leading to effacement
Anti-VEGF of podocyte foot processes, swelling and detachment of
VEGFR2 antibody ECs from the GBM and ultimately the development
of TMA. Similar renal lesions can be seen in patients who
receive anti-​VEGF drugs for the treatment of cancers183.
Normal crosstalk between Pre-eclampsia Anti-VEGF Up to one-​third of patients receiving anti-​VEGF therapy
EC and podocytes (increased sFLT1) therapy develop hypertension, 20–60% of patients receiving anti-​
Fig. 5 | renal damage associated with defective endothelial protection. a | Atypical VEGF therapy develop proteinuria and a few patients will
haemolytic uraemic syndrome is characterized by deficient regulation of the develop TMA187. Importantly, podocyte-​specific deple-
complement alternate pathway due to quantitative or functional deficiency or tion of VEGF induces the same clinical and pathological
autoimmune inhibition of factor H (FH), factor I (FI) or membrane cofactor protein (MCP), phenotype in mice183. Improved understanding of the bal-
leading to uncontrolled alternative pathway activation with formation of the membrane ance between VEGF and anti-​angiogenic factors might
attack complex C5b-9 and endothelial cell (EC) injury. b | Vascular endothelial growth facilitate the development of approaches to manage severe
factor (VEGF) synthesized by podocytes exerts protective effects on glomerular ECs diseases, as exemplified by the use of apheresis to remove
(GECs) by binding VEGF receptors (VEGFRs). VEGF availability is compromised in circulating sFLT1 in women with pre-​eclampsia188, and
pre-eclampsia, in which increased placental secretion of the soluble VEGFR sFLT1 acts as shed new light on the pathophysiology of other vascular
a decoy to reduce the availability of VEGF to signal through VEGFR2 on GECs. Similarly,
renal diseases, such as AAV189.
use of anti-​VEGF therapies also reduces levels of available VEGF, leading to podocyte
and endothelial damage.
Diabetic kidney disease. DKD is the leading cause of
end-​stage renal disease (ESRD) in high-​income coun-
neuropilin 1 (ref.180). The pro-​angiogenic effect of VEGF tries190,191. In patients with type 1 diabetes mellitus
is mainly mediated through VEGFR2; mice lacking this (T1DM) or type 2 diabetes mellitus (T2DM), DKD is
receptor die in utero owing to an absence of endothelial characterized by glomerular hypertrophy and hyper-
and haematopoietic cells181. Two major splice variants filtration, typically associated with albuminuria, and
of VEGFR1 exist: the full-​length transmembrane recep- progressive CKD. Pathological examination shows
tor and a secreted, soluble protein called soluble FLT1 GBM thickening, mesangial expansion and podocyte
(sFLT1), which lacks the transmembrane and intra- detachment; the development of glomerulosclerosis192,
cellular domains. Full-​length VEGFR1 and sFLT1 are tubular atrophy and interstitial fibrosis 193; and EC
decoy receptors and reduce the availability of VEGF for loss and capillary rarefaction in glomeruli and the
signalling through VEGFR2 (ref.180); mice deficient in tubulo-interstitium194 (Fig. 6a).
VEGFR1 die owing to excess EC growth182. Hyperglycaemia induces apoptosis of GECs, leading
The renal GBM provides one of the best examples of to a progressive reduction in the surface of fenestrated
the importance of VEGF in EC and pericyte physiology. endothelia195–197. This process is triggered by local acti-
Podocytes are the main source of VEGF in the glomer- vation of the RAAS194,198, ROS-​induced activation of
ulus183. Secreted VEGF is delivered to GECs across the caspase 3 (ref.199) and reduced activation of protein C
GBM, against the flow of urine filtrate, through ill-​defined by thrombomodulin200. Under physiological conditions,
mechanisms that probably involve diffusion183, where it activated protein C prevents glucose-​induced apopto-
binds to VEGFR2. Podocytes also produce sFLT1, which in sis of GECs and podocytes by binding to endothelial
an autocrine fashion binds to glycosphingolipid GM3 protein C receptor (EPCR) and activating the serine/
in lipid rafts to promote podocyte adhesion to the GBM threonine-​protein kinase, PAR1, leading to a decrease

Nature Reviews | Nephrology


Reviews

a Diabetic kidney disease


Loss of fenestrae

Glycocalyx disruption
Podocyte foot
process detachment
High glucose
ROS

AGEs

Heparanase

EC

Glycocalyx

Podocyte
↓eNOS
• Reduced NO
• Increased apoptosis
• Increased permeability RAGE

b Arteriolar nephrosclerosis
Bowman
capsule Ischaemic collapse
of capillary loops
(hypoperfusion)
• Reduced NO, prostaglandin I
and epoxyeicosatrienoic acids Smooth
• Increased ET1, ANGII and muscle cell
thromboxane A2

Vessel lumen Mesangial


narrowing cell

Arteriolar Myointimal thickening, SMC


hyalinosis hypertrophy and proliferation

c Endothelial dysfunction in CKD


EC

Reduced repair
Release of
procoagulant factors
Uraemic Reduced
Inflammation
toxins EPC survival
EMPs
TF

AhR activation EC detachment

Reduced NO availability Glycocalyx breakdown


Fig. 6 | endothelial dysfunction in common diseases. a | Multiple pathways lead to endothelial dysfunction in diabetic
kidney disease. Hyperglycaemia, binding of advanced glycation end products (AGEs) to their receptor RAGE and oxidative
stress promote glycocalyx disruption through increased production of heparanase, reduced synthesis of heparan sulfate,
apoptosis of glomerular endothelial cells (GECs) and reduced nitric oxide (NO) synthesis. Progressive loss of GEC
fenestrae and detachment of podocyte foot processes increases glomerular permeability. b | Arteriolar nephrosclerosis
(sometimes called hypertension-​associated nephropathy) is characterized by a narrowing of renal arterioles, with smooth
muscle cell (SMC) hypertrophy and proliferation, and arterial hyalinosis, with downstream ischaemic collapse of
glomerular capillary loops. Renal endothelial dysfunction leads to a decrease in endothelium-​derived vasodilators (such
as NO, prostaglandin I and epoxyeicosatrienoic acids) and an increase in vasoconstrictive mediators (such as endothelin 1
(ET1), angiotensin II (ANGII) and thromboxane A2), which contribute to increased vasoconstriction, promoting hypoxia,
inflammation and fibrosis. c | Chronic kidney disease (CKD) itself can cause endothelial dysfunction. Uraemic toxins
induce endothelial oxidative stress and a procoagulant phenotype, with increased expression of tissue factor (TF), and
release of procoagulant endothelial microparticles (EMPs). CKD also leads to reduced NO bioavailability, breakdown of
the glycocalyx and reduced endothelial repair by mature endothelial cells (ECs) and endothelial progenitor cells (EPCs).
AhR, aryl hydrocarbon receptor; eNOS, endothelial NO synthase; ROS, reactive oxygen species.

www.nature.com/nrneph
Reviews

Heparanase in the mitochondrial apoptosis pathway in glucose-​ conditions, saturation of physiological glucose
An enzyme that degrades stressed cells200. Activated protein-​C-mediated inhi- metabolism pathways in ECs leads to the activation of
polymeric heparan sulfate bition of apoptosis within glomeruli protects against deleterious pathways (such as the polyol pathway and for-
molecules into shorter-​ hyperglycaemia-​induced renal dysfunction and there- mation of sorbitol), which produces more oxidative stress
chain-length oligosaccharides.
The action of heparanase
fore identifies the crucial role of endothelial function in and EC damage216. Inhibition of the polyol pathway
disrupts the endothelial the pathogenesis of diabetic nephropathy. Moreover, attenuates proteinuria, decreases GBM thickening in
glycocalyx. in Pima Indians with T2DM, the reduction in GEC diabetic rats and reduces glomerular hyperfiltration
fenestrae correlated with the level of albuminuria and in humans213.
The polyol pathway
loss of GFR197. Hyperglycaemia also induces alterations GECs might have a reciprocal role in supporting
Pathway that converts hexose
sugars such as glucose into
in the glycocalyx. For example, a reduction in heparan podocyte function through the production of protective
sugar alcohols (polyols). In this sulfate synthesis201 contributes to increased albumin factors such as endothelial NO synthase (eNOS). Diabetic
metabolic pathway, glucose is passage201. In rats with streptozotocin-​induced DKD, mice with eNOS deficiency have accelerated DKD and
reduced to sorbitol (by the proteinuria increased concurrently with decreasing severe podocyte injury198. One way in which chronic
aldose reductase), which is
then converted to fructose (by
heparan sulfate content202. In addition to reduced hep- hyperglycaemia affects eNOS expression is through the
sorbitol dehydrogenase). The arin sulfate synthesis, heparan sulfate degradation is formation of AGEs — a group of proteins and/or lipids
polyol pathway is activated in promoted by the increased synthesis of heparanase by that are glycated following exposure to sugars213,217. The
hyperglycaemia, owing to the GECs and podocytes203,204. The increase in heparanase AGE receptor, RAGE, is upregulated in ECs by hyper-
saturation of physiological
synthesis by podocytes occurs as a result of increased glycaemia213, and binding of AGEs to RAGE increases
glucose metabolism, and leads
to a cellular oxidative stress.
ROS and ANGII205 and the activation of NF-​κB by ROS synthesis in ECs through activation of NAD(P)H
advanced glycation end products (AGEs)206. In Zucker oxidase (NOX2)218,219. The production of ROS by GECs
Sorbitol fatty rats, oxidative stress increased glomerular filtration is increased in hyperglycaemic states via PKC-​dependent
Sugar alcohol derived from barrier permeability to macromolecules by increasing activation of NOX2220,221. AGEs also reduce NO bio-
glucose through the action of
aldose reductase. Sorbitol is a
expression of heparanase207. Heparanase might also availability by decreasing eNOS expression and inacti-
component of the polyol contribute to the inflammatory processes that promote vating NO219. In addition, hyperglycaemia reduces NO
pathway. tubulo-interstitial fibrosis in DKD207,208. bioavailability221 by inducing the expression of ADMA
Activation of PKC by diacylglycerol might also medi- (asymmetric dimethylarginine), an endogenous inhibi-
eNOS uncoupling
ate cell signalling and endothelial hyperpermeability in tor of eNOS213, and through eNOS uncoupling195, leading
Occurs when endothelial NO
synthase (eNOS) is not coupled
DKD209. Inhibition of PKC reduces hyperglycaemia-​ to the formation of superoxide anion (O2−) instead of
with its substrate (mainly induced hyperpermeability of GECs209, whereas inhi- NO. ROS also increases expression of pro-​inflammatory
l-arginine) or cofactors. eNOS bition of the PKC isoform, PKCβ, reduces albumin COX2 (refs221,222) and subsequent production of COX-​
uncoupling results in the excretion as well as glomerular and tubulo-​interstitial derived vasoconstrictor prostanoids221–223, which might
production of the pro-​oxidative
superoxide anion (O2−) instead
injury in diabetic rats210. promote glomerular efferent arteriole constriction and
of NO, which characterizes As mentioned above, podocytes synthesize VEGF9,194, glomerular capillary pressure. ROS production in late
endothelial dysfunction. which acts on VEGFRs expressed by GECs to regu- stages of disease might promote renal fibrosis through
late GEC function211. Reduced VEGF levels have been upregulation of the profibrotic molecules TGFβ1 and
Prostanoids
reported in patients and rodents with diabetes, possibly ANGII224,225. Mesangial expansion and the accumulation
Physiologically active lipid
compounds that are
as a consequence of reduced podocyte number; how- of extracellular matrix (ECM) might also be favoured
metabolites of the fatty acid ever, increased VEGF levels have also been reported by the endothelial synthesis of platelet-​derived growth
arachidonic acid. Prostanoids in association with diabetes194,211,212. Hyperglycaemia factor (PDGF)226, the expression of which is induced by
comprise prostaglandins might also alter the responsiveness of GECs to VEGF211. high glucose213. Blockade of PDGF receptor signalling
(which have vasodilator and
anti-​aggregant properties) and
A study of biopsy samples from patients with diabetes inhibits the proliferation of pericytes and their differen-
thromboxanes (which are showed increased binding of VEGF to its receptor in the tiation into myofibroblasts, attenuating the development
potent vasoconstrictors that endothelium of mildly injured glomeruli but decreased of kidney fibrosis213,227. Genetic deletion of a specific
also activate platelet binding in severely injured glomeruli212. Thus, an initial PDGF (PDGFD) attenuated renal fibrosis after UUO or
aggregation).
upregulation of VEGF signalling in response to endothe- ischaemia–reperfusion injury226.
Incretins lial stress could contribute to increased glomerular per- The fibrotic process of DKD also involves EndMT194,
Glucose-​lowering hormones meability in early stages of DKD, whereas subsequent which is promoted by hyperglycaemia, TGFβ, ANGII,
secreted by the stomach downregulation of VEGF might underlie the ineffec- AGEs35 and the enzyme dipeptidyl peptidase 4 (DPP4)228,
during a meal. Incretins tive capillary repair in advanced stages of disease212,213. which degrades incretins. Inhibition of DPP4 with lina-
increase the release of insulin
from pancreatic β-​cells, slow
VEGF inhibition promotes endothelial injury and accel- gliptin reduces kidney fibrosis associated with EndMT
gastric emptying and inhibit erates the progression of glomerular lesions in mice in diabetic mice229.
glucagon release from with diabetes213. Of note, endothelial injury and loss in DKD is not
pancreatic α-​cells. The two In contrast to adipose or muscle cells, ECs do not compensated by increased repair. In fact, the number
main incretins are glucagon-​
require insulin to uptake glucose194, with different EC of EPCs is reduced in patients with T1DM or T2DM194
like peptide 1 (GLP1) and
gastric inhibitory peptide (GIP),
populations regulating glucose uptake through differ- and is associated with progression of renal disease230.
which are both rapidly ent mechanisms214,215. Although information specific to Moreover, the angiogenic properties of EPCs from
inactivated by the enzyme GECs are lacking, the finding that microvascular reti- patients with diabetes are reduced, with impaired
dipeptidyl peptidase 4 (DPP4). nal ECs are not able to limit glucose uptake, whereas proliferation and ability to incorporate into vascular
ECs in the brain and heart can limit glucose uptake214, structures194, reduced eNOS expression, decreased NO
might in part explain why some populations, such as bioavailability and increased pro-​oxidative properties231.
retinal ECs and GECs, are particularly susceptible to Of interest, new agents that have shown promise in
a high-​glucose environment. Under hyperglycaemic patients with DKD — glucagon-​like peptide 1 receptor

Nature Reviews | Nephrology


Reviews

(GLP1R) agonists and sodium–glucose cotransporter 2 of local vascular homeostasis; increase vasoconstric-
(SGLT2) inhibitors — might in part act by improving tion; and, in turn, promote hypoxia, inflammation and
endothelial function232. fibrosis in the artery and surrounding interstitium.
Beyond the kidney, endothelial dysfunction is
Arteriolar nephrosclerosis implicated in the macrovascular complications of
Epidemiological studies reveal a strong association hypertension, including coronary artery disease, heart
between the presence and severity of arterial hyper- microvascular dysfunction, stroke or conduit arterial
tension and the occurrence of ESRD233. In this con- dysfunction and stiffness. This dysfunction in part
text, endothelial dysfunction is suspected to contribute results from the reduced release of endothelium-​derived
substantially to the pathophysiology of hypertension-​ vasodilatory mediators241,251. Structural (myointimal
induced kidney injury. The typical presentation of this thickening) and functional (vasoconstriction) con-
so-​called arteriolar nephrosclerosis234 involves nonspe- sequences of endothelial dysfunction consequently
cific criteria, including a slow decline in GFR, absent increase exposure of the renal vasculature to alterations
or low-​range proteinuria, normal urine sediment and in blood flow and impaired autoregulation, increasing
the absence of an alternative identifiable cause of kid- the risk of glomerular ischaemia and tissue hypoxia.
ney disease in a patient with long-​standing hypertension. Ultimately, vascular rarefaction at the peritubular capil-
Histology typically demonstrates a narrowing of renal lary level, as a result of endothelial damage and EndMT,
arterioles with smooth muscle cell hypertrophy and an drives tubulo-​interstitial fibrosis and the development
accumulation of ECM in the vascular wall. Additional of CKD252–254.
lesions, dependent on the pathogenic factors and the
chronicity of the disease, include ischaemic glomeruli Systemic endothelial dysfunction associated with CKD.
and tubulo-​interstitial fibrosis (Fig. 6b). Of note, CKD in As described above, renal endothelial injury can result
the context of hypertension is not always causally related, in the development of renal dysfunction. However, CKD
as has been discussed elsewhere235–237. itself is a cause of profound systemic endothelial dys-
Despite this caveat, multiple studies have shown function, which contributes to the cardiovascular burden
that patients with essential hypertension have impaired of this disease255. Endothelial dysfunction, characterized
systemic and renal endothelial function238–241, although by elevated soluble biomarkers such as soluble adhesion
whether renal endothelial dysfunction is a cause molecules or EMPs (Table 1) and altered flow-​mediated
or a consequence of hypertension remains unclear. vasodilation256,257, is observed at every stage of CKD4,258,
Pharmacological inhibition of NO with L-​NAME including children with CKD259,260. Patients with CKD
induces endothelial dysfunction, severe hypertension also show evidence of an imbalance between EC injury
and renal vascular lesions similar to those observed (reflected by the number of EMPs and CECs) and
in patients with malignant hypertension242,243, and repair (reflected by the number of EPCs and CACs)4,261.
fawn-​hooded hypertensive rats, which have impaired The accumulation of uraemic toxins that occurs as
autoregulation of renal blood flow, develop progressive a consequence of impaired renal clearance contributes
kidney failure with age. In this inbred model of spon- to CKD-​associated endothelial dysfunction4; endothe-
taneous renal disease, endothelial dysfunction in the lial dysfunction worsens with progression of CKD and
renal artery precedes the development of kidney dam- improves with resolution of uraemia following kid-
Autoregulation age244 and involves direct exposure of the renal arteri- ney transplantation257. The endothelium is constantly
Autoregulation of renal blood is ole endothelium to increased pressure. Renal ECs also exposed to uraemic toxins that accumulate in the blood
a homeostatic mechanism, integrate signalling from circulating vasoactive and pro-​ of patients with CKD262 (Fig. 6c). The increase in uraemic
relying both on myogenic
response of afferent arterioles
inflammatory mediators. Specifically, studies from the toxin concentrations occurs early in CKD and is observed
(vasoconstriction in case of past couple of years have demonstrated a central role in living kidney donors despite maintaining an estimated
transmural pressure elevation) for components of the immune system, particularly GFR >60 ml/min (ref.263). More than 100 uraemic solutes
and tubulo-​glomerular TH17 and γδ T cells, in endothelial dysfunction and have been described, which can be classified into three
feedback (depending on the
target organ damage in ANGII-​induced hypertension. categories: small molecules, medium-​sized molecules
sensing of sodium chloride
delivery to the macula densa). These deleterious effects are dependent on the arte- and protein-​bound solutes262. Among protein-​bound
Autoregulation protects the rial pro-​inflammatory milieu induced by T cell pro-​ toxins, some indolic compounds that result from trypto-
glomerular capillaries from inflammatory cytokines, especially IL-17A and IFNγ. phan metabolism by gut microbiota have documented
elevations in arterial pressure The attributable renal damage may similarly relate to endothelial toxicity264: they inhibit endothelial prolifer-
and allows the kidney to
maintain a fairly constant
immune-​mediated dysfunction of renal ECs, although ation and wound repair; decrease NO production; and
blood flow and glomerular this remains to be established245,246. Oxidative stress promote EC senescence, EMP formation, leukocyte
filtration rate. also contributes to renal damage in hypertension247. adhesion, glycocalyx disruption and EPC apoptosis265.
Experimental studies have demonstrated that cytoplas- The indolic uraemic toxins indoxyl sulfate and indole-3
Indolic compounds
mic and mitochondrial dysfunction can be induced acetic acid (IAA), for example, promote endothelial oxi-
Chemical compounds
comprising an aromatic bicyclic by ANGII, leading to increased ROS and contributing dative stress266 and induce a pro-​inflammatory267 and
structure resembling that of to endothelial dysfunction and atherosclerosis248–250. procoagulant268 phenotype in different types of ECs.
indole (C8H7N). Indolic uraemic Renal endothelial dysfunction leads to a decrease in Indoxyl sulfate and IAA are ligands for the transcription
compounds result from endothelium-​derived vasodilators (such as NO, prosta- factor aryl hydrocarbon receptor (AhR), which induces
tryptophan metabolism by the
gut and include indoxyl sulfate,
glandin I and epoxyeicosatrienoic acids) and an increase endothelial synthesis of TF and COX2, in addition to
indole-3 acetic acid and in vasoconstrictive mediators (such as ET1, ANGII and increasing expression of the cytochromes CYP1A1 and
indoxyl-​β-d-​glucuronide. thromboxane A2), which contribute to the disruption CYP1B1 for the metabolism of exogenous pollutants64,267.

www.nature.com/nrneph
Reviews

Dioxin Chronic exposure to environmental pollutants such as ANGII receptor blockers (ARBs); however, a new study
Also known as 2,3,7,8-tetra- dioxin, an exogenous ligand of AhR, causes endothelial challenges this view. This study demonstrated the ability
chlorodibenzodioxin (TCDD), dysfunction and cardiovascular disease similar to that of renin to cleave C3 into C3b and C3a, as would a C3
dioxin and dioxin-​like seen in CKD269. Chronic exposure of mice to indoxyl convertase. This cleavage, and consequent complement
compounds are chemical
pollutants of the environment,
sulfate following uninephrectomy270, or UUO269, induces activation, was specifically blocked by the renin inhibitor
resulting from industrial endothelial dysfunction. In patients with CKD, serum aliskiren175. Administration of aliskiren to three patients
practices (mostly incineration levels of indoxyl sulfate and IAA are associated with car- with dense deposit disease (a C3 glomerulopathy asso-
processes) and from forest fires diovascular events and mortality267,271, and reduction in ciated with dysregulation of the alternative pathway of
and volcanic eruptions. Dioxin
indoxyl sulfate concentration with use of the oral sorbent complement) led to decreased systemic and renal com-
is the exogenous ligand of the
transcription factor aryl
AST-120 improves markers of endothelial dysfunction in plement activation. In a separate study, aliskiren and the
hydrocarbon receptor (AhR). mice272 and in humans273. ACE inhibitor enalaprilat were successful in controlling
Exposure to dioxins, mostly malignant hypertension in two paediatric patients with
through ingestion of Therapies that target the endothelium aHUS279. Although these studies indicate that RAAS
contaminated food, is
mutagenic and increases the
Understanding of the physiological roles of the endothe- inhibitors might exert their beneficial effects in part
risks of cardiovascular lium as well as the different mechanisms of EC dysfunc- by inhibiting the deleterious effects of complement on
diseases. tion have led to the development of targeted strategies the endothelium, further studies are warranted to eval-
to improve endothelium health. uate the effect of this treatment on complement and
TMA injury279.
Protection of the glycocalyx Blockade of the complement terminal pathway by
As described earlier, the glycocalyx covers GECs and acts eculizumab, a monoclonal anti-​C5 antibody, has dramat-
as a filtration barrier at fenestrations. Factors that cause ically improved outcomes for adults and children with
injury to GECs (for instance, in the setting of DKD) also aHUS280,281, including recurrent aHUS after transplanta-
induce the sulfation and deacetylation of heparan sulfate tion282. The role of eculizumab in the treatment of other
and activate heparanase in podocytes, resulting in gly- complement-​mediated renal diseases, such as typical
cocalyx injury. Mice lacking heparanase are protected HUS and other forms of renal TMA, remains to be deter-
from streptozotocin-​induced DKD274, and treatment of mined283–286. As mentioned earlier, the anaphylatoxin
diabetic mice with anti-​heparanase antibodies reduces C5a and its receptor contribute to the pathogenesis of
albuminuria and kidney injury274. However, two clinical AAV. Avacopan, an orally administered C5aR antagonist,
trials that aimed to replenish the glycocalyx through the has shown promising results in AAV as a replacement for
oral ingestion of sulodexide (a mixture of proteoglycans) high-​dose corticosteroids287. In addition, experimental
failed to demonstrate a renoprotective effect on GFR targeting of pro-​inflammatory cytokines such as IL-1β or
decline or proteinuria275,276. pro-​inflammatory signalling pathways, such as SMAD–
The potent vasoconstrictor, ET1, is produced by ECs TGFβ or NF-​κB signalling pathways, reduces the inflam-
and other renal cell populations in response to stress. matory consequences of renal endothelial dysfunction
Endothelin-​receptor antagonists (ERAs) have shown in murine models of crescentic glomerulonephritis and
beneficial renal effects in animal models of CKD and DKD and in human ECs in vitro146,288,289.
anti-​proteinuric effects in patients with DKD and non-​
diabetic CKD, even in the presence of optimal RAAS Inhibition of coagulation
blockade 28,277. However, treatment of hypertensive FXa is an important component of the coagulation
patients with ERAs is associated with an increased risk cascade. FXa is responsible for thrombin generation,
of fluid retention and congestive heart failure owing to but it also exerts direct effects on a variety of cells via
a dose-​dependent reduction in the fractional excretion its receptors, PAR1 and PAR2 (ref.290). As mentioned
of sodium, possibly through inhibitory effects on the earlier, the PARs participate in the interplay between
ETB receptor28. Nevertheless, the attenuation of albu- coagulation and inflammation. The mesangial pro-
minuria achieved by the ERA atrasentan in a murine liferation, glomerular inflammation and fibrin depo-
model of diabetic nephropathy277 was associated with sition observed in animal models in response to FXa
restoration of glomerular endothelial glycocalyx cov- signalling through PARs are reversed by FXa inhibi-
erage through downregulation of heparanase and was tion21,291,292. In diabetic db/db mice, inhibition of FXa
associated with a shift in the balance of macrophages attenuates glomerular hypertrophy, ECM accumula-
from a pro-​inflammatory phenotype towards a more tion and proteinuria and limits angiogenesis293. In an
regulatory phenotype. Future studies, such as the ongo- experimental model of kidney transplantation-​induced
ing SONAR trial in patients with DKD278, will determine ischaemia–reperfusion injury, administration of the
whether ERAs can safely slow the progression of renal anti-​F Xa anticoagulation therapy fondaparinux to
dysfunction in patients with DKD. the transplanted kidney, before warm ischaemia and
during cold ischaemia, improved renal function post-​
Anti-​inflammatory strategies transplantation, with reduced indices of fibrosis com-
RAAS blockade has been the cornerstone of nephro- pared with organs treated with heparin294. Targeting
protection in CKD for many years. The benefit of FXa might therefore be promising for preventing the
RAAS blockade has been considered until now as fibrotic evolution of glomerulonephritides and early
a class effect, resulting from the haemodynamic, DKD293,294. By contrast, despite promising results in
anti-​inflammatory and anti-​f ibrotic properties of pilot studies, administration of recombinant activated
angiotensin-​converting-enzyme (ACE) inhibitors and protein C failed to improve outcomes in patients with

Nature Reviews | Nephrology


Reviews

septic shock295. Likewise, the benefit of recombinant endothelium, either indirectly through the assessment
thrombomodulin in sepsis coagulopathy remains to be of circulating biomarkers or directly through the use of
clearly demonstrated296. new imaging techniques to study EC structure in vivo
and/or assess EC function ex vivo, as well as develop-
Fibrogenesis as a target ments in the bioengineering of vessels and organoids,
Irrespective of their origin, interstitial myofibroblasts hold great promise for the development and evaluation
drive fibrosis in CKD. The relative contribution of res- of therapeutic strategies for renal diseases associated
ident interstitial fibroblasts, infiltrating fibrocytes and with endothelial injury. Improved understanding of
cells derived from kidney pericytes, epithelial cells endothelial physiology is required to identify therapeu-
and ECs is still a matter of debate, although cell fate stud- tic targets. For example, knowledge of the mechanisms
ies suggest that EndMT-​derived cells represent approx- by which complement drives endothelial lesions paved
imately 10% of interstitial myofibroblasts297. Despite the the way for the use of C5 blocking strategies in patients
likely limited contribution of EndMT to renal myofi- with aHUS. Understanding of endothelial biology is
broblasts, blocking EndMT by targeting core mediators also needed to anticipate adverse effects of therapeutic
of this phenotypic switch, including TGFβ, TWIST1, agents, such as the adverse consequences of VEGF inhi-
SNAI1 or sirtuin 3, might hold important therapeutic bition. Greater understanding of the interaction between
implications in halting CKD progression298–301. complement and coagulation pathways will also likely
lead to the development of new therapeutic strategies
Conclusions for a number of diseases, including DKD and arteri-
Endothelial activation, and the switch of ECs from a reg- olar nephrosclerosis, leading to improved outcomes for
ulatory state to a prothrombotic and pro-​inflammatory patients with renal diseases.
phenotype, is a common feature of multiple renal
diseases. Improvements in techniques to study the Published online xx xx xxxx

1. Verma, S. K. & Molitoris, B. A. Renal endothelial injury components and impairs activation of protein C in related to fibrinolysis factor expression, in vitro.
and microvascular dysfunction in acute kidney injury. human glomerular endothelial cells. J. Immunol. 196, Microvasc. Res. 47, 377–387 (1994).
Semin. Nephrol. 35, 96–107 (2015). 832–845 (2016). 32. Roumenina, L. T. et al. A prevalent C3 mutation in
2. Roumenina, L. T., Rayes, J., Frimat, M. & 19. Rezaie, A. R. Protease-​activated receptor signalling by aHUS patients causes a direct C3 convertase gain of
Fremeaux-Bacchi, V. Endothelial cells: source, barrier, coagulation proteases in endothelial cells. Thromb. function. Blood 119, 4182–4191 (2012).
and target of defensive mediators. Immunol. Rev. 274, Haemost. 112, 876–882 (2014). 33. Du, L. et al. Interleukin-1β increases permeability and
307–329 (2016). 20. Frank, R. D. et al. The synthetic pentasaccharide upregulates the expression of vascular endothelial-​
3. Molitoris, B. A. Therapeutic translation in acute kidney fondaparinux reduces coagulation, inflammation and cadherin in human renal glomerular endothelial cells.
injury: the epithelial/endothelial axis. J. Clin. Invest. neutrophil accumulation in kidney ischemia-​reperfusion Mol. Med. Rep. 11, 3708–3714 (2015).
124, 2355–2363 (2014). injury. J. Thromb. Haemost. 3, 531–540 (2005). 34. Murakami, S. et al. Expression of adhesion molecules
4. Jourde-​Chiche, N., Dou, L., Cerini, C., Dignat-​George, F. 21. Nomura, K. et al. Roles of coagulation pathway and by cultured human glomerular endothelial cells in
& Brunet, P. Vascular incompetence in dialysis factor Xa in rat mesangioproliferative response to cytokines: comparison to human umbilical
patients—protein-​bound uremic toxins and endothelial glomerulonephritis. Lab Invest. 87, 150–160 (2007). vein and dermal microvascular endothelial cells.
dysfunction. Semin. Dial. 24, 327–337 (2011). 22. Moussa, L., Apostolopoulos, J., Davenport, P., Microvasc. Res. 62, 383–391 (2001).
5. Zoccali, C. et al. The systemic nature of CKD. Nat. Rev. Tchongue, J. & Tipping, P. G. Protease-​activated 35. Betzen, C. et al. Shiga toxin 2a-​induced endothelial
Nephrol. 13, 344–358 (2017). receptor-2 augments experimental crescentic injury in hemolytic uremic syndrome: a metabolomic
6. Tarbell, J. M., Simon, S. I. & Curry, F. R. Mechanosensing glomerulonephritis. Am. J. Pathol. 171, 800–808 analysis. J. Infect. Dis. 213, 1031–1040 (2016).
at the vascular interface. Annu. Rev. Biomed. Eng. 16, (2007). 36. Gomez, S. A. et al. The oxidative stress induced in vivo
505–532 (2014). 23. Chung, H., Ramachandran, R., Hollenberg, M. D. by Shiga toxin-2 contributes to the pathogenicity of
7. Aird, W. C. Endothelial cell heterogeneity. Cold Spring & Muruve, D. A. Proteinase-​activated receptor-2 haemolytic uraemic syndrome. Clin. Exp. Immunol.
Harb. Perspect. Med. 2, a006429 (2012). transactivation of epidermal growth factor receptor 173, 463–472 (2013).
8. Chi, J. T. et al. Endothelial cell diversity revealed by and transforming growth factor-​beta receptor signaling 37. Merle, N. S. et al. Characterization of renal injury and
global expression profiling. Proc. Natl Acad. Sci. USA pathways contributes to renal fibrosis. J. Biol. Chem. inflammation in an experimental model of intravascular
100, 10623–10628 (2003). 288, 37319–37331 (2013). hemolysis. Front. Immunol. 9, 179 (2018).
9. Satchell, S. C. & Braet, F. Glomerular endothelial cell 24. Oe, Y. et al. Coagulation factor Xa and protease-​ 38. Merle, N. et al. Intravascular hemolysis activates
fenestrations: an integral component of the glomerular activated receptor 2 as novel therapeutic targets for complement via cell-​free heme and heme-​loaded
filtration barrier. Am. J. Physiol. Renal Physiol. 296, diabetic nephropathy. Arterioscler Thromb. Vasc. Biol. microvesicles. JCI Insight 3, 96910 (2018).
F947–F956 (2009). 36, 1525–1533 (2016). 39. Frimat, M. et al. Complement activation by heme as a
10. Satchell, S. The role of the glomerular endothelium in 25. Kohan, D. E., Inscho, E. W., Wesson, D. & Pollock, D. M. secondary hit for atypical hemolytic uremic syndrome.
albumin handling. Nat. Rev. Nephrol. 9, 717–725 Physiology of endothelin and the kidney. Compr. Physiol. Blood 122, 282–292 (2013).
(2013). 1, 883–919 (2011). 40. Roumenina, L. T., Rayes, J., Lacroix-​Desmazes, S.
11. Rabelink, T. J. & de Zeeuw, D. The glycocalyx—linking 26. Rossi, G. P. et al. Endothelial factors in the pathogenesis & Dimitrov, J. D. Heme: modulator of plasma systems
albuminuria with renal and cardiovascular disease. and treatment of chronic kidney disease Part I: General in hemolytic diseases. Trends Mol. Med. 22, 200–213
Nat. Rev. Nephrol. 11, 667–676 (2015). mechanisms: a joint consensus statement from the (2016).
12. Stan, R. V., Kubitza, M. & Palade, G. E. PV-1 is a European Society of Hypertension Working Group on 41. Dejana, E., Hirschi, K. K. & Simons, M. The molecular
component of the fenestral and stomatal diaphragms Endothelin and Endothelial Factors and The Japanese basis of endothelial cell plasticity. Nat. Commun. 8,
in fenestrated endothelia. Proc. Natl Acad. Sci. USA Society of Hypertension. J. Hypertens. 36, 451–461 14361 (2017).
96, 13203–13207 (1999). (2018). 42. Li, J., Qu, X. & Bertram, J. F. Endothelial-myofibroblast
13. Rabelink, T. J., Wijewickrama, D. C. & de Koning, E. J. 27. Daehn, I. et al. Endothelial mitochondrial oxidative transition contributes to the early development
Peritubular endothelium: the Achilles heel of the stress determines podocyte depletion in segmental of diabetic renal interstitial fibrosis in streptozotocin-​
kidney? Kidney Int. 72, 926–930 (2007). glomerulosclerosis. J. Clin. Invest. 124, 1608–1621 induced diabetic mice. Am. J. Pathol. 175,
14. Shaw, I., Rider, S., Mullins, J., Hughes, J. & Peault, B. (2014). 1380–1388 (2009).
Pericytes in the renal vasculature: roles in health and 28. Kohan, D. E. & Barton, M. Endothelin and endothelin 43. Egorova, A. D. et al. Lack of primary cilia primes
disease. Nat. Rev. Nephrol. 14, 521–534 (2018). antagonists in chronic kidney disease. Kidney Int. 86, shear-​induced endothelial-​to-mesenchymal transition.
15. Kramann, R. & Humphreys, B. D. Kidney pericytes: 896–904 (2014). Circ. Res. 108, 1093–1101 (2011).
roles in regeneration and fibrosis. Semin. Nephrol. 34, 29. Siragy, H. M. & Carey, R. M. Role of the intrarenal 44. Camenisch, T. D. et al. Temporal and distinct TGFβ
374–383 (2014). renin-​angiotensin-aldosterone system in chronic ligand requirements during mouse and avian
16. Rafii, S., Butler, J. M. & Ding, B. S. Angiocrine kidney disease. Am. J. Nephrol. 31, 541–550 (2010). endocardial cushion morphogenesis. Dev. Biol. 248,
functions of organ-​specific endothelial cells. Nature 30. Clark, S. J. et al. Tissue-​specific host recognition by 170–181 (2002).
529, 316–325 (2016). complement factor H is mediated by differential 45. Basile, D. P. et al. Impaired endothelial proliferation
17. Pober, J. S. & Sessa, W. C. Evolving functions of activities of its glycosaminoglycan-​binding regions. and mesenchymal transition contribute to vascular
endothelial cells in inflammation. Nat. Rev. Immunol. J. Immunol. 190, 2049–2057 (2013). rarefaction following acute kidney injury. Am. J.
7, 803–815 (2007). 31. Louise, C. B. & Obrig, T. G. Human renal microvascular Physiol. Renal Physiol. 300, F721–F733 (2011).
18. Sartain, S. E., Turner, N. A. & Moake, J. L. TNF endothelial cells as a potential target in the 46. Zeisberg, E. M., Potenta, S. E., Sugimoto, H.,
regulates essential alternative complement pathway development of the hemolytic uremic syndrome as Zeisberg, M. & Kalluri, R. Fibroblasts in kidney fibrosis

www.nature.com/nrneph
Reviews

emerge via endothelial-​to-mesenchymal transition. experimental glomerulonephritis. Am. J. Pathol. 163, 97. Morigi, M. et al. Alternative pathway activation of
J. Am. Soc. Nephrol. 19, 2282–2287 (2008). 553–562 (2003). complement by Shiga toxin promotes exuberant C3a
47. Xavier, S. et al. Curtailing endothelial TGFβ signaling is 72. Schirutschke, H. et al. Injured kidney endothelium is only formation that triggers microvascular thrombosis.
sufficient to reduce endothelial-​mesenchymal transition marginally repopulated by cells of extrarenal origin. J. Immunol. 187, 172–180 (2011).
and fibrosis in CKD. J. Am. Soc. Nephrol. 26, 817–829 Am. J. Physiol. Renal Physiol. 305, F1042–F1052 98. Nolasco, L. H. et al. Hemolytic uremic syndrome-​
(2015). (2013). associated Shiga toxins promote endothelial-​cell
48. Huang, X. et al. Loss of caveolin-1 promotes 73. Sangidorj, O. et al. Bone marrow-​derived endothelial secretion and impair ADAMTS13 cleavage of
endothelial-​mesenchymal transition during sepsis: progenitor cells confer renal protection in a murine unusually large von Willebrand factor multimers.
a membrane proteomic study. Int. J. Mol. Med. 32, chronic renal failure model. Am. J. Physiol. Renal Blood 106, 4199–4209 (2005).
585–592 (2013). Physiol. 299, F325–F335 (2010). 99. Tsokos, G. C., Lo, M. S., Costa Reis, P. & Sullivan, K. E.
49. Stasi, A. et al. Emerging role of Lipopolysaccharide 74. Hillebrands, J. L., Klatter, F. A., van Dijk, W. D. New insights into the immunopathogenesis of systemic
binding protein in sepsis-​induced acute kidney injury. & Rozing, J. Bone marrow does not contribute lupus erythematosus. Nat. Rev. Rheumatol. 12,
Nephrol. Dial. Transplant. 32, 24–31 (2017). substantially to endothelial-​cell replacement in 716–730 (2016).
50. Xu-​Dubois, Y. C. et al. Markers of endothelial-​ transplant arteriosclerosis. Nat. Med. 8, 194–195 100. Belizna, C., Duijvestijn, A., Hamidou, M. & Tervaert, J. W.
to-mesenchymal transition: evidence for antibody-​ (2002). Antiendothelial cell antibodies in vasculitis and
endothelium interaction during antibody-​mediated 75. Lekakis, J. et al. Methods for evaluating endothelial connective tissue disease. Ann. Rheum. Dis. 65,
rejection in kidney recipients. J. Am. Soc. Nephrol. 27, function: a position statement from the European 1545–1550 (2006).
324–332 (2016). Society of Cardiology Working Group on Peripheral 101. van Paassen, P. et al. Induction of endothelial cell
51. Goligorsky, M. S. Endothelial progenitor cells: from Circulation. Eur. J. Cardiovasc. Prev. Rehabil. 18, apoptosis by IgG antibodies from SLE patients with
senescence to rejuvenation. Semin. Nephrol. 34, 775–789 (2011). nephropathy: a potential role for anti-​endothelial cell
365–373 (2014). 76. Rabelink, T. J., de Boer, H. C. & van Zonneveld, A. J. antibodies. Ann. NY Acad. Sci. 1108, 147–156 (2007).
52. Sabatier, F., Camoin-​Jau, L., Anfosso, F., Sampol, J. Endothelial activation and circulating markers of 102. Perry, G. J. et al. Antiendothelial cell antibodies in
& Dignat-​George, F. Circulating endothelial cells, endothelial activation in kidney disease. Nat. Rev. lupus: correlations with renal injury and circulating
microparticles and progenitors: key players towards Nephrol. 6, 404–414 (2010). markers of endothelial damage. Q. J. Med. 86,
the definition of vascular competence. J. Cell. 77. Noël, L.-H. Atlas de Pathologie Rénale (Médecine 727–734 (1993).
Mol. Med. 13, 454–471 (2009). Sciences Flammarion, 2008). 103. D’Cruz, D. P. et al. Antibodies to endothelial cells in
53. Ridger, V. C. et al. Microvesicles in vascular homeostasis 78. Babickova, J. et al. Regardless of etiology, progressive systemic lupus erythematosus: a potential marker
and diseases. Position paper of the European Society of renal disease causes ultrastructural and functional for nephritis and vasculitis. Clin. Exp. Immunol. 85,
Cardiology (ESC) Working Group on Atherosclerosis and alterations of peritubular capillaries. Kidney Int. 91, 254–261 (1991).
Vascular Biology. Thromb. Haemost. 117, 1296–1316 70–85 (2017). 104. Kondo, A. et al. The level of IgA antibodies to
(2017). 79. Lachmann, P. et al. Interference with Gsα-​coupled endothelial cells correlates with histological evidence
54. Woywodt, A. et al. Isolation and enumeration of receptor signaling in renin-​producing cells leads to of disease activity in patients with lupus nephritis.
circulating endothelial cells by immunomagnetic renal endothelial damage. J. Am. Soc. Nephrol. 28, PLOS ONE 11, e0163085 (2016).
isolation: proposal of a definition and a consensus 3479–3489 (2017). 105. Skaggs, B. J., Hahn, B. H. & McMahon, M. Accelerated
protocol. J. Thromb. Haemost. 4, 671–677 (2006). 80. Lee, C. J. et al. The clinicopathologic significance of atherosclerosis in patients with SLE—mechanisms and
55. Yao, G. et al. Evaluation of renal vascular lesions using endothelial tubuloreticular inclusions in glomerular management. Nat. Rev. Rheumatol 8, 214–223 (2012).
circulating endothelial cells in patients with lupus diseases. Ultrastruct. Pathol. 37, 386–394 (2013). 106. Vasilev, V. V. et al. Functional characterization of
nephritis. Rheumatology (Oxford) 47, 432–436 (2008). 81. Schiessl, I. M., Hammer, A., Riquier-​Brison, A. & autoantibodies against complement component C3 in
56. Koc, M. et al. Circulating endothelial cells are Peti-Peterdi, J. Just look! Intravital microscopy as the patients with lupus nephritis. J. Biol. Chem. 290,
associated with future vascular events in hemodialysis best means to study kidney cell death dynamics. 25343–25355 (2015).
patients. Kidney Int. 67, 1078–1083 (2005). Semin. Nephrol. 36, 220–236 (2016). 107. Denny, M. F. et al. Interferon-​alpha promotes
57. Faure, V. et al. Elevation of circulating endothelial 82. Hackl, M. J. et al. Tracking the fate of glomerular abnormal vasculogenesis in lupus: a potential
microparticles in patients with chronic renal failure. epithelial cells in vivo using serial multiphoton imaging pathway for premature atherosclerosis. Blood 110,
J. Thromb. Haemost. 4, 566–573 (2006). in new mouse models with fluorescent lineage tags. 2907–2915 (2007).
58. Amabile, N. et al. Circulating endothelial microparticles Nat. Med. 19, 1661–1666 (2013). 108. Kahlenberg, J. M. et al. Inflammasome activation of
are associated with vascular dysfunction in patients 83. Diaspro, A. et al. Multi-​photon excitation microscopy. IL-18 results in endothelial progenitor cell dysfunction
with end-​stage renal failure. J. Am. Soc. Nephrol. 16, Biomed. Eng. Online 5, 36 (2006). in systemic lupus erythematosus. J. Immunol. 187,
3381–3388 (2005). 84. Salmon, A. H. et al. Loss of the endothelial glycocalyx 6143–6156 (2011).
59. Karpman, D., Stahl, A. L. & Arvidsson, I. Extracellular links albuminuria and vascular dysfunction. J. Am. 109. Miyakis, S. et al. International consensus statement on
vesicles in renal disease. Nat. Rev. Nephrol. 13, Soc. Nephrol. 23, 1339–1350 (2012). an update of the classification criteria for definite
545–562 (2017). 85. Chrobak, K. M., Potter, D. R. & Tien, J. Formation of antiphospholipid syndrome (APS). J. Thromb. Haemost.
60. Kirsch, T. et al. Engulfment of apoptotic cells by perfused, functional microvascular tubes in vitro. 4, 295–306 (2006).
microvascular endothelial cells induces proinflammatory Microvasc. Res. 71, 185–196 (2006). 110. Bienaime, F., Legendre, C., Terzi, F. & Canaud, G.
responses. Blood 109, 2854–2862 (2007). 86. Onoe, H. et al. Metre-​long cell-​laden microfibres Antiphospholipid syndrome and kidney disease.
61. Haubitz, M., Dhaygude, A. & Woywodt, A. Mechanisms exhibit tissue morphologies and functions. Nat. Mater. Kidney Int. 91, 34–44 (2017).
and markers of vascular damage in ANCA-​associated 12, 584–590 (2013). 111. de Groot, P. G. & Urbanus, R. T. The significance of
vasculitis. Autoimmunity 42, 605–614 (2009). 87. Huling, J., Ko, I. K., Atala, A. & Yoo, J. J. Fabrication of autoantibodies against β2-glycoprotein I. Blood 120,
62. Erdbruegger, U. et al. Diagnostic role of endothelial biomimetic vascular scaffolds for 3D tissue constructs 266–274 (2012).
microparticles in vasculitis. Rheumatology (Oxford) using vascular corrosion casts. Acta Biomater. 32, 112. Du, V. X., Kelchtermans, H., de Groot, P. G. & de Laat, B.
47, 1820–1825 (2008). 190–197 (2016). From antibody to clinical phenotype, the black box of
63. Gao, C. et al. Thrombotic role of blood and endothelial 88. Soo, J. Y., Jansen, J., Masereeuw, R. & Little, M. H. the antiphospholipid syndrome: pathogenic
cells in uremia through phosphatidylserine exposure Advances in predictive in vitro models of drug-​induced mechanisms of the antiphospholipid syndrome.
and microparticle release. PLOS ONE 10, e0142835 nephrotoxicity. Nat. Rev. Nephrol. 14, 378–393 (2018). Thromb. Res. 132, 319–326 (2013).
(2015). 89. Xinaris, C. et al. In vivo maturation of functional renal 113. Steinkasserer, A., Estaller, C., Weiss, E. H., Sim, R. B.
64. Gondouin, B. et al. Indolic uremic solutes increase organoids formed from embryonic cell suspensions. & Day, A. J. Complete nucleotide and deduced amino
tissue factor production in endothelial cells by the J. Am. Soc. Nephrol. 23, 1857–1868 (2012). acid sequence of human beta 2-glycoprotein I.
aryl hydrocarbon receptor pathway. Kidney Int. 84, 90. Fakhouri, F., Zuber, J., Fremeaux-​Bacchi, V. Biochem. J. 277, 387–391 (1991).
733–744 (2013). & Loirat, C. Haemolytic uraemic syndrome. Lancet 114. Nonaka, M., Matsuda, Y., Shiroishi, T., Moriwaki, K.
65. Combes, V. et al. In vitro generation of endothelial 390, 681–696 (2017). & Natsuume-​Sakai, S. Molecular cloning of mouse
microparticles and possible prothrombotic activity in 91. Keepers, T. R., Gross, L. K. & Obrig, T. G. Monocyte beta 2-glycoprotein I and mapping of the gene to
patients with lupus anticoagulant. J. Clin. Invest. 104, chemoattractant protein 1, macrophage inflammatory chromosome 11. Genomics 13, 1082–1087 (1992).
93–102 (1999). protein 1 alpha, and RANTES recruit macrophages to 115. Giannakopoulos, B. & Krilis, S. A. The pathogenesis of
66. Dignat-​George, F. et al. Endothelial microparticles: the kidney in a mouse model of hemolytic-​uremic the antiphospholipid syndrome. N. Engl. J. Med. 368,
a potential contribution to the thrombotic complications syndrome. Infect. Immun. 75, 1229–1236 (2007). 1033–1044 (2013).
of the antiphospholipid syndrome. Thromb. Haemost. 92. Mallick, E. M. et al. A novel murine infection model for 116. Corban, M. T. et al. Antiphospholipid syndrome: role
91, 667–673 (2004). Shiga toxin-​producing Escherichia coli. J. Clin. Invest. of vascular endothelial cells and implications for risk
67. Karpman, D. et al. Complement interactions with blood 122, 4012–4024 (2012). stratification and targeted therapeutics. J. Am. Coll.
cells, endothelial cells and microvesicles in thrombotic 93. Warnier, M. et al. Trafficking of Shiga toxin/Shiga-​like Cardiol. 69, 2317–2330 (2017).
and inflammatory conditions. Adv. Exp. Med. Biol. 865, toxin-1 in human glomerular microvascular endothelial 117. Holers, V. M. et al. Complement C3 activation is
19–42 (2015). cells and human mesangial cells. Kidney Int. 70, required for antiphospholipid antibody-​induced fetal
68. Renner, B. et al. Cyclosporine induces endothelial cell 2085–2091 (2006). loss. J. Exp. Med. 195, 211–220 (2002).
release of complement-​activating microparticles. J. Am. 94. Obrig, T. G. et al. Endothelial heterogeneity in Shiga 118. Pierangeli, S. S. et al. Requirement of activation of
Soc. Nephrol. 24, 1849–1862 (2013). toxin receptors and responses. J. Biol. Chem. 268, complement C3 and C5 for antiphospholipid
69. Asahara, T. et al. Isolation of putative progenitor 15484–15488 (1993). antibody-mediated thrombophilia. Arthritis Rheum.
endothelial cells for angiogenesis. Science 275, 95. Basu, D. & Tumer, N. E. Do the A subunits contribute 52, 2120–2124 (2005).
964–967 (1997). to the differences in the toxicity of Shiga toxin 1 and 119. Girardi, G. et al. Complement C5a receptors and
70. Ito, T., Suzuki, A., Imai, E., Okabe, M. & Hori, M. Bone Shiga toxin 2? Toxins (Basel) 7, (1467–1485 (2015). neutrophils mediate fetal injury in the
marrow is a reservoir of repopulating mesangial cells 96. Petruzziello-​Pellegrini, T. N. et al. The CXCR4/CXCR7/ antiphospholipid syndrome. J. Clin. Invest. 112,
during glomerular remodeling. J. Am. Soc. Nephrol. SDF-1 pathway contributes to the pathogenesis of 1644–1654 (2003).
12, 2625–2635 (2001). Shiga toxin-​associated hemolytic uremic syndrome in 120. Oku, K. et al. Complement and thrombosis in the
71. Rookmaaker, M. B. et al. Bone-​marrow-derived cells humans and mice. J. Clin. Invest. 122, 759–776 antiphospholipid syndrome. Autoimmun Rev. 15,
contribute to glomerular endothelial repair in (2012). 1001–1004 (2016).

Nature Reviews | Nephrology


Reviews

121. Loupy, A. et al. The Banff 2015 Kidney Meeting anti-​proteinase-3 or anti-​myeloperoxidase antibodies. 171. Vernon, K. A. et al. Partial complement factor H
Report: current challenges in rejection classification J. Leukoc. Biol. 80, 1424–1433 (2006). deficiency associates with C3 glomerulopathy and
and prospects for adopting molecular pathology. Am. 146. Choi, M. et al. Endothelial NF-​κB blockade thrombotic microangiopathy. J. Am. Soc. Nephrol. 27,
J. Transplant. 17, 28–41 (2017). abrogates ANCA-​induced GN. J. Am. Soc. Nephrol. 1334–1342 (2016).
122. Everly, M. J. et al. Incidence and impact of de novo 28, 3191–3204 (2017). 172. Pickering, M. C. et al. Spontaneous hemolytic uremic
donor-​specific alloantibody in primary renal allografts. 147. Schreiber, A. et al. Neutrophil serine proteases syndrome triggered by complement factor H lacking
Transplantation 95, 410–417 (2013). promote IL-1β generation and injury in necrotizing surface recognition domains. J. Exp. Med. 204,
123. Wiebe, C. et al. Rates and determinants of progression crescentic glomerulonephritis. J. Am. Soc. Nephrol. 1249–1256 (2007).
to graft failure in kidney allograft recipients with 23, 470–482 (2012). 173. Ueda, Y. et al. Murine systemic thrombophilia and
de novo donor-​specific antibody. Am. J. Transplant. 148. Jarrot, P. A. & Kaplanski, G. Pathogenesis of ANCA-​ hemolytic uremic syndrome from a factor H point
15, 2921–2930 (2015). associated vasculitis: an update. Autoimmun Rev. 15, mutation. Blood 129, 1184–1196 (2017).
124. Lefaucheur, C. et al. Preexisting donor-​specific HLA 704–713 (2016). 174. Ueda, Y. et al. Blocking properdin prevents
antibodies predict outcome in kidney transplantation. 149. Schreiber, A., Luft, F. C. & Kettritz, R. Phagocyte NADPH complement-​mediated hemolytic uremic syndrome
J. Am. Soc. Nephrol. 21, 1398–1406 (2010). oxidase restrains the inflammasome in ANCA-induced and systemic thrombophilia. J. Am. Soc. Nephrol. 29,
125. Stegall, M. D., Chedid, M. F. & Cornell, L. D. The role GN. J. Am. Soc. Nephrol. 26, 411–424 (2015). 1928–1937 (2018).
of complement in antibody-​mediated rejection in 150. Sorensen, O. E. & Borregaard, N. Neutrophil 175. Bekassy, Z. D. et al. Aliskiren inhibits renin-​mediated
kidney transplantation. Nat. Rev. Nephrol. 8, extracellular traps - the dark side of neutrophils. complement activation. Kidney Int. 94, 689–700
670–678 (2012). J. Clin. Invest. 126, 1612–1620 (2016). (2018).
126. Jindra, P. T., Jin, Y. P., Rozengurt, E. & Reed, E. F. HLA 151. Xiao, H., Schreiber, A., Heeringa, P., Falk, R. J. 176. Sansbury, F. H. et al. Factors determining penetrance
class I antibody-​mediated endothelial cell proliferation & Jennette, J. C. Alternative complement pathway in in familial atypical haemolytic uraemic syndrome.
via the mTOR pathway. J. Immunol. 180, 2357–2366 the pathogenesis of disease mediated by anti-neutrophil J. Med. Genet. 51, 756–764 (2014).
(2008). cytoplasmic autoantibodies. Am. J. Pathol. 170, 52–64 177. Rodriguez de Cordoba, S., Hidalgo, M. S., Pinto, S.
127. Tible, M. et al. Pathologic classification of antibody-​ (2007). & Tortajada, A. Genetics of atypical hemolytic uremic
mediated rejection correlates with donor-​specific 152. Wang, H., Wang, C., Zhao, M. H. & Chen, M. syndrome (aHUS). Semin. Thromb. Hemost. 40,
antibodies and endothelial cell activation. J. Heart Neutrophil extracellular traps can activate alternative 422–430 (2014).
Lung Transplant. 32, 769–776 (2013). complement pathways. Clin. Exp. Immunol. 181, 178. Roumenina, L. T. et al. Hyperfunctional C3 convertase
128. Blogowski, W. et al. Clinical analysis of perioperative 518–527 (2015). leads to complement deposition on endothelial cells
complement activity during ischemia/reperfusion 153. Katayama, S. et al. Markers of acute kidney injury in and contributes to atypical hemolytic uremic
injury following renal transplantation. Clin. J. Am. Soc. patients with sepsis: the role of soluble thrombomodulin. syndrome. Blood 114, 2837–2845 (2009).
Nephrol. 7, 1843–1851 (2012). Crit. Care 21, 229 (2017). 179. Noris, M. et al. Dynamics of complement activation in
129. Roumenina, L. T., Zuber, J. & Fremeaux-​Bacchi, V. 154. Merle, N. S., Church, S. E., Fremeaux-​Bacchi, V. & aHUS and how to monitor eculizumab therapy. Blood
Physiological and therapeutic complement regulators Roumenina, L. T. Complement system part I: molecular 124, 1715–1726 (2014).
in kidney transplantation. Curr. Opin. Organ mechanisms of activation and regulation. Front. 180. Ferrara, N., Gerber, H. P. & LeCouter, J. The biology of
Transplant. 18, 421–429 (2013). Immunol. 6, 262 (2015). VEGF and its receptors. Nat. Med. 9, 669–676 (2003).
130. Jane-​Wit, D. et al. Alloantibody and complement 155. Merle, N. S., Noe, R., Halbwachs-​Mecarelli, L., 181. Shalaby, F. et al. Failure of blood-​island formation and
promote T cell-​mediated cardiac allograft Fremeaux-​Bacchi, V. & Roumenina, L. T. Complement vasculogenesis in Flk-1-deficient mice. Nature 376,
vasculopathy through noncanonical nuclear factor-​κB system part II: role in immunity. Front. Immunol. 6, 62–66 (1995).
signaling in endothelial cells. Circulation 128, 257 (2015). 182. Fong, G. H., Rossant, J., Gertsenstein, M.
2504–2516 (2013). 156. Blanc, C. et al. Overall neutralization of complement & Breitman, M. L. Role of the Flt-1 receptor tyrosine
131. Stites, E., Le Quintrec, M. & Thurman, J. M. factor H by autoantibodies in the acute phase of the kinase in regulating the assembly of vascular
The complement system and antibody-​mediated autoimmune form of atypical hemolytic uremic endothelium. Nature 376, 66–70 (1995).
transplant rejection. J. Immunol. 195, 5525–5531 syndrome. J. Immunol. 189, 3528–3537 (2012). 183. Eremina, V. et al. VEGF inhibition and renal thrombotic
(2015). 157. Jozsi, M. et al. Anti factor H autoantibodies block microangiopathy. N. Engl. J. Med. 358, 1129–1136
132. Hirohashi, T. et al. A novel pathway of chronic allograft C-terminal recognition function of factor H in hemolytic (2008).
rejection mediated by NK cells and alloantibody. Am. uremic syndrome. Blood 110, 1516–1518 (2007). 184. Jin, J. et al. Soluble FLT1 binds lipid microdomains in
J. Transplant. 12, 313–321 (2012). 158. Merinero, H. M. et al. Complete functional podocytes to control cell morphology and glomerular
133. Roda, J. M. et al. Natural killer cells produce T cell-​ characterization of disease-​associated genetic variants barrier function. Cell 151, 384–399 (2012).
recruiting chemokines in response to antibody-​coated in the complement factor H gene. Kidney Int. 93, 185. Venkatesha, S. et al. Soluble endoglin contributes to
tumor cells. Cancer Res. 66, 517–526 (2006). 470–481 (2018). the pathogenesis of preeclampsia. Nat. Med. 12,
134. Hidalgo, L. G. et al. NK cell transcripts and NK cells in 159. Liszewski, M. K. & Atkinson, J. P. Complement regulator 642–649 (2006).
kidney biopsies from patients with donor-​specific CD46: genetic variants and disease associations. 186. Levine, R. J. et al. Soluble endoglin and other circulating
antibodies: evidence for NK cell involvement in Hum. Genom. 9, 7 (2015). antiangiogenic factors in preeclampsia. N. Engl. J. Med.
antibody-​mediated rejection. Am. J. Transplant. 10, 160. Bienaime, F. et al. Mutations in components of 355, 992–1005 (2006).
1812–1822 (2010). complement influence the outcome of Factor 187. Izzedine, H. et al. Kidney diseases associated with anti-​
135. Loupy, A. et al. Gene expression profiling for the I-associated atypical hemolytic uremic syndrome. vascular endothelial growth factor (VEGF): an 8-year
identification and classification of antibody-​mediated Kidney Int. 77, 339–349 (2010). observational study at a single center. Medicine
heart rejection. Circulation 135, 917–935 (2017). 161. Fremeaux-​Bacchi, V. et al. Mutations in complement (Baltimore) 93, 333–339 (2014).
136. Legris, T. et al. Antibody-​dependent NK cell activation C3 predispose to development of atypical hemolytic 188. Thadhani, R. et al. Removal of soluble Fms-​like tyrosine
is associated with late kidney allograft dysfunction and uremic syndrome. Blood 112, 4948–4952 (2008). kinase-1 by dextran sulfate apheresis in preeclampsia.
the complement-​independent alloreactive potential of 162. Schramm, E. C. et al. Mapping interactions between J. Am. Soc. Nephrol. 27, 903–913 (2016).
donor-​specific antibodies. Front. Immunol. 7, 288 complement C3 and regulators using mutations in 189. Le Roux, S. et al. Elevated soluble Flt1 inhibits
(2016). atypical hemolytic uremic syndrome. Blood 125, endothelial repair in PR3-ANCA-​associated vasculitis.
137. Cardinal, H., Dieude, M. & Hebert, M. J. The emerging 2359–2369 (2015). J. Am. Soc. Nephrol. 23, 155–164 (2012).
importance of non-​HLA autoantibodies in kidney 163. Goicoechea de Jorge, E. et al. Gain-​of-function 190. Ritz, E. & Orth, S. R. Nephropathy in patients with
transplant complications. J. Am. Soc. Nephrol. 28, mutations in complement factor B are associated with type 2 diabetes mellitus. N. Engl. J. Med. 341,
400–406 (2017). atypical hemolytic uremic syndrome. Proc. Natl Acad. 1127–1133 (1999).
138. Cross, A. R., Glotz, D. & Mooney, N. The role of the Sci. USA 104, 240–245 (2007). 191. Thomas, M. C., Cooper, M. E. & Zimmet, P. Changing
endothelium during antibody-​mediated rejection: from 164. Marinozzi, M. C. et al. Complement factor B epidemiology of type 2 diabetes mellitus and
victim to accomplice. Front. Immunol. 9, 106 (2018). mutations in atypical hemolytic uremic syndrome-​ associated chronic kidney disease. Nat. Rev. Nephrol.
139. Lahat, N., Bitterman, H., Weiss-​Cerem, L. & Rahat, M. A. disease-relevant or benign? J. Am. Soc. Nephrol. 25, 12, 73–81 (2016).
Hypoxia increases membranal and secreted HLA-​DR in 2053–2065 (2014). 192. Tervaert, T. W. et al. Pathologic classification of
endothelial cells, rendering them T cell activators. 165. Lemaire, M. et al. Recessive mutations in DGKE cause diabetic nephropathy. J. Am. Soc. Nephrol. 21,
Transpl. Int. 24, 1018–1026 (2011). atypical hemolytic-​uremic syndrome. Nat. Genet. 45, 556–563 (2010).
140. Pober, J. S., Merola, J., Liu, R. & Manes, T. D. Antigen 531–536 (2013). 193. Najafian, B., Alpers, C. E. & Fogo, A. B. Pathology of
presentation by vascular cells. Front. Immunol. 8, 166. Delvaeye, M. et al. Thrombomodulin mutations in human diabetic nephropathy. Contrib. Nephrol. 170,
1907 (2017). atypical hemolytic-​uremic syndrome. N. Engl. J. Med. 36–47 (2011).
141. Taflin, C. et al. Human endothelial cells generate Th17 361, 345–357 (2009). 194. Gilbert, R. E. The endothelium in diabetic nephropathy.
and regulatory T cells under inflammatory conditions. 167. Bruneau, S. et al. Loss of DGKepsilon induces Curr. Atheroscler. Rep. 16, 410 (2014).
Proc. Natl Acad. Sci. USA 108, 2891–2896 (2011). endothelial cell activation and death independently of 195. Cheng, H., Wang, H., Fan, X., Paueksakon, P.
142. Jennette, J. C. & Falk, R. J. Small-​vessel vasculitis. complement activation. Blood 125, 1038–1046 & Harris, R. C. Improvement of endothelial nitric
N. Engl. J. Med. 337, 1512–1523 (1997). (2015). oxide synthase activity retards the progression of
143. Lee, K. H. et al. Neutrophil extracellular traps (NETs) 168. Ueda, Y., Gullipalli, D. & Song, W. C. Modeling diabetic nephropathy in db/db mice. Kidney Int. 82,
in autoimmune diseases: a comprehensive review. complement-​driven diseases in transgenic mice: values 1176–1183 (2012).
Autoimmun Rev. 16, 1160–1173 (2017). and limitations. Immunobiology 221, 1080–1090 196. Toyoda, M., Najafian, B., Kim, Y., Caramori, M. L.
144. Williams, J. M. et al. Activation of the Gi heterotrimeric (2016). & Mauer, M. Podocyte detachment and reduced
G protein by ANCA IgG F(ab’)2 fragments is necessary 169. Pickering, M. C. et al. Uncontrolled C3 activation glomerular capillary endothelial fenestration in
but not sufficient to stimulate the recruitment of causes membranoproliferative glomerulonephritis in human type 1 diabetic nephropathy. Diabetes 56,
those downstream mediators used by intact ANCA mice deficient in complement factor H. Nat. Genet. 31, 2155–2160 (2007).
IgG. J. Am. Soc. Nephrol. 14, 661–669 (2003). 424–428 (2002). 197. Weil, E. J. et al. Podocyte detachment and reduced
145. Reumaux, D., Hordijk, P. L., Duthilleul, P. & Roos, D. 170. de Jorge, E. G. et al. The development of atypical glomerular capillary endothelial fenestration promote
Priming by tumor necrosis factor-​alpha of human hemolytic uremic syndrome depends on complement kidney disease in type 2 diabetic nephropathy.
neutrophil NADPH-​oxidase activity induced by C5. J. Am. Soc. Nephrol. 22, 137–145 (2011). Kidney Int. 82, 1010–1017 (2012).

www.nature.com/nrneph
Reviews

198. Peng, H. et al. High glucose induces activation of the oxidative stress, reduced NO, and enhanced PGH(2)/ 249. Dikalova, A. E. et al. Therapeutic targeting of
local reninangiotensin system in glomerular TXA(2) mediation. Am. J. Physiol. Heart Circ. Physiol. mitochondrial superoxide in hypertension. Circ. Res.
endothelial cells. Mol. Med. Rep. 9, 450–456 (2014). 293, H3096–H3104 (2007). 107, 106–116 (2010).
199. Ho, F. M., Liu, S. H., Liau, C. S., Huang, P. J. & 224. Ha, H. & Lee, H. B. Reactive oxygen species amplify 250. Choi, H., Tostes, R. C. & Webb, R. C. Mitochondrial
Lin-Shiau, S. Y. High glucose-​induced apoptosis in glucose signalling in renal cells cultured under aldehyde dehydrogenase prevents ROS-​induced
human endothelial cells is mediated by sequential high glucose and in diabetic kidney. Nephrology (Carlton) vascular contraction in angiotensin-​II hypertensive
activations of c-​Jun NH2-terminal kinase and 10 (Suppl), S7–S10 (2005). mice. J. Am. Soc. Hypertens. 5, 154–160 (2011).
caspase-3. Circulation 101, 2618–2624 (2000). 225. Sutariya, B., Jhonsa, D. & Saraf, M. N. TGF-​β: the 251. Landmesser, U. & Drexler, H. Endothelial function and
200. Isermann, B. et al. Activated protein C protects against connecting link between nephropathy and fibrosis. hypertension. Curr. Opin. Cardiol. 22, 316–320 (2007).
diabetic nephropathy by inhibiting endothelial and Immunopharmacol. Immunotoxicol. 38, 39–49 (2016). 252. Guerrot, D. et al. Progression of renal fibrosis:
podocyte apoptosis. Nat. Med. 13, 1349–1358 226. Buhl, E. M. et al. The role of PDGF-​D in healthy and the underestimated role of endothelial alterations.
(2007). fibrotic kidneys. Kidney Int. 89, 848–861 (2016). Fibrogen. Tissue Repair 5(Suppl. 1), S15 (2012).
201. Singh, A. et al. High glucose causes dysfunction of the 227. Chen, Y. T. et al. Platelet-​derived growth factor 253. Ponnuchamy, B. & Khalil, R. A. Cellular mediators of
human glomerular endothelial glycocalyx. Am. J. receptor signaling activates pericyte-​myofibroblast renal vascular dysfunction in hypertension. Am. J.
Physiol. Renal Physiol. 300, F40–F48 (2011). transition in obstructive and post-​ischemic kidney Physiol. Regul. Integr. Comp. Physiol. 296,
202. van den Born, J. et al. Reduction of heparan sulphate-​ fibrosis. Kidney Int. 80, 1170–1181 (2011). R1001–R1018 (2009).
associated anionic sites in the glomerular basement 228. Shi, S. et al. Interactions of DPP-4 and integrin β1 254. Seccia, T. M. et al. Endothelin-1 drives epithelial-​
membrane of rats with streptozotocin-​induced diabetic influences endothelial-​to-mesenchymal transition. mesenchymal transition in hypertensive
nephropathy. Diabetologia 38, 1169–1175 (1995). Kidney Int. 88, 479–489 (2015). nephroangiosclerosis. J. Am. Heart Assoc. 5, e003888
203. van den Hoven, M. J. et al. Increased expression of 229. Kanasaki, K. et al. Linagliptin-​mediated DPP-4 (2016).
heparanase in overt diabetic nephropathy. Kidney Int. inhibition ameliorates kidney fibrosis in streptozotocin-​ 255. Hirata, Y. et al. Endothelial function and cardiovascular
70, 2100–2108 (2006). induced diabetic mice by inhibiting endothelial-​ events in chronic kidney disease. Int. J. Cardiol. 173,
204. Rabelink, T. J. et al. Heparanase: roles in cell survival, to-mesenchymal transition in a therapeutic regimen. 481–486 (2014).
extracellular matrix remodelling and the development Diabetes 63, 2120–2131 (2014). 256. Chen, J. et al. Interrelationship of multiple endothelial
of kidney disease. Nat. Rev. Nephrol. 13, 201–212 230. Makino, H. et al. Decreased circulating CD34+ cells dysfunction biomarkers with chronic kidney disease.
(2017). are associated with progression of diabetic PLOS ONE 10, e0132047 (2015).
205. van den Hoven, M. J. et al. Regulation of glomerular nephropathy. Diabet. Med. 26, 171–173 (2009). 257. Recio-​Mayoral, A., Banerjee, D., Streather, C. &
heparanase expression by aldosterone, angiotensin II 231. Jarajapu, Y. P. et al. Blockade of NADPH oxidase Kaski, J. C. Endothelial dysfunction, inflammation and
and reactive oxygen species. Nephrol. Dial. Transplant. restores vasoreparative function in diabetic CD34+ cells. atherosclerosis in chronic kidney disease—a cross-​
24, 2637–2645 (2009). Invest. Ophthalmol. Vis. Sci. 52, 5093–5104 (2011). sectional study of predialysis, dialysis and kidney-​
206. An, X. et al. The receptor for advanced glycation 232. Shigiyama, F. et al. Effectiveness of dapagliflozin on transplantation patients. Atherosclerosis 216,
endproducts mediates podocyte heparanase vascular endothelial function and glycemic control in 446–451 (2011).
expression through NF-​κB signaling pathway. Mol. Cell patients with early-​stage type 2 diabetes mellitus: 258. Verbeke, F. H. et al. Flow-​mediated vasodilation in
Endocrinol. (2017). DEFENCE study. Cardiovasc. Diabetol. 16, 84 (2017). end-stage renal disease. Clin. J. Am. Soc. Nephrol. 6,
207. Goldberg, R. et al. Role of heparanase-​driven 233. Klag, M. J. et al. Blood pressure and end-​stage renal 2009–2015 (2011).
inflammatory cascade in pathogenesis of diabetic disease in men. N. Engl. J. Med. 334, 13–18 (1996). 259. Khandelwal, P. et al. Dyslipidemia, carotid intima-​
nephropathy. Diabetes 63, 4302–4313 (2014). 234. Freedman, B. I. & Cohen, A. H. Hypertension-​ media thickness and endothelial dysfunction in
208. Xu, G. et al. Heparanase-​driven inflammation from the attributed nephropathy: what’s in a name? Nat. Rev. children with chronic kidney disease. Pediatr. Nephrol.
AGEs-​stimulated macrophages changes the functions Nephrol. 12, 27–36 (2016). 31, 1313–1320 (2016).
of glomerular endothelial cells. Diabetes Res. Clin. 235. Ehret, G. B. et al. The genetics of blood pressure 260. Kari, J. A. et al. Physiology and biochemistry of
Pract. 124, 30–40 (2017). regulation and its target organs from association endothelial function in children with chronic renal
209. Sward, P. & Rippe, B. Acute and sustained actions of studies in 342,415 individuals. Nat. Genet. 48, failure. Kidney Int. 52, 468–472 (1997).
hyperglycaemia on endothelial and glomerular barrier 1171–1184 (2016). 261. Brunet, P. et al. Does uremia cause vascular dysfunction?
permeability. Acta Physiol. (Oxf.) 204, 294–307 (2012). 236. Ehret, G. B. et al. Genetic variants in novel pathways Kidney Blood Press Res. 34, 284–290 (2011).
210. Kelly, D. J. et al. Protein kinase C beta inhibition influence blood pressure and cardiovascular disease 262. Duranton, F. et al. Normal and pathologic
attenuates the progression of experimental diabetic risk. Nature 478, 103–109 (2011). concentrations of uremic toxins. J. Am. Soc. Nephrol.
nephropathy in the presence of continued 237. Freedman, B. I. et al. The apolipoprotein L1 (APOL1) 23, 1258–1270 (2012).
hypertension. Diabetes 52, 512–518 (2003). gene and nondiabetic nephropathy in African Americans. 263. Rossi, M. et al. Uremic toxin development in living
211. Majumder, S. & Advani, A. VEGF and the diabetic J. Am. Soc. Nephrol. 21, 1422–1426 (2010). kidney donors: a longitudinal study. Transplantation
kidney: more than too much of a good thing. J. 238. Maruhashi, T. et al. Endothelial function is impaired in 97, 548–554 (2014).
Diabetes Complicat. 31, 273–279 (2017). patients receiving antihypertensive drug treatment 264. Vanholder, R., Schepers, E., Pletinck, A., Nagler, E. V.
212. Hohenstein, B. et al. Local VEGF activity but not VEGF regardless of blood pressure level: FMD-​J study (flow-​ & Glorieux, G. The uremic toxicity of indoxyl sulfate
expression is tightly regulated during diabetic mediated dilation Japan). Hypertension 70, 790–797 and p-​cresyl sulfate: a systematic review. J. Am. Soc.
nephropathy in man. Kidney Int. 69, 1654–1661 (2017). Nephrol. 25, 1897–1907 (2014).
(2006). 239. Higashi, Y. et al. Effects of L-​arginine infusion on renal 265. Jourde-​Chiche, N. et al. Levels of circulating
213. Cheng, H. & Harris, R. C. Renal endothelial dysfunction hemodynamics in patients with mild essential endothelial progenitor cells are related to uremic
in diabetic nephropathy. Cardiovasc. Hematol. Disord. hypertension. Hypertension 25, 898–902 (1995). toxins and vascular injury in hemodialysis patients.
Drug Targets 14, 22–33 (2014). 240. Panza, J. A., Quyyumi, A. A., Brush, J. E. Jr J. Thromb. Haemost. 7, 1576–1584 (2009).
214. Rajah, T. T., Olson, A. L. & Grammas, P. Differential & Epstein, S. E. Abnormal endothelium-​dependent 266. Dou, L. et al. The uremic solute indoxyl sulfate induces
glucose uptake in retina- and brain-​derived endothelial vascular relaxation in patients with essential oxidative stress in endothelial cells. J. Thromb.
cells. Microvasc. Res. 62, 236–242 (2001). hypertension. N. Engl. J. Med. 323, 22–27 (1990). Haemost. 5, 1302–1308 (2007).
215. Alpert, E. et al. Delayed autoregulation of glucose 241. Bellien, J. et al. Epoxyeicosatrienoic acids contribute 267. Dou, L. et al. The cardiovascular effect of the uremic
transport in vascular endothelial cells. Diabetologia with altered nitric oxide and endothelin-1 pathways to solute indole-3 acetic acid. J. Am. Soc. Nephrol. 26,
48, 752–755 (2005). conduit artery endothelial dysfunction in essential 876–887 (2015).
216. Mapanga, R. F. & Essop, M. F. Damaging effects of hypertension. Circulation 125, 1266–1275 (2012). 268. Addi, T. et al. Mechanisms of tissue factor induction
hyperglycemia on cardiovascular function: spotlight on 242. Zoccali, C. Endothelial dysfunction in subcutaneous by the uremic toxin indole-3 acetic acid through
glucose metabolic pathways. Am. J. Physiol. Heart small resistance arteries and cardiovascular events. aryl hydrocarbon receptor/nuclear factor-​κ B
Circ. Physiol. 310, H153–H173 (2016). J. Hypertens. 24, 1900–1901; author reply signaling pathway in human endothelial cells. Arch.
217. Frimat, M. et al. Kidney, heart and brain: three organs 1901–1902 (2006). Toxicol. https://doi.org/10.1007/s00204-018-2328-3
targeted by ageing and glycation. Clin. Sci. (Lond.) 243. Boffa, J. J., Tharaux, P. L., Dussaule, J. C. & (2018).
131, 1069–1092 (2017). Chatziantoniou, C. Regression of renal vascular fibrosis 269. Sallee, M. et al. The aryl hydrocarbon receptor-​
218. Wautier, M. P. et al. Activation of NADPH oxidase by by endothelin receptor antagonism. Hypertension 37, activating effect of uremic toxins from tryptophan
AGE links oxidant stress to altered gene expression 490–496 (2001). metabolism: a new concept to understand
via RAGE. Am. J. Physiol. Endocrinol. Metab. 280, 244. Ochodnický, P. et al. Renal vascular dysfunction cardiovascular complications of chronic kidney
E685–E694 (2001). precedes the development of renal damage in the disease. Toxins (Basel) 6, 934–949 (2014).
219. Daroux, M. et al. Advanced glycation end-​products: hypertensive Fawn-​Hooded rat. Am. J. Physiol. Renal 270. Yang, K. et al. Klotho protects against indoxyl
implications for diabetic and non-​diabetic Physiol. 298, F625–F633 (2010). sulphate-​induced myocardial hypertrophy. J. Am. Soc.
nephropathies. Diabetes Metab. 36, 1–10 (2010). 245. Caillon, A. et al. γδ T cells mediate angiotensin Nephrol. 26, 2434–2446 (2015).
220. Schaffer, S. W., Jong, C. J. & Mozaffari, M. Role of II-induced hypertension and vascular injury. Circulation 271. Barreto, F. C. et al. Serum indoxyl sulfate is associated
oxidative stress in diabetes-​mediated vascular 135, 2155–2162 (2017). with vascular disease and mortality in chronic
dysfunction: unifying hypothesis of diabetes revisited. 246. Norlander, A. E. et al. A salt-​sensing kinase in kidney disease patients. Clin. J. Am. Soc. Nephrol. 4,
Vascul. Pharmacol. 57, 139–149 (2012). T lymphocytes, SGK1, drives hypertension and 1551–1558 (2009).
221. Jaimes, E. A., Hua, P., Tian, R. X. & Raij, L. Human hypertensive end-​organ damage. JCI Insight 2, 92801 272. Six, I. et al. Deleterious vascular effects of indoxyl
glomerular endothelium: interplay among glucose, free (2017). sulfate and reversal by oral adsorbent AST-120.
fatty acids, angiotensin II, and oxidative stress. Am. J. 247. Mennuni, S. et al. Hypertension and kidneys: Atherosclerosis 243, 248–256 (2015).
Physiol. Renal Physiol. 298, F125–F132 (2010). unraveling complex molecular mechanisms underlying 273. Yu, M., Kim, Y. J. & Kang, D. H. Indoxyl sulfate-​induced
222. Kiritoshi, S. et al. Reactive oxygen species from hypertensive renal damage. J. Hum. Hypertens. 28, endothelial dysfunction in patients with chronic kidney
mitochondria induce cyclooxygenase-2 gene expression 74–79 (2014). disease via an induction of oxidative stress. Clin. J.
in human mesangial cells: potential role in diabetic 248. Puddu, P., Puddu, G. M., Galletti, L., Cravero, E. & Am. Soc. Nephrol. 6, 30–39 (2011).
nephropathy. Diabetes 52, 2570–2577 (2003). Muscari, A. Mitochondrial dysfunction as an initiating 274. Gil, N. et al. Heparanase is essential for the
223. Toth, E. et al. Contribution of polyol pathway to event in atherogenesis: a plausible hypothesis. development of diabetic nephropathy in mice.
arteriolar dysfunction in hyperglycemia. Role of Cardiology 103, 137–141 (2005). Diabetes 61, 208–216 (2012).

Nature Reviews | Nephrology


Reviews

275. Packham, D. K. et al. Sulodexide fails to demonstrate 288. Ka, S. M. et al. Kidney-​targeting Smad7 gene vein graft remodeling. Sci. Transl Med. 6, 227ra34
renoprotection in overt type 2 diabetic nephropathy. transfer inhibits renal TGF-​β/MAD homologue (SMAD) (2014).
J. Am. Soc. Nephrol. 23, 123–130 (2012). and nuclear factor κB (NF-​κB) signalling pathways, and 300. Mahmoud, M. M. et al. TWIST1 integrates endothelial
276. Lewis, E. J. et al. Sulodexide for kidney protection in improves diabetic nephropathy in mice. Diabetologia responses to flow in vascular dysfunction and
type 2 diabetes patients with microalbuminuria: a 55, 509–519 (2012). atherosclerosis. Circ. Res. 119, 450–462 (2016).
randomized controlled trial. Am. J. Kidney Dis. 58, 289. Shen, W. C. et al. Indoxyl sulfate enhances 301. Mahmoud, M. M. et al. Shear stress induces
729–736 (2011). IL-1β-induced E-​selectin expression in endothelial cells endothelial-​to-mesenchymal transition via the
277. Boels, M. G. et al. Atrasentan reduces albuminuria by in acute kidney injury by the ROS/MAPKs/NFκB/AP-1 transcription factor Snail. Sci. Rep. 7, 3375 (2017).
restoring the glomerular endothelial glycocalyx barrier pathway. Arch. Toxicol. 90, 2779–2792 (2016).
in diabetic nephropathy. Diabetes 65, 2429–2439 290. Borensztajn, K., Peppelenbosch, M. P. & Spek, C. A. Acknowledgements
(2016). Factor Xa: at the crossroads between coagulation and The authors are grateful to all members of the French Renal
278. Heerspink, H. J. L. et al. Rationale and protocol of the signaling in physiology and disease. Trends Mol. Med. Endothelial Society (FRENDS) for fruitful discussions.
Study Of diabetic Nephropathy with AtRasentan 14, 429–440 (2008). FRENDS is a collaborative group of nephrologists and
(SONAR) trial: a clinical trial design novel to diabetic 291. Tanaka, M. et al. Role of coagulation factor Xa and researchers working on the endothelium in different renal
nephropathy. Diabetes Obes. Metab. 20, 1369–1376 protease-​activated receptor 2 in human mesangial cell diseases. The research was supported by grants from the
(2018). proliferation. Kidney Int. 67, 2123–2133 (2005). Agence Nationale de la Recherche (ANR) JCJC–INFLACOMP
279. Raghunathan, V. et al. Targeting renin-​angiotensin 292. Shimosawa, M. et al. Lipopolysaccharide-​triggered 2015–2018 (ANR-15-CE15-0001; to L.T.R.); INSERM (to
system in malignant hypertension in atypical hemolytic acute aggravation of mesangioproliferative L.T.R.); EU FP7 grant 2012–305608 (EURenOmics;
uremic syndrome. Indian J. Nephrol. 27, 136–140 glomerulonephritis through activation of coagulation to V.F.-B.); Assistance Publique-​Hôpitaux de Paris (AP-​HP)-
(2017). in a high IgA strain of ddY mice. Nephron Exp. Programme Hospitaliser de Recherche Clinique (PHRC)
280. Verhave, J. C., Wetzels, J. F. & van de Kar, N. C. Novel Nephrol. 112, e81–e91 (2009). AOM08198 (to V.F.-B.); Association pour l'Information et la
aspects of atypical haemolytic uraemic syndrome and 293. Sumi, A. et al. Roles of coagulation pathway and factor Recherche sur les maladies rénales Génétiques (AIRG) (to
the role of eculizumab. Nephrol. Dial. Transplant. Xa in the progression of diabetic nephropathy in db/db V.F.-B.); the French Society of Nephrology (to V.F.-B.); a grant
29(Suppl. 4), iv131–iv141 (2014). mice. Biol. Pharm. Bull. 34, 824–830 (2011). from Appel d'Offre de Recherche Clinique (AORC) 2011-
281. Mortara, A. Lo studio RELAX-​AHF [Italian]. G. Ital. 294. Tillet, S. et al. Kidney graft outcome using an anti-​Xa A01347 (to G.K.); and Assistance Publique-​Hôpitaux de
Cardiol. (Rome) 16, 193–197 (2015). therapeutic strategy in an experimental model of Marseille (AP-​HM)-PHRC grant 2012-A00217-36 (to G.K.).
282. Legendre, C. et al. Eculizumab in renal severe ischaemia-​reperfusion injury. Br. J. Surg. 102,
transplantation. Transplant Rev. (Orlando) 27, 90–92 132–142; discussion 142 (2015). Author contributions
(2013). 295. Annane, D. et al. Recombinant human activated All authors contributed to researching data for the article,
283. Noris, M., Mescia, F. & Remuzzi, G. STEC-​HUS, protein C for adults with septic shock: a randomized discussing the article’s content and writing, reviewing and
atypical HUS and TTP are all diseases of controlled trial. Am. J. Respir. Crit. Care Med. 187, editing the manuscript before submission.
complement activation. Nat. Rev. Nephrol. 8, 1091–1097 (2013).
622–633 (2012). 296. Yamakawa, K. et al. Recombinant human soluble Competing interests
284. Loos, S., Oh, J. & Kemper, M. J. Eculizumab in thrombomodulin in severe sepsis: a systematic review F.F. has received consultancy fees and/or speaker honoraria
STEC-HUS: need for a proper randomized controlled and meta-​analysis. J. Thromb. Haemost. 13, 508–519 from Alexion and Roche. M.L.Q. has received consultancy
trial. Pediatr. Nephrol. 33, 1277–1281 (2018). (2015). fees and/or speaker honoraria from Alexion. The other
285. Le Quintrec, M. et al. Patterns of clinical response to 297. LeBleu, V. S. et al. Origin and function of authors declare no competing interests.
eculizumab in patients with C3 glomerulopathy. Am. J. myofibroblasts in kidney fibrosis. Nat. Med. 19,
Kidney Dis. 72, 84–92 (2018). 1047–1053 (2013). Publisher’s note
286. Oosterveld, M. J. et al. Eculizumab in pediatric 298. Lin, J. R. et al. Suppression of endothelial-​ Springer Nature remains neutral with regard to jurisdictional
dense deposit disease. Clin. J. Am. Soc. Nephrol. 10, to-mesenchymal transition by SIRT (Sirtuin) 3 claims in published maps and institutional affiliations.
1773–1782 (2015). alleviated the development of hypertensive renal
287. Jayne, D. R. W. et al. Randomized trial of C5a receptor injury. Hypertension 72, 350–360 (2018). Supplementary information
inhibitor avacopan in ANCA-​associated vasculitis. 299. Cooley, B. C. et al. TGF-​β signaling mediates Supplementary information is available for this paper at
J. Am. Soc. Nephrol. 28, 2756–2767 (2017). endothelial-​to-mesenchymal transition (EndMT) during https://doi.org/10.1038/s41581-018-0098-z.

www.nature.com/nrneph

You might also like