You are on page 1of 16

REVIEWS

The emerging role of coagulation


proteases in kidney disease
Thati Madhusudhan1, Bryce A.Kerlin2 and Berend Isermann1
Abstract | A role of coagulation proteases in kidney disease beyond their function in normal
haemostasis and thrombosis has long been suspected, and studies performed in the past 15years
have provided novel insights into the mechanisms involved. The expression of protease-activated
receptors (PARs) in renal cells provides a molecular link between coagulation proteases and renal
cell function and revitalizes research evaluating the role of haemostasis regulators in renal disease.
Renal cell-specific expression and activity of coagulation proteases, their regulators and their
receptors are dynamically altered during disease processes. Furthermore, renal inflammation
andtissue remodelling are not only associated, but are causally linked with altered coagulation
activation and protease-dependent signalling. Intriguingly, coagulation proteases signal through
more than one receptor or induce formation of receptor complexes in a cell-specific manner,
emphasizing context specificity. Understanding these cell-specific signalosomes and their
regulation in kidney disease is crucial to unravelling the pathophysiological relevance of
coagulation regulators in renal disease. In addition, the clinical availability of small molecule
targeted anticoagulants as well as the development of PAR antagonists increases the need
forin-depth knowledge of the mechanisms through which coagulation proteases might
regulaterenal physiology.

Tight regulation of coagulation proteases is required coagulation system modulates renal health3,4 (TABLE1).
for normal haemostasis in the kidney. Coagulation is Such knowledge might enable nephrologists to exploit
intimately associated with inflammation and the func the beneficial effects of such interventions while simulta
tions of the haemostatic system extend beyond main neously avoiding the detrimental aspects. In this Review
tenanceof vascular integrity and prevention of excessive we summarize current knowledge regarding the role of
blood loss. This crosstalk is reflected phylogenetically by coagulation proteases in renal disease.
the haemocyte or amoebocyte, the sole circulating blood
element of the horseshoe crab (Limulus polyphemus)1, The coagulation system
which simultaneously fulfils the functions of platelets The coagulation system is traditionally viewed as a
and phagocytic cells. Vertebrates do not have a single cascade-like system with two activation pathways: the
1
Institute of Clinical Chemistry
cell type that unifies the coagulation system and innate tissue factor (TF; also known as extrinsic) pathway and
and Pathobiochemistry,
Medical Faculty, Otto-von- inflammatory regulators, but soluble coagulation pro the contact (also known as intrinsic) pathway5 (FIG.1).
Guericke-University, teases regulate haemostasis as well as inflammation and TFis a type1 transmembrane receptor that shares
Magdeburg, Leipziger tissue remodelling 2. substantial structural homology with typeII cytokine
Strasse44, Magdeburg The close association of inflammation and tissue receptors. Full length TF is a 263 amino acid, single-
D39120, Germany.
2
Center for Clinical and
remodelling with renal disease has prompted research chain polypeptide with a 219 amino acid extracellular
Translational Research, ers to address the functions of haemostatic regulators in Nterminus, a 23 amino acid transmembrane domain
Nationwide Childrens this setting. The discovery of protease-activated recep and a 21 amino acid intracellular Cterminus6. As TF is
Hospital, 700 Childrens tors (PARs), the pivotal cellular receptors for coagulation physiologically expressed by perivascular cells but not
Drive, W325 Columbus,
proteases, provided new impetus leading to novel patho by resting endothelial cells, leucocytes or other cells in
Ohio43205, USA.
Correspondence to B.I.
genic concepts in various organ systems. Moreover, the direct contact with blood (with the exception of placen
berend.isermann@ increasing clinical utilization of anticoagulants targeting tal trophoblasts and cancer cells), it comes into contact
med.ovgu.de specific coagulation proteases and the advent of PAR with blood only upon vascular injury or following its
doi:10.1038/nrneph.2015.177 antagonists warrant additional investigation to gain induction (for example by inflammatory cytokines) on
Published online 23 Nov 2015 deeper insights into the mechanisms through which the endothelial cells or leucocytes.

94 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

Key points Several physiological anticoagulant mechanisms pre


vent excessive coagulation activation. Tissue factor path
In addition to their pivotal and well-established functions in haemostasis, coagulation way inhibitor (TFPI) is a Kunitz-type protease inhibitor
regulators and receptors mediate non-haemostatic functions in the kidney that constrains the activity of the TF/factorVIIa and
Derangements of the coagulation system and altered coagulation-protease- prothrombinase complexes, thus restricting initiation of
dependent signalling in renal disease might alter disease progression coagulation. The serine protease inhibitor antithrombin
Coagulation proteases alter the function of a variety of renal cell types via distinct (also known as antithrombin III) inhibits multiple coagu
protease-activated receptors (PARs) and co-receptors lation proteases, including its primary target thrombin
Activated proteinC has nephroprotective effects that are at least partly independent and factors VIIa, IXa, Xa, XIa and XIIa. Antithrombin
of its anticoagulant function also inhibits other serine proteases, such as kallikrein,
The new drug classes of target-specific oral anticoagulants and PAR inhibitors might plasmin and trypsin as well as the complement lec
interfere with the functions of coagulation proteases in renal disease, with potential tin pathway. The inhibitory activity of antithrombin is
beneficial or adverse effects enhanced ~2,0004,000 fold in the presence of heparin14.
A more specific and equally important anticoagu
lant mechanism is initiated by endothelial thrombo
TF is induced in diverse renal diseases and modu modulin, which modulates the function of thrombin.
lates renal function through various mechanisms. It When bound to thrombomodulin, thrombin loses its
lacks intrinsic proteolytic activity, but can bind factorVII pro-coagulant function but efficiently activates pro
orfactor VIIa in a Ca2+-dependent manner, promoting tein C15. Activated protein C (aPC) in turn inactivates
factor VII activation or enhancing the catalytic activity cofactors Va and VIIIa, thus efficiently dampening
of factor VIIa7. The TF/VIIa complex interacts with and amplification of thrombin generation. In addition, the
activates factor IX and factor X. Activated factor X ena thrombomodulinthrombin complex activates TAFI,
bles formation of the prothrombinase complex (com which removes Cterminal lysine and arginine residues
prised of factor Xa, factor Va, calcium and negatively from peptides. In the case of fibrin these Cterminal resi
charged phospholipids of activated cells such as platelets). dues are required for tissue-type plasminogen activator
The prothrombinase complex promotes the conversion (tPA)-mediated activation of plasminogen. All of these
of prothrombin to thrombin (also known as factorIIa). anticoagulant regulators have functions in renal diseases.
Although the initial quantity generated is small, throm The formation of a fibrinplatelet aggregate is not the
bin initiates an amplification loop via factorXIa genera final step in haemostasis and is succeeded by the equally
tion, activates the non-proteolytic c ofactors V and VIII well-controlled process of fibrinolysis. This proteolytic
(resulting in an ~1,000fold acceleration in thrombin process is not restricted to fibrin but might also target
generation) and recruits activated platelets, thus ensur the extracellular matrix. Plasmin-mediated fibrinolysis
ing the localized production of haemostatic thrombin and degradation of extracellular matrix is highly relevant
concentrations (FIG.2). Subsequently, thrombin acti in the context of fibrotic renal diseases. In addition, the
vates fibrinogen and platelets (via PARs), generating proteases involved in fibrinolysis might either activate
fibrin-platelet aggregates known as bloodclots. or disarm PARs and can directly modulate cellular func
The role of the contact pathway in coagulation has tion through urokinase plasminogen activator receptor
been redefined in the past decade8. A series of elegant (uPAR)16,17. The roles of fibrinolysis and uPAR in renal
studies have established a factorXII-dependent mech disease have been reviewed previously 1719.
anism in the pathophysiology of thrombo-occlusive dis
eases, even though factor XII activation is dispensable Protease activated receptors
for haemostasis8. These studies demonstrate that haemo Beyond its indispensable haemostatic functions, the
stasis (prevention of excessive blood loss) and throm coagulation system has non-haemostatic functions,
bosis (excess thrombus formation) are mechanistically for example in inflammation and tissue remodel
separable, suggesting that pharmaceutical interventions ling. The discovery of PARs provided insight into the
that target pathological thrombosis without interfering non-haemostatic functions of coagulation proteases.
with normal haemostasis are feasible9. Although well accepted in basic research, knowledge
The dependence of factor XII activation on negatively of these non-haemostatic functions has not yet been
charged surfaces has been known for almost a century 10, translated into clinical applications.
but the importance of this phenomenon was eluci PARs are members of the class A family of rhodopsin-
dated only in the past decade, with the discovery that like G protein-coupled receptors (GPCRs) and are pre
polyphosphates have the ability to enhance factor XII dominantly organized within lipid rafts20. Four distinct
activation8,11,12. This effect and additional polyphosphate- PARs have been identified (PAR1, PAR2, PAR3 and
dependent effects on the coagulation system, such as PAR4). Unlike the metabotropic members of the class C
promoting factor V activation and enhancing the anti family of GPCRs, which are obligate dimers, PARs can
fibrinolytic activity of thrombin activable fibrinolysis function as protomers as well as heterodimers. This abil
inhibitor (TAFI, also known as carboxypeptidase B2), ity enables PARs to activate diverse signalling pathways
depend on the polymer length, which varies depending in tissue, context and temporally specific fashions. PARs
on the source (for example, very long polymers pres transmit cellular responses initiated by a number of coag
ent in microorganisms and shorter polymers present ulation regulators, for example factor IIa (PAR1, PAR3
inplatelets)11,13. and PAR4), aPC (PAR1 and PAR3), factor Xa (PAR2) or

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 95

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | Coagulation regulators with known functions in renal physiology and pathophysiology
Coagulation regulator Function Expression Effects in renal physiology and pathophysiology Refs
(alternative name(s))
Procoagulant
Tissue factor Initiator; cofactor for Subendothelium Induced in CGN, TMA, DN and endotoxaemia 3338,40,41,
(thromboplastin, factor VIIa in factor IX 4345,98,
Inhibition protects against CGN and TMA
CD142, factor III) and factor X activation 102, 190
Factor Xa Enzyme Plasma Inhibition is protective in DN, endotoxaemia, MsPGN and IR 4951,97,136
Factor VIII Cofactor for activated Plasma Links impaired kidney function with risk of venous 75
(antihaemophilic factor) factor IX in factor X thrombosis
activation
Factor V Cofactor for factor Platelets Factor Vdependent initiation of coagulation in human 42,112
(labile factor) Xa in prothrombin andplasma mesangial cells invitro
activation
FV Leiden mutation conveys protective effects in DN
Prothrombin/ thrombin Zymogen and Plasma Elevated in AKI, nephrotic syndrome, CKD, CGN and other 55,56,90,106,
(factor IIa) protease renal diseases 108,135,
54, 165
Inhibition protects against tubular atrophy, AKI and
proinflammatory and proliferative responses in proximal
tubular cells and mesangial cells
Fibrinogen Fibrin precursor Plasma, platelets Plasma levels are elevated in diabetic patients and after 48,119,147,
(factor I) renal IR 149151
Heterozygous but not homozygous depletion is protective
in AKI, CGN and IR
B 1542 peptide protects against renal IRinjury
Anticoagulant
Thrombomodulin Cofactor for thrombin Endothelial Shedding of thrombomodulin and elevated plasma levels of 81,86,101,
in protein C activation surface soluble thrombomodulin in diabetes and CKD 103105,131,
133,181
Soluble thrombomodulin is protective in experimental
glomerulonephritis and IR
Thrombomodulin lectin-like domain deficiency exacerbates
HUS and DN
Protein C Zymogen Plasma Renal-specific expression is reduced in various kidney 25,28,29,46,
andprotease diseases 82,100,167,
168,172
Activation is impaired in animals and patients with diabetes
and in renal insufficiency
Treatment with activated protein C is protective in
experimental DN, IR andendotoxaemia
Antithrombin Protease inhibitor Plasma Deficient in experimental nephrotic syndrome 145,191
(heparin cofactor)
Protects against tubular atrophy, AKI and PAN-induced
nephrosis
Tissue factor Protease inhibitor Endothelial Induced in CGN Elevated levels partially compensate 125,182,
pathwayinhibitor surface via GPI, coagulation activation in diabetes 184,185
(extrinsic pathway platelets, plasma
Deficiency inhibits experimental glomerulonephritis
inhibitor)
Inhibition is protective inexperimental kidney fibrosis
AKI, acute kidney injury; CGN, chronic glomerulonephritis; CKD, chronic kidney disease; DN, diabetic nephropathy; GPI, glycosylphosphatidyl inositol; HUS, haemolytic
uraemic syndrome; IR, ischaemiareperfusion; MsPGN, mesangioproliferative glomerulonephritis; PAN, puromycin aminonucleoside; TMA, thrombotic microangiopathy.

plasmin (PAR1 and PAR4), as well as non-coagulation Nterminal sequence, which acts as a tethered ligand
proteases, for example matrix m etalloproteinase1 (PAR1), inducing a conformational rearrangement and irreversi
tryptase or matriptase (PAR2), cathepsin G (PAR4) and ble activation of heterotrimericG proteins4,26 (FIG.3). This
cathepsinS (PAR2)15,16,2128 (TABLE2). mechanism is in contrast to that of most GPCRs, which
The functions of individual PAR protomers and are reversibly activated.
downstream signalling cascades have been extensively The expression of PARs is highly heterogeneous and
studied, but the mechanisms of heterodimerization and more than one PAR is expressed on many cell types. In
their pathophysiological relevance remain incompletely addition, expression of PARs in some cell types is spe
explored. PARs have a unique activation mechanism: cies specific. For example, human platelets express PAR1
following cell surface localization the protease cleaves and PAR4 and human podocytes predominatelyexpress
the extracellular Nterminus, unmasking a cryptic PAR2 and PAR3, whereas murine platelets expressPAR3

96 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

andPAR3. Despite activation of PAR1 and PAR3 by both


Tissue factor pathway Contact activation pathway Polyphosphates
(haemostasis) (thrombosis) thrombin and aPC, these proteases convey opposing
cellular effects20. Biased (that is, functionally selective)
Negatively Activated
charged surfaces platelets and cell-specific PAR-mediated signalling is thought to
Tissue injury
TF Cytokines result from distinct cleavage sites, which result in distinct
neoN-termini, and cell-specific supramolecular recep
FXIIa FXII
TFPI
tor complexes (that is, PAR signalosomes)15,2325. PARs
can form homodimers or heterodimers either consti
AT FXIa FXI tutively or upon activation, depending on the cell type,
FVII TF/FVIIa extracellular protease, specific PAR that is activated and
aPC the presence of co-receptors. In addition to PARPAR
AT FIXa FIX
FVIIIa interactions, PARs can interact with other receptors (for
example sphingosine 1phosphate receptor1 [S1P1], epi
dermal growth factor receptor [EGFR] and P2Y purino
aPC ceptor12), emphasizing the involvement of cell-specific
FX AT FXa FX signalosomes2326 (FIG.3).
FVa
Coagulation regulators and receptors
Coagulation regulators and receptors for coagula
Common pathway tion protease signalling are widely expressed in renal
FII AT (thrombin generation)
Thrombin cell types (FIG.4). For example, thrombomodulin and
endothelial protein C receptor (EPCR) are expressed
aPC aPC PC onendothelial cells. Expression of thrombomodulin and
EPCR is reduced in acute inflammatory states (for exam
Feedback ple sepsis) or chronic diseases (for example diabetes
Signalling Fibrinogen amplication
mellitus)3234. In a model of endotoxaemia, genetically
modified mice with reduced EPCR expression (10%
Platelet Feedback Thrombomodulin
Fibrin activation inhibition and thrombin of wild-type levels) displayed enhanced albuminuria
Tissue
remodelling and renal haemorrhage, demonstrating a crucial role
Inammation Blood clotting Feedback regulation of EPCR in the regulation of vascular permeability in
the kidney 35. EPCR was likewise required for vascular
Figure 1 | The coagulation system. The tissue factor (TF) and contact
Nature activation
Reviews pathways protection in lung and brain, but to a lesser extent in
| Nephrology
lead to a common pathway that generates thrombin. In the tissue factor pathway, tissue other organs an observation that is consistent with the
injury or inflammatory cytokines induce cell-surface expression of TF. The TF/FVIIa complex concept of organ-specific regulation of coagulation35,36.
activates FX, which converts FII to thrombin. In the contact activation pathway, negatively An organ-specific function has likewise been identi
charged surfaces (such as phospholipids and polyphosphates from activated platelets)
fied for the cytoplasmic domain of TF37. Mice that lack
activate FXII, initiating a cascade leading to FX activation and thrombin generation.
this domain express tumour necrosis factor (TNF) on
Thrombin triggers formation of blood clots, provides feed-back amplification or inhibition
of the coagulation activation process and engages in receptor-dependent signalling. their glomeruli from the age of 6weeks and sponta
Localization of the coagulation cascade to cell surfaces ensures a spatial constraint on neously develop albuminuria, podocyte effacement
thrombin generation. Feed-back amplification is provided by activation of thenon-catalytic and loss . They are sensitive to glomerular injury, but
38

cofactors FV and FVIII, platelet activation and thrombin-mediated activation of FXI. Excess resistant to lipopolysaccharide-induced systemic inflam
coagulation activation is averted through several anticoagulant mechanisms, including mation or arthritis3840. The spontaneous phenotypes
inhibition of the TF/FVIIa/FXa complex by tissue factor pathway inhibitor (TFPI), inhibition ofthese mice exemplify the non-haemostatic functionof
of several coagulation factors by antithrombin (AT) and proteolytic inactivation of FVa and the coagulation system, as the cytoplasmic domain is
FVIIIa by activated protein C (aPC). As aPC is generated by the FIIa/thrombomodulin not required for the pro-coagulant function of TF, but
complex on undisturbed endothelial cells, local generation of FIIa following endothelial
is involved in TF signalling. The renal pathology is exa
orvascular injury triggers aPC formation in a spatially and temporally limited fashion.
cerbated in a glomerulonephritis model independent of
Thrombin, aPC and other coagulation proteases interact with protease activated
receptors,initiating cellular signalling that regulates inflammation and tissue remodelling. leucocyte TF expression, suggesting a pathogenic func
a,activated; F, factor; PC, protein C. tion of renal TF expression38,41. Although the pathophysi
ological relevance of TF and the cytoplasmic domain has
been shown in murine models, the expression pattern
and PAR4 29 and murine podocytes predominately of TF in renal cell types remains to be characterized in
express PAR1 and PAR330. The diversity of protease- mice4244. Conversely, in humans and rabbits, glomeru
dependent signalling is furthermore exemplified by the lar TF expression has been demonstrated inpodocytes,
TF/VIIa/Xa complex and the serine proteases thrombin parietal epithelial cells and possibly also in mesangial
and aPC. The TF/VIIa/Xa complex can activate PAR2, cells4548. Notably, increased TF expression has been
but the cytoplasmic domain of TF (which can be phos detected in acute and chronic kidney diseases (CKDs)
phorylated but is dispensable for coagulation activation) in humans and rodents4953. AsTFPI is expressed in
independently regulates cellular functions such as adhe interstitial blood vessels, but not in glomeruli, glom
sion or migration31. Thrombin directly interacts with erular TF expression is not opposed by c oncomitant
PAR1, PAR3 and PAR4, whereas aPC interacts with PAR1 TFPIexpression54.

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 97

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

PAR2 and PAR4 varies across species, whereas PAR3


Extrinsic tenase Vessel wall injury Intrinsic tenase
Endothelium expression is conserved across humans, mice and rats30.
In human podocytes PAR3 dimerizes with PAR2 follow
ing aPC-mediated PAR3 activation, whereas in murine
TF FVIIa podocytes the cytoprotective effect of aPC depends
Platelets on PAR3/PAR1 heterodimerization30. Murinemesan
FVIIIa
gial cells express PAR1 and PAR230,63 and murine and
Thrombin Platelet activation
human tubular cells express PAR1, PAR2 and EPCR;
FX FIX FXa FIXa Fibrin formation reports regarding the expression of PAR3 and PAR4 are
Feedback inhibition conflicting, possibly reflecting different cell sources34,64,65.
Signalling through PARs
FVIII Integrins (for example M2, 1 and 3) and other
receptors such as apolipoprotein E receptor 2, platelet
Platelet glycoprotein Ib chain and S1P123 also interact with
polyphosphates FVa FII coagulation proteases, but little if anything is known
about these interactions in thekidney.
Considering the wide expression of coagulation
Vessel wall injury regulators, their receptors and potential co-receptors by
renal cells, autocrine and paracrine effects of coagulation
Figure 2 | Initiation, amplification and propagation of coagulation. Upon vessel proteases on renal function seem likely. This concept is
wall injury and/or activation of endothelial cells, tissue factorNature
(TF) is Reviews Nephrology
exposed |to blood supported by the experimental data discussed below.
and binds to FVII or FVIIa, promoting FVII activation or enhancing its catalytic activity. Endothelial dysfunction resulting in altered coagulation
The TFFVIIa (extrinsic tenase) complex activates small amounts of FIX and FX. FXa activation might, therefore, not only promote thrombus
associates with FVa to form the prothrombinase complex, which cleaves FII to generate
formation, but also impact other renal cell types, such as
asmall amount of thrombin. This initiation phase is followed by the amplification phase,
in which thrombin activates cell-surface (predominately platelet) bound FV and FVIII
podocytes and tubular cells6668.
andplatelet-bound FXI. FIXa binds to FVIIIa on negatively charged surfaces
(predominately platelet-derived phospholipids), activating FX (intrinsic tenase) and Role of the glycocalyx
initiating a burst of thrombin generationthe propagation phase. Thrombin has The glycocalyx regulates both capillary permeability
pleotropic functions including feedback inhibition, fibrin formation, platelet activation and activation of coagulation69,70. Although glycocalyx
and signalling through protease activated receptors. a, activated; F, factor. function remains incompletely characterized (reflect
ing largely unmet analytical challenges), important
insights have been gained for the endothelial glycocalyx,
Intriguingly protein C and protein C inhibitor (PCI), including that of glomerular endothelial cells70.
which were initially thought to be specific to the liver, are The glycocalyx consists of covalently linked struc
also expressed in renal cells55. Moreover, renal proteinC tures such as membrane-bound glycoproteins (for
expression is reduced in mice with systemic inflamma example, syndecan1 and thrombomodulin) and
tion or diabetes55. PCI co-localizes with urokinase in attached negatively charged glycosaminoglycans71. The
proximal tubular cells (PTCs) and urokinasePCI com membrane-bound, multifunctional, anticoagulant pro
plexes are readily detectable in urine56. Proinflammatory tein thrombomodulin contains a chondroitin sulphate
stimuli induce glomerular factor V expression, whereas side chain, to which glycosaminoglycans are attached71.
ischaemiareperfusion injury (IRI) induces tubular This loosely attached coat consists of secreted proteo
fibrinogen expression 53,5759. Although endogenous glycans, including perlecan and hyaluronan. The glyco
expression of coagulation regulators such as TFPI, calyx has important roles in the transduction of shear
protein C, PCI, and factor V within renal cell types has stress, regulation of leucocyteendothelial cell inter
been demonstrated, their pathophysiological relevance actions, selective permeability, growth-factor binding,
in regulating local coagulation and protease signal and complement and coagulation regulation71,72. Some of
ling in kidney disease remains unknown and warrants the endothelium and plasma-derived soluble molecules
additionalinvestigation. bound to the glycocalyx are important regulators of the
Direct effects of coagulation proteases such as fac glomerular filtration barrier and/or the coagulation sys
tor IIa or factor Xa on glomerular cells have long been tem (for example vascular endothelial growth factor A
known, implying receptor-dependent modulation of [VEGFA], protein C and antithrombin)7173.
intracellular signalling pathways53,60. A physiologi Coagulation regulators that bind to the glycocalyx
cal function of PARs was first demonstrated in 1991 include thrombin, antithrombin, heparin cofactor2 and
(REF.61); renal PAR1 activation conveys vasoconstriction TFPI. Heparan sulphates provide a scaffold for the inter
and reduces glomerular filtration rate (GFR), whereas action of antithrombin with procoagulant enzymes (such
PAR2 activation causes vasodilation, partially reverses as thrombin, factor Xa and factor IXa), thus enhancing
the vasoconstriction induced by PAR1 agonists or angi the anticoagulant effect several thousand-fold. TFPI is
otensinII, and increases GFR62. Renal endothelial cells thought to bind to the glycocalyx via heparan sulphates,
express PAR1, PAR2 and EPCR, whereas podocytes but other proteins could also be involved74. Uptake and
express PAR3, but lack EPCR, demonstrating the pres degradation of TFPIfactor Xa complexes also depends
ence of distinct cell-specific receptor complexes30 (FIG.3). on heparan sulphates in the glycocalyx 75. Heparin
Intriguingly, podocyte-specific expression of PAR1, cofactor2 is a thrombin-specific protease inhibitor,

98 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 2 | Activating and disarming (inactivating) proteases of PARs15,16,2628


PAR Activating proteases Disarming proteases
Coagulation Other Coagulation Other
PAR1 Thrombin, aPC, EPCR, Granzyme A, trypsin IV, KLK1, KLK4, Plasmin KLK1, KLK14, ADAM17,
activated factor X, KLK6, KLK14, MMP1, elastase, protease 3, trypsin,
plasmin cathepsin G, proatherocytin*, Pen C 13 cathepsin G, elastase
PAR2 Tissue factor, activated Trypsin, mast cell tryptase, acrosin, Plasmin Protease 3, calpain,
factor X, factor VIIa matripatase/MTSP1, HAT, trypsin IV, cathepsin G, elastase
granzyme A, TMPRSS2, chitinase, KLK2,
KLK4, KLK5, KLK6, KLK14, bacterial
gingipains, Der P1, P2, P3||, Pen C 13
PAR3 Thrombin, aPC, NA NA Cathespin G, elastase
activated factor X
PAR4 Thrombin, plasmin, Trypsin, cathepsin G, trypsin IV, MASP1, NA NA
activated factor X bacterial gingipains, KLK1, KLK14
Functional relevance of only a few proteases and corresponding PAR signalling has been demonstrated in renal physiology and
pathophysiology. ADAM17, disintegrin and metalloproteinase domain-containing protein 17; aPC, activated protein C; EPCR,
endothelial protein C receptor; HAT, human airway trypsin-like protease; KLK, kallikrein-related peptidase; MASP1,
mannan-binding lectin serine protease 1; MMP1, matrix metalloproteinase1; NA, not applicable. *Isolated from snake venom.

Mould allergen. From Streptomyces griseus. ||House dust mite cysteine (P1) or serine (P2, P3) proteases.

which is activated by dermatan sulphate in the glyco with advanced CKD, plasma levels of soluble thrombo
calyx 71. Likewise PCI inhibition of aPC requires binding modulin are elevated, reflecting endothelial dysfunc
to heparan sulphate71. All of these anticoagulant mol tion, whereas levels of aPC are reduced, providing a
ecules within the glycocalyx contribute to the thrombo potential link between CKD, endothelial dysfunction
resistant nature of healthy endothelium. Quantitative and hypercoagulability 87,8991. These clinical observa
and qualitative alterations of the glycocalyx, for example tions provided the rationale for mechanistic studies of
following toxic stimuli such as IRI or hyperglycaemia, the endothelial thrombomodulinprotein C system
modulate the bioavailability of these proteins69,76. Loss in renal disease. Following kidney transplantation,
of endothelial glycocalyx during acute hyperglycaemia markers of hypercoagulability and endothelial dys
coincides with endothelial dysfunction and coagulation function (such as soluble thrombomodulin and EPCR)
activation invivo69. Importantly, damage to the endothe normalize, suggesting that impaired renal function is
lial glycoc alyx has been demonstrated in patients causally related to hypercoagulability, potentially via
with type1 diabetes mellitus, the severity of which is endothelialdysfunction9294.
increased in the presence of microalbuminuria77. These In addition to hypercoagulability, fibrin clots in
studies highlight the crucial pathophysiological rele patients with end-stage renal disease (ESRD) are mark
vance of the endothelial glycocalyx. Further investiga edly altered and characterized by a reduced porous
tion is warranted to gain insight into the mechanistic structure, resistance to fibrinolysis and increased overall
relevance of glycocalyx components in the regulation fibre thickness95,96. These characteristics are associated
of coagulation a ctivation and glomerularpermeability. with increased mortality and with a pro-inflammatory
state, but not with azotaemia95,96. The increased fre
Modulation of haemostasis in renal disease quencies of cardiovascular disease and cardiovascular
Thrombotic disease is a significant morbidity of renal death in patients with CKD have been postulated to
diseases, especially in the settings of nephrotic syndrome be partly accounted for by coagulation activation and
and haemodialysis78,79. The likelihood of thrombosis altered blood clot structure, but causality has not yet
is also heightened in other kidney diseases, includ been established.
ing diabetic nephropathy, hypertensive nephropathy,
glomerulonephritis, interstitial nephritis and polycystic Effect of anticoagulation on renal function
kidney disease78. In general the mechanisms underlying In acute renal diseases, such as newly-diagnosed
this increased risk remain ill-defined8086. nephrotic syndrome, hypercoagulability is an impor
In CKD, hypercoagulability is associated with mark tant co-morbidity 9799, but prophylactic anticoagulation
ers of endothelial dysfunction (for example, increased remains controversial100,101. A similar situation exists
levels of soluble thrombomodulin and changes in for chronic renal diseases. In the context of diabetic
flow-mediated dilatation) and inflammation (such nephropathy, researchers have evaluated whether hepa
as IL6)87,88. Factor VIII and von Willebrand factor rins or other glycosaminoglycans improve renal func
have been reported to contribute to venous thrombo tion102,103. In general these studies did not address the role
embolism in patients with CKD, but both of these of altered coagulation protease activity or signalling, but
coagulation regulators are acute phase reactant pro rather evaluated whether glycosaminoglycans can recon
teins, hence this observation might simply reflect an stitute negative charges within the glomerular filtration
association of endothelial dysfunction (and secondary barrier a concept that remains unproven104. Direct
hypercoagulability) with inflammation84. In patients effects of anticoagulants on kidney homeostasis and

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 99

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

Procoagulant, proinammatory
be dependent on PAR1 signalling in renal cells116,119.
Anticoagulant, anti-inammatory
Inthis context the protective effect of low but sustained
Thrombin / aPC thrombin activation observed in diabetic nephropathy,
and the cytoprotective effect of low thrombin concentra
N-terminus tions invitro on endothelial cells, podocytes and tubular
PAR1 (3860) LDPR SFLLR NPNDKYEPFWEDEEKNESGL
cells are noteworthy, as they support the hypothesis that
PAR3 (3457) TLPIK TFR GAPPNSFEEFPFSALE a low level of coagulation activation might be nephro
PAR protective.65,120 Such protective effects might be lost
following supratherapeutic anticoagulation or direct
thrombininhibition.
Cross-talk
The effects of coagulation protease inhibitors have
been investigated in several preclinical studies121,122.
Receptor level Notably, in a mouse model of sickle cell disease, the
factor Xa inhibitor rivaroxaban had a greater anti-
Intracellular inflammatory effect (characterized by a reduction in
level PAR1, PAR2, PAR4 PAR3 PAR1, PAR2 PAR3 non-PAR plasma levels of IL6) than the thrombin inhibitor
receptors
dabigatran, despite a less potent anticoagulant dose,
Heterodimerization GPCR-dependent or
Classic GPCR signal Altered GPCR signal GPCR-independent signal indicating specific differences between these inhib
itors in relation to blood clotting and cytoprotective
Figure 3 | Potential mechanisms of PAR activation by thrombin and aPC.
Nature Reviews An
| Nephrology effects123. Mechanistically, direct suppression of the
example scheme of protease activated receptors (PARs) and the Nterminal sequences thrombin-induced negative-feedback system by inhib
ofhuman PAR1 and PAR3 depicting distinct cleavage sites for thrombin and activated iting thrombomodulinthrombin-mediated protein C
protein C (aPC; arrows). The qualitatively distinct signalling mechanisms of thrombin
activation might underlie these differential effects of
andaPC can be attributed to the distinct proteolytic activation mechanisms of the
Gprotein-coupled receptor (GPCR) Nterminus, resulting in protease-specific factor Xa versus thrombin inhibition124,125. In addition
tetheredligands (shown in red for thrombin and blue for aPC) or induction of distinct to their essential role in blood clotting, both factor Xa
protease-specific signalling complexes. PAR3 is not considered to be signalling and thrombin can elicit multiple cellular effects via
competent and the function of the tethered ligands remains incompletely resolved. PARs and their co-receptors. Thrombin activates PAR1,
Activation of PARs might elicit protease-specific classical GPCR signalling by activation whereas factorXa can also signal via PAR2, directly
of individual PAR receptors (that is protomers) or ligand-specific PARPAR heterodimers. or togetherwith the TFfactor VIIa complex 122,123,126.
In addition, coagulation-protease-dependent signalling might engage non-PAR Consistent withthese observations, inhibition of fac
receptors, enabling biased signalling and thus leading to signalling diversity. Other torXa but not of thrombin suppressed expression of the
proteases can cleave PARs at different sites, for example matrix metalloproteinase1 proinflammatory cytokine IL6, emphasizing the bene
cleaves PAR1 at Asp39 and neutrophil elastase cleaves PAR1 at Leu45.
ficial effects of protease-specific inhibition in a mouse
model of sickle cell disease123. Determining whether
renal disease progression remain undefined in acute and direct anticoagulants differ in regard to their efficacy
chronic renal diseases, but given the wide use of these and safety in the context of renal function and disease
therapies, such effects might have important clinical will be of great clinical interest. Furthermore, elucidat
implications. The first study to evaluate the effect of the ing the molecular mechanisms of coagulation protease
novel direct coagulation protease inhibitor rivaroxaban signalling in renal cells might lead to new therapies that
on renal function is now underway in patients with CKD targetthe involved receptors and pathways without
and atrial fibrillation105. increasing the risk of glomerularhaemorrhage.
Both pharmacological anticoagulation and natu
rally occurring anticoagulants have been demonstrated Coagulation proteases in renal disease
to positively affect nephron health106115. By contrast, Acute glomerular disease
supratherapeutic anticoagulation has been associated The implications of glomerular coagulation activity and
with accelerated loss of GFR116118. These effects might signalling have been thoroughly studied in the setting of
potentially result in a vicious cycle, as many anti rapidly progressive glomerulonephritis (RPGN). A key
coagulants are dependent on renal clearance and might feature of RPGN is the formation of a fibrin matrix in the
accumulate with worsening renal function. Although Bowman capsule, which forms the basis for the patho
glomerular haemorrhage and tubular obstruction, par gnomonic crescent-shaped scar seen in kidney biopsy
ticularly in patients with pre-existing CKD, has been samples. Fibrinogen-deficient mice are partially resist
proposed to be causative, gross haematuria has only ant to antibody-mediated (anti-glomerular b asement
rarely been observed in these patients118. Other mech membrane) RPGN127.
anisms of anticoagulant-mediated glomerular injury Despite beneficial effects in pre-clinical and early
should, therefore, be considered. non-randomized clinical observational studies, therapies
Interestingly, in rats PAR1 inhibition and the direct aimed at reducing fibrin deposition have not been suc
thrombin inhibitor dabigatran have similar detrimental cessfully translated into the clinic128,129. This failure might
effects on renal function to supratherapeutic anticoagu reflect the inherent haemorrhage risk of such therapies
lation with warfarin116,119. These findings demonstrate and the now well-established multifaceted functions of
that these adverse effects are not specific to warfarin the coagulation system, which are partially independ
and as rat platelets do not express PAR1 might ent of fibrin formation. Once formed, fibrin is regulated

100 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

that TF is the major invivo initiator of fibrin deposition


and renal failure in RPGN47. Specific thrombin inhibi
tion using the naturally occurring anticoagulant hirudin
ameliorated crescent formation, glomerular Tcell and
macrophage infiltration, and serum creatinine eleva
tion, emphasizing the pathophysiological relevance of
Endothelial cells
Human and mouse: thrombin in this disease model137. Similarly, factor Xa
PAR1, PAR2, EPCR, thrombomodulin inhibition ameliorated the disease phenotype in rats
with experimental mesangioproliferative glomerulo
nephritis in a manner that was suggested to be depend
ent on mesangial cell PAR2 signalling 53,106,138. Indeed,
signalling by procoagulant proteases that are activated
Mesangal cells
Human: PAR1, PAR2, TF downstream of TF might contribute to RPGN independ
Mouse: PAR1, PAR2 ent of fibrin formation and dissolution. Thus, PAR1 or
PAR2 knockout diminished progression of experimen
Podocytes tal glomerulonephritis in mice, demonstrating that PAR
Human: PAR2, PAR3, PAR4, TF signalling drives this immune-cell-mediated glomerular
Mouse: PAR1, PAR3, PAR4
disease137,139. Although these studies clearly establish a
pathogenic role of coagulation proteases and PARs in
acute glomerular diseases, the dynamics of receptor
Tubular epithelial cells
Human and mouse: activation and interaction remain unknown.
PAR1, PAR2, PAR4, EPCR In a mouse model of thrombotic microangiopathy
(TMA) induced by various murine monoclonal and
Figure 4 | Expression of coagulation protease receptorsNature in renal cells. Protease
Reviews | Nephrology human antiphospholipid antibodies, glomerular TF
activated receptor (PAR) 2 is expressed on all types of human renal cells but is not expression was markedly enhanced and TF seemed
expressed on murine podocytes, whereas PAR3 expression is predominantly restricted to to be a common mediator of glomerular injury 43.
podocytes. Endothelial protein C receptor (EPCR), which promotes protein C activation Following exposure to these antibodies, which had anti-
and activated protein C signalling, is expressed by glomerular endothelial and tubular phospholipid activity, both TF expression and com
epithelial cells. Tissue factor (TF) is expressed by podocytes and mesangial cells in
plement activation (that is, glomerular C3 deposition)
humans, whether it is expressed on these cells in other species remains unproven.
Invarious disease models (for example sepsis and diabetic nephropathy) TF expression were increased; C5a receptor (C5aR) deficiency partially
isinduced, but expression of thrombomodulin and EPCR is impaired. protected against these effects. Importantly, genetic or
pharmacological (using pravastatin) reduction of TF
expression prevented renal injury irrespective of whether
by the plasmin system (FIG.5). Mice with plasminogen the antibody-induced glomerular injury, and increased
or tPA deficiency are prone to exacerbated RPGN129. TF expression occurred via a complement-dependent
Interestingly, deficiency of urokinase plasminogen acti or complement-independent pathway 43. These studies
vator (uPA), or its receptor uPAR, reduced glomerular established a causal relationship of TF in regulation of
macrophage infiltration, but did not significantly alter the acute glomerular injury, which is partially potentiated by
RPGN disease course130. The proinflammatory cytokine complement activation.
IL1, which is released from infiltrating mononuclear Induction of TF expression following C5aR sig
cells in experimental RPGN, upregulates glomerular nalling has also been shown in human and murine
expression of tPA, but not uPA, and downregulates the neutrophils140,141 and the functional interaction of TF
tPA inhibitor plasminogen activator inhibitor1 (PAI1) and complement might reflect the well-established
in cultured mesangial cells131133. Mice deficient in PAI1 communication between these systems142. In addition
are partially protected from experimental RPGN, whereas to inducing TF expression, the complement C5b7 com
those that transgenically overexpress PAI1 have exac plex might activate TF by oxidizing its disulphide bond
erbated disease, suggesting that compensatory changes (via protein-disulphide-isomerase-dependent thiol-
in tPA and PAI1 expression are inadequate to control disulphide exchange), resulting in optimalTF folding
experimental RPGN134. A function of the fibrinolytic and oxidation and thus contributing to TFactivation7,143.
system nephroprotection is further supported by a study Intriguingly, thrombomodulin, the functional opponent
in which pharmacological inhibition of TAFI conveyed of TF in coagulation activation, dampens complement
protective effects in mice with acute glomerulonephritis activation144 and thrombomodulin polymorphisms
by promotingfibrinolysis135. are associated with atypical haemolytic uraemic syn
During RPGN, coagulation activation and subse drome (HUS)145. Thrombomodulin dampens comple
quent fibrin deposition seem to be dependent on TF ment activation via activation of TAFI (via removal
expressed by infiltrating mononuclear cells47,136. An of carboxyterminal arginines from C3a andC5a) and
association of increased TF expression with glomerular through a poorly defined mechanism involving the
fibrin deposition and renal failure has been shown in lectin-like domain144,146. Deficiency of the lectin-like
human RPGN and in rabbit models47. Administration domain of thrombomodulin worsened Shiga-toxin-
of anti-TF antibody in rabbits ameliorated these changes induced experimental HUS in mice; these mice showed
without affecting macrophage infiltration, indicating enhanced renal impairment and increased glomerular

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 101

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Acute glomerular injury b Acute tubular injury

Inammation IRI

Inammatory cell Glomerular cell Inammatory cell Tubular cell


recruitment activation? recruitment activation?

TF expression TF expression

FXa Fibrin Thrombin Fibrin

Hirudin Glomerular
Heparin PAR2 Crescent formation Excessive Moderate
injury
AT

Thrombin Fibrinolysis Tubular Tissue Fibrinolysis


PAR1 injury
injury
+
PAR1 Plasmin Plasmin B15-42
TGF-

Complement activation TM ECM ICAM-1



PAI-1
PAR1 Tubular Tissue
Heparin and/or TM TAFIa RAAS aPC EPCR YB-1 repair
repair

Figure 5 | Coagulation regulators in acute kidney injury. a | In acute glomerular injury, inflammation induces tissue
factor (TF) expression on inflammatory cells recruited into the glomeruli or potentially on glomerular Nature Reviews
cells | Nephrology
themselves,
resulting in coagulation activation. FVa and the FXa complex assemble on these cells and enhance thrombin generation.
FXa and thrombin induce glomerular cell dysfunction via protease activated receptor (PAR) 2 and PAR1, respectively.
Thrombin signalling via PAR1 might involve transactivation of PAR2. Increased fibrin deposition contributes to the
formation of glomerular crescents, which is inhibited by plasmin-mediated fibrinolysis. Inhibition of plasmin by
plasminogen activator inhibitor1 (PAI1), which might be induced via the reninangiotensinaldosterone system (RAAS)
or activated thrombin activatable fibrinolysis inhibitor (TAFIa), abolishes this effect. b | In acute tubular injury
inflammation, for example in the context of ischaemiareperfusion injury (IRI), induces TF expression on inflammatory
cells or potentially on tubular epithelial cells themselves, triggering coagulation activation within the tubular
compartment. Thrombin signalling via PAR1 leads to transforming growth factor (TGF)-dependent tissue remodelling
and tubular injury, whereas activated protein C (aPC) signalling via PAR1endothelial protein C receptor (EPCR) inhibits
TGF-dependent tubular fibrosis and preserves expression of YB1 by restricting its ubiquitin-dependent degradation,
thereby preventing tubular injury. Excess fibrin formation induces tubular injury, whereas moderate fibrin generation
triggers fibrinolysis, leading to extracellular matrix (ECM) degradation and generation of the fibrin-derived peptide
B1542, which blocks the interaction of fibrin with intercellular adhesion molecule1 (ICAM1). Moderate fibrin
deposition, therefore, contributes to renal recovery. Green inhibitory arrows indicate that inhibition promotes repair; red
inhibitory arrow indicates that inhibition promotes injury. a, activated; AT, antithrombin; F, factor; TM, thrombomodulin.

complement and fibrin deposition compared with activate PAR1 by proteolytic cleavage and might also sig
wild-type controls 147. Whether thrombomodulin nal via PAR4 (TABLE2). The implications of PAI1 altera
functionally interacts with TF via complement regula tions on plasmin-mediated cell signalling in the kidney
tion in the context of g lomerular disease has not yet remain unexplored in the contexts of RAAS-mediated
beenaddressed. renal fibrosis and glomerulonephritis151,152.
The reninangiotensinaldosterone system (RAAS) Nonspecific coagulation inhibition with hepa
is integrally involved in the progression of many kid rin or antithrombin reduces proteinuria in the puro
ney diseases148. Signalling through angiotensin II type1 mycin aminonucleoside and adriamycin experimental
and angiotensin IV receptors upregulates expression models of nephrotic syndrome153156. Unfortunately,
of PAI1148. In addition, aldosterone stimulates renal owing to the nonspecific nature of these inhibitors it
PAI1 expression and PAI1deficient animals are remains unclear which protease(s) are involved in per
protected from aldosterone-induced glomerulosclero sistent proteinuria. Circulating protease activity from
sis149,150. These effects are thought to occur via amelio patients with nephrotic syndrome has, however, been
ration of PAI1mediated suppression of plasmin and demonstrated to injure cultured podocytes in what
matrix-metalloproteinase-mediated degradation of seems to be a PAR1dependent manner, implicat
extracellular matrix proteins that are involved in renal ing PAR1 cleaving proteases as p romising targets for
fibrosis148. Interestingly, plasmin can either disarm or furtherinvestigation151,157.

102 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

Acute tubular disease protease signalling in acute kidney diseases likely extend
The effects of coagulation proteases on renal tubular beyond those described in this Review. For example,
health have been addressed in several studies. In the con coagulation is known to be activated in complement
text of IRI, limited fibrin formation is required for tissue (HUS) and non-complement (TTP) mediated thrombotic
repair; complete fibrinogen deficiency severely impairs microangiopathy (TMA)43. Notably thrombomodulin has
renal function, whereas reduced fibrinogen expres been shown to regulate complement in atypical HUS145.
sion protects against ischaemic injury in heterozygous Renal injury in the setting of TMA is thought to be medi
fibrinogen-knockout mice (FIG.4)57,158. The fibrin-derived ated, at least in part, by ischaemic effects secondary to
peptide B1542 protects kidneys from IRI, potentially small artery thrombosis. However, the implications of
by competitively blocking binding of fibrin to integrins, downstream cellular injury mediated by activated coag
ICAM1 or VEcadherin, thus altering cellular signalling ulation protease signalling remains unexplored in these
and reducing apoptosis57,159,160. Aprotective effect of the settings. Anticoagulation with heparin might modulate
B1542 peptide is apparent even if applied post-injury complement system activity, reflecting the close inter
and might involve preservation of endothelial and vascu action of the complement and coagulation systems and
lar integrity 57,161. The observation that urinary fibrinogen suggesting that it might be possible to pharmaceutically
might be a marker of acute k idney injury (AKI) is also of target and utilize these systems inAKI167.
potential clinical relevance162.
The coagulation system clearly also has functions that Chronic kidney disease
are independent of fibrin formation in AKI. Thus, defi CKD (defined as estimated GFR <60ml/min/1.73m2) is
ciencies in TF or PAR1 are protective in renal IRI and an increasing medical problem, which afflicts ~26mil
impaired thrombomodulin-dependent protein C acti lion people in the USA alone. As outlined above,
vation worsens disease outcomes34,52. In renal IRI, aPC hypercoagulability in CKD is well documented, but
independent of its anticoagulant function sustains the underlying mechanisms remain poorly defined.
intracellular levels of the cold-shock protein YB1 by Hypercoagulability might be linked to altered clearance
restricting its ubiquitination and degradation through or inactivation of coagulation regulators, uraemic toxins,
PAR1 EPCR signalling in tubular cells34. These stud electrolyte and acidbase abnormalities and/or endothe
ies have identified a new mechanism through which lial dysfunction. The loss of renal thrombomodulin
coagulation proteases modulate cellular function inAKI. expression and other procoagulant and proinflamma
Additional roles for coagulation signalling in tubular tory regulators during ageing might depend on NF-B
epithelial cells might influence renal outcomes. A prom signalling, consistent with a role of this transcription
inent example is the discovery that plasmin activates factor in the regulation of coagulation regulators32,168.
sodium and calcium channels of PTCs during nephrotic- Fibrinolysis regulators are also activated in CKD169 and
range proteinuria163,164. These protease-mediated altera both impaired fibrinolytic activity and hyperfibrinolysis
tions in ion channel function might at least partly explain have been described88,170,171. Plasma levels of PAI1 are
oedema formation in nephrotic syndrome. Furthermore, increased in patients with CKD, including those with
thrombin stimulates proliferation and pro-inflammatory diabetic nephropathy 18. To date, most existing studies of
responses in cultured human PTCs64,165. These responses the role of haemostatic regulators in CKD have focused
can be recapitulated with PAR2 activating peptides but on diabetic nephropathy a leading cause ofESRD.
not with other PAR agonists; this finding is interesting
because thrombin is not known to cleave PAR216,151. Diabetic nephropathy
When cleaved, however, the PAR1tethered ligand might Fibrin deposition is increased in diabetic glomeruli
transactivate PAR2 that is heterodimerized to PAR1, and is associated with glomerular extracellular matrix
potentially explaining these data27. These responses to accumulation (FIG.6)172,173. Thrombin-induced mesangial
thrombin seem to be TGF-dependent. PAR1 cleav TGF expression and an increased peripheral blood
age by aPC bound to EPCR seems to counteract the PAI1 to tPA ratio in diabetic patients might contrib
effect of thrombin by downregulating TGF-mediated ute to these changes174,175. The function of the endothe
production of profibrotic extracellular matrix proteins lial thrombomodulinproteinC system is impaired
byPTCs64,65. in patients with diabetic nephropathy, as reflected by
The fibrinolytic system might also be involved in increased plasma levels of soluble thrombomodulin and
tubular injury. Mice that overexpress PAI1 develop sig reduced levels of aPC176,177.
nificantly aggravated renal fibrosis in response to ureteral Animal studies have been instrumental in decipher
obstruction, compared with wild-type mice166. These ing the role of coagulation proteases in diabetic nephro
effects might be mediated through a downstream reduc pathy. TF expression is increased in diabetic mice (db/db
tion in extracellular matrix degradation, as proposed to and streptozotocin models), along with increased expres
occur for the RAAS in the glomerulus148. The effects of sion of PAR2 (db/db mice show a transient increase at
PAI1 modulation on downstream plasmin PAR1 20weeks) and factorV51,58. Increased TF expression has
signalling have not yet been investigated. been observed in tubular cells and glomerular parietal
Together the available data suggest that coagulation cells51, whereas glomerular thrombomodulin expression
derangements and egress of coagulation proteins into the is reduced in diabetic mice33. Studies involving genetic
renal interstitium and/or urinary space might influence modification of the thrombomodulinproteinC system
disease progression. The implications of coagulation established that loss of thrombomodulin-dependent

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 103

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Chronic glomerular injury b Chronic tubular injury


Subclinical inammation Subclinical inammation
Cellular dysfunction Cellular dysfunction

Inammatory cell Glomerular cell Inammatory cell Tubular cell


recruitment activation recruitment activation?

Altered expression of
coagulation regulators
TF expression

Complement
TF/FV/PAR2 Thrombomodulin activation

Thrombin
Thrombin aPC

EC:EPCR/PAR1
PAI-1/tPA Fibrin TGF- P:PAR3 Tubular Fibrin
injury

ECM p66Shc
Bax
Fibrinolysis TM
Glomerulosclerosis
ROS
Mitochrondrial Plasmin TAFIa
Glomerular dysfunction
injury

Figure 6 | Coagulation regulators in chronic kidney injury. a | In chronic kidney disease, subclinical inflammation
Nature Reviews | Nephrology
induces expression of tissue factor (TF) on glomerular cells or potentially on inflammatory cells recruited into the
glomeruli, thus triggering coagulation activation. FXa and thrombin induce glomerular cell dysfunction via protease
activated receptor (PAR) signalling, fibrin induction and extracellular matrix (ECM) deposition. In chronic diabetic kidney
disease, thrombomodulin (TM)dependent proteinC activation is impaired resulting in exacerbated glucose toxicity and
glomerular cell dysfunction. Impaired protein C activation also triggers mitochondrial localization of Bax and p66Shc,
resulting in mitochondrial dysfunction in glomerular cells. Reconstitution of activated protein C (aPC) signalling, for
example by exogenous administration, restores cellular function via endothelial protein C receptor (EPCR)PAR1
signalling in endothelial cells and PAR3mediated signalling in podocytes, thus preventing diabetic nephropathy.
Independent of aPC generation, thrombomodulin inhibits complement activation via its lectin-like domain and
ameliorates diabetic nephropathy. b | Chronic inflammation triggers fibrin generation in the tubulointerstitial
compartment, contributing to tubular injury. Plasmin-mediated degradation of fibrin and ECM inhibits this process,
butthis tissue-protective mechanism is inhibited by thrombin-mediated activation of thrombin activable fibrinolysis
inhibitor (TAFI). Green inhibitory arrow indicates that inhibition promotes repair; red inhibitory arrow indicates that
inhibition promotes injury. a, activated; EC, endothelial cells; F, factor; P, podocytes; PAI1 plasminogen activator
inhibitor1; ROS, reactive oxygen species; TGF, transforming growth factor ; tPA, tissue-type plasminogen activator.

proteinC activation aggravates diabetic nephropathy, Association of the PAR3/PAR2 dimer with caveolin1
whereas compensation for impaired proteinC activation is also required for the cytoprotective effect of aPC
protects against the disease33. Exogenous application invitro 30. aPC reduces caveolin1Tyr14 phosphory
of aPC likewise ameliorated diabetic nephropathy in lation in a time-dependent manner and this aPC-
mice, demonstrating that the underlying mechanism mediated dephosphorylation enables the dissociation
can, in principle, be pharmacologically targeted178,179. of caveolin1 from PAR2 and PAR3, effectively inhibit
As the nephroprotective effect of aPC is independent ing podocyte apoptosis30. The mechanistic importance
of its anticoagulant properties, the possibility exists of PARcaveolin interactions has likewise been estab
that the underlying mechanism could be utilized with lished in endothelial cells, in which compartmentaliza
out interfering with the haemostatic system30,179. aPC tion of PAR1 into caveolar microdomains determines
engages distinct receptor complexes (PAR1EPCR on the barrier protective versus barrier disruptive effects
endothelial cells and PAR3 in podocytes) to ameliorate of aPC and thrombin, respectively 20. Whether these
glucose toxicity. In podocytes aPC signalling via PAR3 observations made in HUVECderived EA.hy926
transactivates PAR2 in humans or PAR1 in mice30. Given cellscan be extrapolated to glomerular endothelial cells
the species-specific expression of PARs in podocytes, remains unknown. Based on the available data, however,
results obtained in mice, for example when evaluating it can be concluded that proteases convey their effects
new therapeutic strategies or the risk of adverse effects, in renal cells through cell-specific PAR signalosomes180.
cannot generally be extrapolated tohumans. The characterization of these signalosomes and the

104 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

downstream signalling events might facilitate the identi with increased plasmin and matrix metalloproteinase2
fication of small molecules that target these protease-in activity, but seems to also involve signalling via uPAR
duced cytoprotectivemechanisms. and the ERK/MAPK pathway 187190. Notably loss or gain
In diabetic nephropathy, aPC epigenetically supresses of PAI1 expression resulted in tubulointerstitial injury
the redox enzyme p66Shc and thereby limits sustained in aged non-diabetic mice, illustrating a crucial role of
generation of mitochondrial reactive oxygen species179. PAI1 in renal homeostasis in health and disease187.
These effects are dependent on thrombomodulin- Similar to acute renal diseases (such as glomerulo
mediated proteinC activation on endothelial cells and nephritis, haemolyticuraemic syndrome and other
aPC/PAR3 signalling in podocytes, establishing an complement-mediated diseases), in diabetic nephro
endothelial-to-podocyte crosstalk mechanism179. This pathy thrombomodulin suppresses complement activa
coagulation-protease-dependent crosstalk complements tion in addition to providing a functional switch between
the VEGF and angiopoietin crosstalk at the glomerular thrombin and aPC signalling. The lectin-like domain of
filtration barrier, which is controlled by podocytes181. In thrombomodulin inhibits complement activation through
the diabetic milieu this coagulation-protease-dependent a poorly characterized mechanism in glucose-stressed
crosstalk is disturbed, impairing the glomerular filtra endothelial cells and podocytes and ameliorates diabetic
tion barrier and podocyte function, thus contributing nephropathy in mice independent of blood coagulation191.
to diabeticglomerulopathy.
Consistent with the invitro concentration-dependent Tubulointerstitial fibrosis
effects of thrombin in tubular cells and podocytes, mice In diabetic patients on dialysis, levels of TAFI and acti
with low but sustained thrombin generation owing to vated TAFI are elevated, indicating impaired fibrino
the factor V Leiden (FVL) mutation are partially pro lysis192. A potential role of activated TAFI in tubular
tected from diabetic nephropathy 120. This effect is lost injury is supported by its induction by glucose-stressed
following anticoagulation with the direct and irreversi tubular cells invitro, resulting in reduced plasmin activ
ble thrombin inhibitor hirudin, indicating a protective ity and an increase in extracellular matrix 193. Moreover,
role of thrombin generation at low levels120. Whether low inhibition of TAFI reduced tubulointerstitial and glom
thrombin concentrations directly convey cytoprotective erular fibrosis following subtotal nephrectomy in mice194,
signalling, as suggested by invitro studies, or whether consistent with observations made in TAFI-deficient
indirect effects such as protein C activation convey mice challenged with chronic glomerulonephritis195.
cytoprotection, remains unknown120,182. Importantly, the Regarding the possible involvement of PARs in tubu
FVL mutation is associated with reduced albuminuria in lointerstitial fibrosis, the increased expression of tubular
diabetic and pre-diabetic individuals, demonstrating the PAR2 in human chronic renal disease (IgA nephropathy)
relevance of this phenomenon in patients120,183. and in the murine model of unilateral ureteral obstruc
The apparent nephroprotective effect of low throm tion is noteworthy 196,197. Congruent with its profibrino
bin concentrations might relate to controversial data genic role in other organs (such as the liver and lung),
obtained from the use of anticoagulants in animal PAR2 promotes early renal tubular injury and fibrosis
models of diabetic nephropathy. Initially, heparins were in the murine unilateral ureteral obstruction model197.
thought to replenish negatively charged glycosamino Analyses of human PTCs invitro suggest that the profi
glycans in the glomerular filtration barrier, thus restoring brinogenic effects of PAR2 are mechanistically linked to
its function184,185. This possibility cannot be completely TGF and EGFR transactivation197. The roleof coagula
refuted, but other mechanisms for the nephroprotective tion proteases in PAR-activation, modulation ofreceptor
effect should be considered. Efficient thrombin inhib transactivation or tubulointerstitial fibrosis in chronic
ition is expected to impair both protein C activation and renal disease models remains unresolved.
direct cytoprotective signalling. Furthermore, thrombin
and factor Xa activate different receptors; hence anti Renal transplantation
coagulation with direct thrombin or factor Xa inhibitors Given the roles of coagulation proteases in chronic renal
might have distinct consequences. Thus, fondaparinux, diseases their impact on transplant nephropathy is of
aselective factor Xa inhibitor, conveys partially protec potential interest. Enhanced PAI1 expression, which
tive effects in db/db mice (that is, it has a minor effect on seems to be reduced by rapamycin treatment, might
albuminuria and slightly reduces glomerular size), con promote chronic allograft nephropathy 198,199. Increased
trasting with the disadvantageous effects of hirudin58,120. vascular expression of TF has been observed in chronic,
Considering the increasing availability and use of small but not in acute allograft nephropathy 200. In addition,
molecule anticoagulants, and the recent introduction of acute ciclosporin-induced nephropathy was associated
the first PAR-antagonist for clinical use, studies deci with increased TF immunoreactivity in the tubular
phering the effects of these new drugs on glomerular brush border, suggesting that TF expression might be a
function and renal disease areneeded. useful diagnostic marker for this condition200. The rele
As diabetic nephropathy is characterized by extra vance of this observation remains to be established. In
cellular matrix accumulation, a number of studies models ofxenotransplantation, inhibition of coagula
have addressed the role of fibrinolysis regulators in tion activation during the ischaemic period with either
this disease. Expression of PAI1 is induced in diabetic antithrombin or aPC markedly reduced chronic kidney
nephropathy 186. Loss of PAI1 expression in mice reduces graft fibrosis201. This observation implies that excessive
extracellular matrix accumulation, which is associated coagulation activation during the ischaemic period

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 105

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS

harms the transplant and that exvivo anticoagulation clinically approved PAR antagonist (voraxapar), which
of the transplant would be beneficial without increasing might alter coagulation signalling in the kidney, should
the risk of haemorrhage in the recipient. Decipheringthe include renal end points to provide additional insight into
mechanism of such long-lasting effects, including their renal benefits and/or adverse effects. Such clini
thepotential involvement of PARs199, might lead to novel cal studies have been initiated and data might become
clinical interventions. available in the nearfuture105.

Unresolved questions Conclusions


Although studies evaluating the role of the haemostatic Considerable advances have been made in understand
system in renal disease have provided novel pathophysio ing the physiology and pathophysiology of coagula
logical insights, a number of questions remain. For tion proteases, their regulators and their receptors in
example the kinetics of altered expression of coagulation renal disease. Knowledge now exists of the functions
regulators and their receptors in renal disease remain of the haemostatic system conveyed via intravascular
largely undefined. Whether coagulation proteases gain blood clotting, fibrin formation and fibrinolysis, but
access to extravascular renal cell compartments through also through direct modulation of renal cell function
controlled mechanisms and the role of locally expressed through protease-dependent signalling. The identifi
coagulation regulators also need to be defined. Some cation of cell-specific, PAR-dependent signalosomes in
insights into cell-type-specific signalosomes have been renal tissue has provided new pathogenic concepts and,
gained, but further studies are warranted to characterize importantly, identified potential therapeutic targets. The
the interaction of PARs with other receptors, such as Toll- translation of these new insights into the clinic, however,
like receptors and receptor tyrosine kinases (for example requires further mechanistic studies. In addition, sur
EGFR or VEGFR), in the context of renal diseases28,197. veillance of renal function in clinical studies evaluating
Such insights might enable the design of kidney-specific new anticoagulants or therapeutics targeting PARs is
therapeutic approaches. Studies addressing these ques required to gain insights into the function of coagula
tions will likely provide new insights into the regulation tion proteases and their receptors in humans. Based on
of intracellular signalling pathways, tissue remodelling the available data, the possibility exists, but remains to be
and sterile inflammation in renal disease. While these explored, that targeting specific coagulation proteases,
questions are being experimentally addressed, clini their receptors or underlying mechanisms might enable
cal studies evaluating novel anticoagulants or the first clinicians to improve the treatment of renal disease.

1. Hoess, A., Watson, S., Siber, G.R. & Liddington, R. 13. Smith, S.A. etal. Polyphosphate exerts differential inflammatory disease. Br. J.Pharmacol. 171,
Crystal structure of an endotoxin-neutralizing protein effects on blood clotting, depending on polymer size. 11801194 (2014).
from the horseshoe crab, Limulus anti-LPS factor, at Blood 116, 43534359 (2010). 27. Lin, H., Liu, A.P., Smith, T.H. & Trejo, J. Cofactoring
1.5 resolution. EMBO J. 12, 33513356 (1993). 14. Jordan, R.E., Oosta, G.M., Gardner, W.T. & and dimerization of proteinase-activated receptors.
2. Esmon, C.T. The interactions between inflammation Rosenberg, R.D. The kinetics of hemostatic enzyme Pharmacol. Rev. 65, 11981213 (2013).
and coagulation. Br. J.Haematol. 131, 417430 antithrombin interactions in the presence of low 28. Gieseler, F., Ungefroren, H., Settmacher, U.,
(2005). molecular weight heparin. J.Biol. Chem. 255, Hollenberg, M.D. & Kaufmann, R. Proteinase-
3. Benmira, S., Banda, Z.K. & Bhattacharya, V. Old 1008110090 (1980). activated receptors (PARs) focus on receptor
versus new anticoagulants: focus on pharmacology. 15. Griffin, J.H., Zlokovic, B.V. & Mosnier, L.O. receptorinteractions and their physiological and
Recent Pat. Cardiovasc. Drug Discov. 5, 120137 Activatedprotein C: biased for translation. Blood 125, pathophysiological impact. Cell Commun. Signal. 11,
(2010). 28982907 (2015). 86 (2013).
4. Ramachandran, R., Noorbakhsh, F., Defea, K. & 16. Adams, M.N. etal. Structure, function and 29. Coughlin, S.R. Thrombin signalling and protease-
Hollenberg, M.D. Targeting proteinase-activated pathophysiology of protease activated receptors. activated receptors. Nature 407, 258264 (2000).
receptors: therapeutic potential and challenges. Pharmacol. Ther. 130, 248282 (2011). 30. Madhusudhan, T. etal. Cytoprotective signaling by
Nat.Rev. Drug Discov. 11, 6986 (2012). 17. Zhang, G. & Eddy, A.A. Urokinase and its receptors in activated protein C requires protease-activated
5. Adams, R.L. & Bird, R.J. Review article: coagulation chronic kidney disease. Front. Biosci. 13, 54625478 receptor3 in podocytes. Blood 119, 874883 (2012).
cascade and therapeutics update: relevance to (2008). 31. Ruf, W., Disse, J., Carneiro-Lobo, T.C., Yokota, N. &
nephrology. Part 1: Overview of coagulation, 18. Malgorzewicz, S., Skrzypczak-Jankun, E. & Jankun, J. Schaffner, F. Tissue factor and cell signalling in cancer
thrombophilias and history of anticoagulants. Plasminogen activator inhibitor1 in kidney pathology progression and thrombosis. J.Thromb. Haemost. 9,
Nephrology (Carlton) 14, 462470 (2009). (review). Int. J.Mol. Med. 31, 503510 (2013). S306S315 (2011).
6. Eisenreich, A. & Rauch, U. Regulation and differential 19. Luft, F.C. uPAR signaling is under par for the podocyte 32. Song, D., Ye, X., Xu, H. & Liu, S.F. Activation of
role of the tissue factor isoforms in cardiovascular course. J.Mol. Med. (Berl.) 90, 13571359 (2012). endothelial intrinsic NFB pathway impairs protein C
biology. Trends Cardiovasc. Med. 20, 199203 20. Russo, A., Soh, U.J., Paing, M.M., Arora, P. & anticoagulation mechanism and promotes coagulation
(2010). Trejo,J. Caveolae are required for protease-selective in endotoxemic mice. Blood 114, 25212529 (2009).
7. Monroe, D.M. & Key, N.S. The tissue factorfactor signaling by protease-activated receptor1. Proc. Natl 33. Isermann, B. etal. Activated protein C protects
VIIa complex: procoagulant activity, regulation, and Acad. Sci. USA 106, 63936397 (2009). against diabetic nephropathy by inhibiting endothelial
multitasking. J.Thromb. Haemost. 5, 10971105 21. Elmariah, S.B., Reddy, V.B. & Lerner, E.A. Cathepsin and podocyte apoptosis. Nat. Med. 13, 13491358
(2007). S. signals via PAR2 and generates a novel tethered (2007).
8. Bjorkqvist, J., Nickel, K.F., Stavrou, E. & Renne, T. ligand receptor agonist. PLoS ONE 9, e99702 (2014). 34. Dong, W. etal. The protease aPC ameliorates renal
Invivo activation and functions of the protease factor 22. Boire, A. etal. PAR1 is a matrix metalloprotease1 I/R-injury by restricting YB1 ubiquitination. J.Am.
XII. Thromb. Haemost. 112, 868875 (2014). receptor that promotes invasion and tumorigenesis Soc. Nephrol. http://dx.doi.org/10.1681/
9. Kenne, E. & Renne, T. Factor XII: a drug target for ofbreast cancer cells. Cell 120, 303313 (2005). ASN.2014080846.
safeinterference with thrombosis and inflammation. 23. Weiler, H. Multiple receptor-mediated functions of 35. von Drygalski, A., Furlan-Freguia, C., Ruf, W.,
Drug Discov. Today 19, 14591464 (2014). activated protein C. Hamostaseologie 31, 185195 Griffin,J.H. & Mosnier, L.O. Organ-specific protection
10. Griffin, J.H. Role of surface in surface-dependent (2011). against lipopolysaccharide-induced vascular leak is
activation of Hageman factor (blood coagulation 24. Shahzad, K. & Isermann, B. The evolving plasticity dependent on the endothelial protein C receptor.
factorXII). Proc. Natl Acad. Sci. USA 75, 19982002 ofcoagulation protease-dependent cytoprotective Arterioscler. Thromb. Vasc. Biol. 33, 769776 (2013).
(1978). signalling. Hamostaseologie 31, 179184 (2011). 36. Weiler, H. etal. Characterization of a mouse model for
11. Smith, S.A. etal. Polyphosphate modulates blood 25. Bouwens, E.A., Stavenuiter, F. & Mosnier, L.O. thrombomodulin deficiency. Arterioscler. Thromb.
coagulation and fibrinolysis. Proc. Natl Acad. Sci. USA Mechanisms of anticoagulant and cytoprotective Vasc. Biol. 21, 15311537 (2001).
103, 903908 (2006). actions of the protein C pathway. J.Thromb. Haemost. 37. Apostolopoulos, J. etal. The cytoplasmic domain of
12. Muller, F. etal. Platelet polyphosphates are 11, S242S253 (2013). tissue factor in macrophages augments cutaneous
proinflammatory and procoagulant mediators invivo. 26. Hollenberg, M.D. etal. Biased signalling and delayed-type hypersensitivity. J.Leukoc. Biol. 83,
Cell 139, 11431156 (2009). proteinase-activated receptors (PARs): targeting 902911 (2008).

106 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS
38. Apostolopoulos, J., Moussa, L. & Tipping, P.G. 62. Gui, Y., Loutzenhiser, R. & Hollenberg, M.D. 86. Zwaginga, J.J., Koomans, H.A., Sixma, J.J. &
Thecytoplasmic domain of tissue factor restricts Bidirectional regulation of renal hemodynamics by Rabelink, T.J. Thrombus formation and plateletvessel
physiological albuminuria and pathological proteinuria activation of PAR1 and PAR2 in isolated perfused rat wall interaction in the nephrotic syndrome under flow
associated with glomerulonephritis in mice. Nephron kidney. Am. J.Physiol. Renal Physiol. 285, F95F104 conditions. J.Clin. Invest. 93, 204211 (1994).
Exp. Nephrol. 116, e72e83 (2010). (2003). 87. Dubin, R. etal. Kidney function and multiple
39. Sharma, L. etal. The cytoplasmic domain of tissue 63. DAndrea, M.R. etal. Characterization of protease- hemostatic markers: cross sectional associations
factor contributes to leukocyte recruitment and death activated receptor2 immunoreactivity in normal inthe multi-ethnic study of atherosclerosis. BMC
in endotoxemia. Am. J.Pathol. 165, 331340 human tissues. J.Histochem. Cytochem. 46, Nephrol. 12, 3 (2011).
(2004). 157164 (1998). 88. Adams, M.J., Irish, A.B., Watts, G.F., Oostryck, R.
40. Yang, Y.H. etal. Reduction in arthritis severity and 64. Vesey, D.A. etal. Thrombin stimulates &Dogra, G.K. Hypercoagulability in chronic kidney
modulation of immune function in tissue factor proinflammatory and proliferative responses in disease is associated with coagulation activation but
cytoplasmic domain mutant mice. Am. J.Pathol. 164, primary cultures of human proximal tubule cells. not endothelial function. Thromb. Res. 123, 374380
109117 (2004). Kidney Int. 67, 13151329 (2005). (2008).
41. Cunningham, M.A., Kitching, A.R., Tipping, P.G. & 65. Bae, J.S., Kim, I.S. & Rezaie, A.R. Thrombin down- 89. Adams, G.N. etal. Murine prolylcarboxypeptidase
Holdsworth, S.R. Fibrin independent proinflammatory regulates the TGFmediated synthesis of collagen depletion induces vascular dysfunction with
effects of tissue factor in experimental crescentic and fibronectin by human proximal tubule epithelial hypertension and faster arterial thrombosis. Blood
glomerulonephritis. Kidney Int. 66, 647654 (2004). cells through the EPCR-dependent activation of PAR1. 117, 39293937 (2011).
42. Mackman, N., Sawdey, M.S., Keeton, M.R. & J.Cell Physiol. 225, 233239 (2010). 90. Bao, Y.S. etal. Characterization of soluble
Loskutoff, D.J. Murine tissue factor gene expression 66. Molitoris, B.A. & Sutton, T.A. Endothelial injury and thrombomodulin levels in patients with stage 35
invivo. Tissue and cell specificity and regulation by dysfunction: role in the extension phase of acute renal chronic kidney disease. Biomarkers 19, 275280
lipopolysaccharide. Am. J.Pathol. 143, 7684 failure. Kidney Int. 66, 496499 (2004). (2014).
(1993). 67. Nawroth, P.P. & Isermann, B. Mechanisms of diabetic 91. Faioni, E.M. etal. Low levels of the anticoagulant
43. Seshan, S.V. etal. Role of tissue factor in a mouse nephropathyold buddies and newcomers part 2. activity of protein C in patients with chronic renal
model of thrombotic microangiopathy induced by Exp. Clin. Endocrinol. Diabetes 118, 667672 (2010). insufficiency: an inhibitor of protein C is present in
antiphospholipid antibodies. Blood 114, 16751683 68. Nawroth, P.P. & Isermann, B. Mechanisms of diabetic uremic plasma. Thromb. Haemost. 66, 420425
(2009). nephropathyold buddies and newcomers part 1. (1991).
44. Osterud, B. & Bjorklid, E. Sources of tissue factor. Exp. Clin. Endocrinol. Diabetes 118, 571576 (2010). 92. Nampoory, M.R. etal. Hypercoagulability, a serious
Semin. Thromb. Hemost. 32, 1123 (2006). 69. Nieuwdorp, M. etal. Loss of endothelial glycocalyx problem in patients with ESRD on maintenance
45. Drake, T.A., Morrissey, J.H. & Edgington, T.S. during acute hyperglycemia coincides with endothelial hemodialysis, and its correction after kidney
Selective cellular expression of tissue factor in human dysfunction and coagulation activation invivo. transplantation. Am. J.Kidney Dis. 42, 797805
tissues. Implications for disorders of hemostasis and Diabetes 55, 480486 (2006). (2003).
thrombosis. Am. J.Pathol. 134, 10871097 (1989). 70. Haraldsson, B., Nystrom, J. & Deen, W.M. Properties 93. Ghisdal, L. etal. Thrombophilic factors in stage V
46. Flossel, C., Luther, T., Muller, M., Albrecht, S. & of the glomerular barrier and mechanisms of chronic kidney disease patients are largely corrected
Kasper, M. Immunohistochemical detection of tissue proteinuria. Physiol. Rev. 88, 451487 (2008). by renal transplantation. Nephrol. Dial. Transplant.
factor (TF) on paraffin sections of routinely fixed 71. Xu, D. & Esko, J.D. Demystifying heparan sulfate 26, 27002705 (2011).
human tissue. Histochemistry 101, 449453 (1994). protein interactions. Annu. Rev. Biochem. 83, 94. Keven, K. etal. Soluble endothelial cell protein C
47. Erlich, J.H., Holdsworth, S.R. & Tipping, P.G. Tissue 129157 (2014). receptor and thrombomodulin levels after renal
factor initiates glomerular fibrin deposition and 72. Salmon, A.H. & Satchell, S.C. Endothelial glycocalyx transplantation. Int. Urol. Nephrol 42, 10931098
promotes major histocompatibility complex class II dysfunction in disease: albuminuria and increased (2010).
expression in crescentic glomerulonephritis. microvascular permeability. J.Pathol. 226, 562574 95. Undas, A., Kolarz, M., Kopec, G. & Tracz, W. Altered
Am.J.Pathol. 150, 873880 (1997). (2012). fibrin clot properties in patients on long-term
48. Ettelaie, C., Su, S., Li, C. & Collier, M.E. Tissue factor- 73. Reitsma, S., Slaaf, D.W., Vink, H., haemodialysis: relation to cardiovascular mortality.
containing microparticles released from mesangial vanZandvoort,M.A. & oude Egbrink, M.G. The Nephrol. Dial. Transplant. 23, 20102015 (2008).
cells in response to high glucose and AGE induce tube endothelial glycocalyx: composition, functions, and 96. Sjoland, J.A. etal. Fibrin clot structure in patients
formation in microvascular cells. Microvasc. Res. 76, visualization. Pflugers Arch. 454, 345359 (2007). with end-stage renal disease. Thromb. Haemost. 98,
152160 (2008). 74. Kato, H. Regulation of functions of vascular wall cells 339345 (2007).
49. Wiggins, R.C., Njoku, N. & Sedor, J.R. Tissue factor by tissue factor pathway inhibitor: basic and clinical 97. Kerlin, B.A., Ayoob, R. & Smoyer, W.E. Epidemiology
production by cultured rat mesangial cells. Stimulation aspects. Arterioscler. Thromb. Vasc. Biol. 22, and pathophysiology of nephrotic syndrome-
by TNF and lipopolysaccharide. Kidney Int. 37, 539548 (2002). associated thromboembolic disease. Clin. J.Am. Soc.
12811285 (1990). 75. Ho, G., Broze, G.J. Jr & Schwartz, A.L. Role of Nephrol 7, 513520 (2012).
50. Ono, T. etal. Coagulation process proceeds on heparan sulfate proteoglycans in the uptake and 98. Kerlin, B.A. etal. Disease severity correlates with
cultured human mesangial cells via expression of degradation of tissue factor pathway inhibitor thrombotic capacity in experimental nephrotic
factor V. Kidney Int. 60, 10091017 (2001). coagulation factor Xa complexes. J.Biol. Chem. 272, syndrome. J.Am. Soc. Nephrol. http://dx.doi.org/
51. Sommeijer, D.W. etal. Renal tissue factor expression 1683816844 (1997). 10.1681/ASN.2014111097.
is increased in streptozotocin-induced diabetic mice. 76. van Golen, R.F. etal. The mechanisms and 99. Loscalzo, J. Venous thrombosis in the nephrotic
Nephron Exp. Nephrol. 101, e86e94 (2005). physiological relevance of glycocalyx degradation in syndrome. N.Engl. J.Med. 368, 956958 (2013).
52. Sevastos, J. etal. Tissue factor deficiency and PAR1 hepatic ischemia/reperfusion injury. Antioxid. Redox 100. Glassock, R.J. Prophylactic anticoagulation in
deficiency are protective against renal ischemia Signal. 21, 10981118 (2014). nephrotic syndrome: a clinical conundrum. J.Am. Soc.
reperfusion injury. Blood 109, 577583 (2007). 77. Nieuwdorp, M. etal. Endothelial glycocalyx damage Nephrol. 18, 22212225 (2007).
53. Nomura, K. etal. Roles of coagulation pathway coincides with microalbuminuria in type1 diabetes. 101. Lee, T. etal. Personalized prophylactic anticoagulation
andfactor Xa in rat mesangioproliferative Diabetes 55, 11271132 (2006). decision analysis in patients with membranous
glomerulonephritis. Lab. Invest. 87, 150160 78. Christiansen, C.F. etal. Kidney disease and risk of nephropathy. Kidney Int. 85, 14121420 (2014).
(2007). venous thromboembolism: a nationwide population- 102. Gambaro, G. & van der Woude, F.J.
54. Cunningham, M.A. etal. Tissue factor pathway based case-control study. J.Thromb. Haemost. 12, Glycosaminoglycans: use in treatment of diabetic
inhibitor expression in human crescentic 14491454 (2014). nephropathy. J.Am. Soc. Nephrol. 11, 359368
glomerulonephritis. Kidney Int. 55, 13111318 79. Kerlin, B.A., Smoyer, W.E., Tsai, J. & Boulet, S.L. (2000).
(1999). Healthcare burden of venous thromboembolism in 103. Lewis, E.J. & Xu, X. Abnormal glomerular permeability
55. Yamamoto, K. & Loskutoff, D.J. Extrahepatic childhood chronic renal diseases. Pediatr. Nephrol. 30, characteristics in diabetic nephropathy: implications
expression and regulation of protein C in the mouse. 829837 (2014). for the therapeutic use of low-molecular weight
Am. J.Pathol. 153, 547555 (1998). 80. Burton, J.O. etal. Elevated levels of procoagulant heparin. Diabetes Care 31, S202S207 (2008).
56. Song, Z. etal. Intracellular localization of protein C plasma microvesicles in dialysis patients. PLoS ONE 8, 104. Li, J., Wu, H.M., Zhang, L., Zhu, B. & Dong, B.R.
inhibitor (PCI) and urinary plasminogen activator in e72663 (2013). Heparin and related substances for preventing
renal tubular epithelial cells from humans and human 81. Hrafnkelsdottir, T., Ottosson, P., Gudnason, T., diabetic kidney disease. Cochrane Database Syst. Rev.
PCI gene transgenic mice. Histochem. Cell Biol. 128, Samuelsson, O. & Jern, S. Impaired endothelial CD005631 (2010).
293300 (2007). release of tissue-type plasminogen activator in 105. Suzuki, M. etal. Rationale and design of the efficacy
57. Sorensen-Zender, I. etal. Role of fibrinogen in acute patients with chronic kidney disease and hypertension. ofrivaroxaban on renal function in patients with non-
ischemic kidney injury. Am. J.Physiol. Renal Physiol. Hypertension 44, 300304 (2004). valvular atrial fibrillation and chronic kidney disease:
305, F777F785 (2013). 82. Jeong, J.C., Kim, J.E., Ryu, J.W., Joo, K.W. & the XNOAC study. Int. J.Cardiol. 188, 5253 (2015).
58. Sumi, A. etal. Roles of coagulation pathway and factor Kim,H.K. Plasma haemostatic potential of 106. Floege, J., Eng, E., Young, B.A., Couser, W.G. &
Xa in the progression of diabetic nephropathy in db/db haemodialysis patients assessed by thrombin Johnson, R.J. Heparin suppresses mesangial cell
mice. Biol. Pharm. Bull. 34, 824830 (2011). generation assay: hypercoagulability in patients proliferation and matrix expansion in experimental
59. Tillet, S. etal. Kidney graft outcome using an anti-Xa withvascular access thrombosis. Thromb. Res. 132, mesangioproliferative glomerulonephritis. Kidney Int.
therapeutic strategy in an experimental model of 604609 (2013). 43, 369380 (1993).
severe ischaemia-reperfusion injury. Br. J.Surg. 102, 83. Kourtzelis, I. etal. Complement anaphylatoxin C5a 107. van der Pijl, J.W. etal. Danaparoid sodium lowers
132142 (2015). contributes to hemodialysis-associated thrombosis. proteinuria in diabetic nephropathy. J.Am. Soc.
60. Xu, Y. etal. Constitutive expression and modulation of Blood 116, 631639 (2010). Nephrol. 8, 456462 (1997).
the functional thrombin receptor in the human kidney. 84. Ocak, G. etal. Role of hemostatic factors on the risk 108. Ikeguchi, H. etal. Effects of human soluble
Am. J.Pathol. 146, 101110 (1995). ofvenous thrombosis in people with impaired kidney thrombomodulin on experimental glomerulonephritis.
61. Vu, T.K., Hung., D.T., Wheaton, V.I. & Coughlin, S.R. function. Circulation 129, 683691 (2014). Kidney Int. 61, 490501 (2002).
Molecular cloning of a functional thrombin receptor 85. Schlegel, N. Thromboembolic risks and complications 109. Matsuyama, M. etal. Tissue factor antisense
reveals a novel proteolytic mechanism of receptor in nephrotic children. Semin. Thromb. Hemost. 23, oligonucleotides prevent renal ischemia-reperfusion
activation. Cell 64, 10571068 (1991). 271280 (1997). injury. Transplantation 76, 786791 (2003).

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 107

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS
110. Gupta, A. etal. Activated protein C ameliorates LPS- 134. Kitching, A.R. etal. Plasminogen activator inhibitor1 159. Devarajan, P. Update on mechanisms of ischemic
induced acute kidney injury and downregulates renal is a significant determinant of renal injury in acute kidney injury. J.Am. Soc. Nephrol. 17,
INOS and angiotensin 2. Am. J.Physiol. Renal experimental crescentic glomerulonephritis. 15031520 (2006).
Physiol. 293, F245F254 (2007). J.Am.Soc. Nephrol. 14, 14871495 (2003). 160. Krishnamoorthy, A. etal. Fibrinogen -derived B1542
111. Ozaki, T. etal. Intrarenal administration of 135. Muto, Y., Suzuki, K., Iida, H. & Ishii, H. EF6265, peptide protects against kidney ischemia/reperfusion
recombinant human soluble thrombomodulin anovel plasma carboxypeptidase B inhibitor, injury. Blood 118, 19341942 (2011).
ameliorates ischaemic acute renal failure. Nephrol. protectsagainst renal dysfunction in rat thrombotic 161. Urbschat, A. etal. The small fibrinopeptide B1542 as
Dial. Transplant. 23, 110119 (2008). glomerulonephritis through enhancing fibrinolysis. renoprotective agent preserving the endothelial and
112. Sharfuddin, A.A. etal. Soluble thrombomodulin Nephron Exp. Nephrol. 106, e113e121 (2007). vascular integrity in early ischemia reperfusion injury
protects ischemic kidneys. J.Am. Soc. Nephrol. 20, 136. Erlich, J.H. etal. Renal expression of tissue factor in the mouse kidney. PLoS ONE 9, e84432 (2014).
524534 (2009). pathway inhibitor and evidence for a role in crescentic 162. Hoffmann, D. etal. Fibrinogen excretion in the urine
113. Thuillier, R. etal. Thrombin inhibition during kidney glomerulonephritis in rabbits. J.Clin. Invest. 98, and immunoreactivity in the kidney serves as a
ischemia-reperfusion reduces chronic graft 325335 (1996). translational biomarker for acute kidney injury.
inflammation and tubular atrophy. Transplantation 90, 137. Cunningham, M.A. etal. Protease-activated receptor1 Am.J.Pathol. 181, 818828 (2012).
612621 (2010). mediates thrombin-dependent, cell-mediated renal 163. Svenningsen, P. etal. Plasmin in nephrotic urine
114. Abdel-Bakky, M.S., Hammad, M.A., Walker, L.A. & inflammation in crescentic glomerulonephritis. activates the epithelial sodium channel. J.Am. Soc.
Ashfaq, M.K. Silencing of tissue factor by antisense J.Exp. Med. 191, 455462 (2000). Nephrol. 20, 299310 (2009).
deoxyoligonucleotide prevents monocrotaline/LPS renal 138. Tanaka, M. etal. Role of coagulation factor Xa and 164. Tudpor, K. etal. Urinary plasmin inhibits TRPV5 in
injury in mice. Arch. Toxicol. 85, 12451256 (2011). protease-activated receptor 2 in human mesangial nephrotic-range proteinuria. J.Am. Soc. Nephrol. 23,
115. Rajashekhar, G. etal. Soluble thrombomodulin cellproliferation. Kidney Int. 67, 21232133 (2005). 18241834 (2012).
reduces inflammation and prevents microalbuminuria 139. Moussa, L., Apostolopoulos, J., Davenport, P., 165. Grandaliano, G. etal. Regenerative and
induced by chronic endothelial activation in transgenic Tchongue, J. & Tipping, P.G. Protease-activated proinflammatory effects of thrombin on human
mice. Am. J.Physiol. Renal Physiol. 302, F703F712 receptor2 augments experimental crescentic proximal tubular cells. J.Am. Soc. Nephrol. 11,
(2012). glomerulonephritis. Am. J.Pathol. 171, 800808 10161025 (2000).
116. Ryan, M. etal. Warfarin-related nephropathy is the tip (2007). 166. Matsuo, S. etal. Multifunctionality of PAI1 in
of the iceberg: direct thrombin inhibitor dabigatran 140. Redecha, P. etal. Tissue factor: a link between C5a fibrogenesis: evidence from obstructive nephropathy
induces glomerular hemorrhage with acute kidney and neutrophil activation in antiphospholipid antibody in PAI1overexpressing mice. Kidney Int. 67,
injury in rats. Nephrol. Dial. Transplant. 29, induced fetal injury. Blood 110, 24232431 (2007). 22212238 (2005).
22282234 (2014). 141. Ritis, K. etal. A novel C5a receptor-tissue factor 167. Zaferani, A. etal. Heparin/heparan sulphate
117. van Blijderveen, J.C. etal. Overanticoagulation is cross-talk in neutrophils links innate immunity to interactions with complementa possible target
associated with renal function decline. J.Nephrol. 26, coagulation pathways. J.Immunol. 177, 47944802 forreduction of renal function loss? Nephrol. Dial.
691698 (2013). (2006). Transplant. 29, 515522 (2014).
118. Brodsky, S.V. etal. Warfarin-related nephropathy 142. Amara, U. etal. Molecular intercommunication 168. Wiggins, J.E. etal. NFB promotes inflammation,
occurs in patients with and without chronic kidney between the complement and coagulation systems. coagulation, and fibrosis in the aging glomerulus.
disease and is associated with an increased mortality J.Immunol. 185, 56285636 (2010). J.Am. Soc. Nephrol. 21, 587597 (2010).
rate. Kidney Int. 80, 181189 (2011). 143. Langer, F. etal. Rapid activation of monocyte tissue 169. Pawlak, K., Ulazka, B., Mysliwiec, M. & Pawlak, D.
119. Arachiche, A., de la Fuente, M. & Nieman, M.T. factor by antithymocyte globulin is dependent on Vascular endothelial growth factor and uPA/suPAR
Platelet specific promoters are insufficient to express complement and protein disulfide isomerase. Blood system in early and advanced chronic kidney
protease activated receptor 1 (PAR1) transgene in 121, 23242335 (2013). diseasepatients: a new link between angiogenesis
mouse platelets. PLoS ONE 9, e97724 (2014). 144. Conway, E.M. Thrombomodulin and its role in andhyperfibrinolysis? Transl. Res. 160, 346354
120. Wang, H. etal. Low but sustained coagulation inflammation. Semin. Immunopathol. 34, 107125 (2012).
activation ameliorates glucose-induced podocyte (2012). 170. Ito, T., Niwa, T. & Matsui, E. Fibrinolytic activity in
apoptosis: protective effect of factor V Leiden in 145. Delvaeye, M. etal. Thrombomodulin mutations in renal disease. Clin. Chim. Acta 36, 145151 (1972).
diabetic nephropathy. Blood 117, 52315242 (2011). atypical hemolytic-uremic syndrome. N.Engl. J.Med. 171. Mezzano, D. etal. Hemostatic disorder of uremia: the
121. Borensztajn, K. & Spek, C.A. Blood coagulation factor 361, 345357 (2009). platelet defect, main determinant of the prolonged
Xa as an emerging drug target. Expert Opin. Ther. 146. Weiler, H. & Isermann, B.H. Thrombomodulin. bleeding time, is correlated with indices of activation
Targets 15, 341349 (2011). J.Thromb. Haemost. 1, 15151524 (2003). of coagulation and fibrinolysis. Thromb. Haemost. 76,
122. Ruf, W. FXa takes center stage in vascular 147. Zoja, C. etal. Lack of the lectin-like domain of 312321 (1996).
inflammation. Blood 123, 16301631 (2014). thrombomodulin worsens Shiga toxin-associated 172. Farquhar, A., MacDonald, M.K. & Ireland, J.T. The
123. Sparkenbaugh, E.M. etal. Differential contribution of hemolytic uremic syndrome in mice. J.Immunol. 189, role of fibrin deposition in diabetic glomerulosclerosis:
FXa and thrombin to vascular inflammation in a mouse 36613668 (2012). a light, electron and immunofluorescence microscopy
model of sickle cell disease. Blood 123, 17471756 148. Fogo, A.B. The role of angiotensin II and plasminogen study. J.Clin. Pathol. 25, 657667 (1972).
(2014). activator inhibitor1 in progressive glomerulosclerosis. 173. Nakagawa, T. etal. Diabetic endothelial nitric oxide
124. Furugohri, T. etal. Different antithrombotic properties Am. J.Kidney Dis. 35, 179188 (2000). synthase knockout mice develop advanced diabetic
of factor Xa inhibitor and thrombin inhibitor in rat 149. Brown, N.J. etal. Aldosterone modulates plasminogen nephropathy. J.Am. Soc. Nephrol. 18, 539550
thrombosis models. Eur. J.Pharmacol. 514, 3542 activator inhibitor1 and glomerulosclerosis invivo. (2007).
(2005). Kidney Int. 58, 12191227 (2000). 174. Yamabe, H. etal. Thrombin stimulates production
125. Furugohri, T., Sugiyama, N., Morishima, Y. & 150. Ma, J. etal. Plasminogen activator inhibitor1 oftransforming growth factor- by cultured human
Shibano,T. Antithrombin-independent thrombin deficiency protects against aldosterone-induced mesangial cells. Nephrol. Dial. Transplant. 12,
inhibitors, but not direct factor Xa inhibitors, enhance glomerular injury. Kidney Int. 69, 10641072 438442 (1997).
thrombin generation in plasma through inhibition of (2006). 175. Ito, Y. etal. Kinetics of connective tissue growth
thrombinthrombomodulinprotein C system. 151. Soh, U.J., Dores, M.R., Chen, B. & Trejo, J. factorexpression during experimental proliferative
Thromb.Haemost. 106, 10761083 (2011). Signaltransduction by protease-activated receptors. glomerulonephritis. J.Am. Soc. Nephrol. 12,
126. Rana, S., Yang, L., Hassanian, S.M. & Rezaie, A.R. Br. J.Pharmacol. 160, 191203 (2010). 472484 (2001).
Determinants of the specificity of protease-activated 152. Kuliopulos, A. etal. Plasmin desensitization of the 176. Mormile, A. etal. Physiological inhibitors of blood
receptors 1 and 2 signaling by factor Xa and thrombin. PAR1 thrombin receptor: kinetics, sites of truncation, coagulation and prothrombin fragment F 1+2 in
J.Cell. Biochem. 113, 977984 (2012). and implications for thrombolytic therapy. type2 diabetic patients with normoalbuminuria and
127. Drew, A.F. etal. Crescentic glomerulonephritis is Biochemistry 38, 45724585 (1999). incipient nephropathy. Acta Diabetol. 33, 241245
diminished in fibrinogen-deficient mice. Am. J.Physiol. 153. Baroni, E.A., Costa, R.S., da Silva, C.G. & (1996).
Renal Physiol. 281, F1157F1163 (2001). Coimbra,T.M. Heparin treatment reduces glomerular 177. Matsumoto, K. etal. Inverse correlation between
128. Hariharan, S., Pollak, V.E., Kant, K.S., Weiss, M.A. injury in rats with adriamycin-induced nephropathy activated protein C generation and carotid
&Wadhwa, N.K. Diffuse proliferative lupus nephritis: but does not modify tubulointerstitial damage or the atherosclerosis in Type2 diabetic patients.
long-term observations in patients treated with renal production of transforming growth factor-. Diabet.Med. 24, 13221328 (2007).
ancrod. Clin. Nephrol. 34, 6169 (1990). Nephron 84, 248257 (2000). 178. Gil-Bernabe, P. etal. Exogenous activated protein C
129. Craciun, F.L. etal. Pharmacological and genetic 154. Benchetrit, S. etal. Low molecular weight heparin inhibits the progression of diabetic nephropathy.
depletion of fibrinogen protects from kidney fibrosis. reduces proteinuria and modulates glomerular TNF- J.Thromb. Haemost. 10, 337346 (2012).
Am. J.Physiol. Renal Physiol. 307, F471F484 production in the early phase of adriamycin 179. Bock, F. etal. Activated protein C ameliorates diabetic
(2014). nephropathy. Nephron 87, 155160 (2001). nephropathy by epigenetically inhibiting the redox
130. Kitching, A.R. etal. Plasminogen and plasminogen 155. Diamond, J.R. & Karnovsky, M.J. Nonanticoagulant enzyme p66Shc. Proc. Natl Acad. Sci. USA 110,
activators protect against renal injury in crescentic protective effect of heparin in chronic aminonucleoside 648653 (2013).
glomerulonephritis. J.Exp. Med. 185, 963968 nephrosis. Ren. Physiol. 9, 366374 (1986). 180. Bock, F., Shahzad, K., Vergnolle, N. & Isermann, B.
(1997). 156. Yamashita, J. etal. Protective effects of antithrombin Activated protein C based therapeutic strategies in
131. Arai, K.I. etal. Cytokines: coordinators of immune on puromycin aminonucleoside nephrosis in rats. chronic diseases. Thromb. Haemost. 111, 610617
and inflammatory responses. Annu. Rev. Biochem. 59, Eur.J.Pharmacol. 589, 239244 (2008). (2014).
783836 (1990). 157. Harris, J.J. etal. Active proteases in nephrotic plasma 181. Fu, J., Lee, K., Chuang, P.Y., Liu, Z. & He, J.C.
132. Wilson, H.M., Haites, N.E., Reid, F.J. & Booth, N.A. lead to a podocin-dependent phosphorylation of VASP Glomerular endothelial cell injury and cross talk in
Interleukin1 up-regulates the plasminogen activator/ in podocytes via protease activated receptor1. diabetic kidney disease. Am. J.Physiol. Renal Physiol.
plasmin system in human mesangial cells. Kidney Int. J.Pathol. 229, 660671 (2013). 308, F287F297 (2015).
49, 10971104 (1996). 158. Ajay, A.K., Saikumar, J., Bijol, V. & Vaidya, V.S. 182. Kerlin, B.A. etal. Survival advantage associated with
133. Sedor, J.R., Nakazato, Y. & Konieczkowski, M. Heterozygosity for fibrinogen results in efficient heterozygous factor V Leiden mutation in patients with
Interleukin1 and the mesangial cell. Kidney Int. 41, resolution of kidney ischemia reperfusion injury. severe sepsis and in mouse endotoxemia. Blood 102,
595599 (1992). PLoSONE 7, e45628 (2012). 30853092 (2003).

108 | FEBRUARY 2016 | VOLUME 12 www.nature.com/nrneph

2016 Macmillan Publishers Limited. All rights reserved


REVIEWS
183. Peter, A. etal. Lower plasma creatinine and urine 192. Malyszko, J., Malyszko, J.S., Hryszko, T. & 199. Grandaliano, G. etal. Protease-activated receptor 1
albumin in individuals at increased risk of type2 Mysliwiec,M. Thrombin activatable fibrinolysis and plasminogen activator inhibitor 1 expression in
diabetes with factor v leiden mutation. ISRN inhibitor (TAFI) and markers of endothelial cell injury chronic allograft nephropathy: the role of coagulation
Endocrinol. 2014, 530830 (2014). in dialyzed patients with diabetic nephropathy. and fibrinolysis in renal graft fibrosis. Transplantation
184. Pugliese, F., Mene, P. & Cinotti, G.A. Glomerular Thromb. Haemost. 91, 480486 (2004). 72, 14371443 (2001).
polyanion and control of cell function. Am. J.Nephrol. 193. Atkinson, J.M., Pullen, N. & Johnson, T.S. An 200. Osterholm, C., Veress, B., Simanaitis, M., Hedner, U.
10, S14S18 (1990). inhibitor of thrombin activated fibrinolysis inhibitor &Ekberg, H. Differential expression of tissue factor
185. Gambaro, G. & Kong, N.C. Glycosaminoglycan (TAFI) can reduce extracellular matrix accumulation in (TF) in calcineurin inhibitor-induced nephrotoxicity and
treatment in glomerulonephritis? An interesting an invitro model of glucose induced ECM expansion. rejectionimplications for development of a possible
option to investigate. J.Nephrol. 23, 244252 Matrix Biol. 32, 277287 (2013). diagnostic marker. Transpl. Immunol. 15, 165172
(2010). 194. Atkinson, J.M., Pullen, N., Da Silva-Lodge, M., (2005).
186. Hagiwara, H. etal. Expression of type1 plasminogen Williams, L. & Johnson, T.S. Inhibition of thrombin- 201. Ramackers, W. etal. Recombinant human
activator inhibitor in the kidney of diabetic rat models. activated fibrinolysis inhibitor increases survival in antithrombin prevents xenogenic activation of
Thromb. Res. 111, 301309 (2003). experimental kidney fibrosis. J.Am. Soc. Nephrol. 26, hemostasis in a model of pigtohuman kidney
187. Lassila, M. etal. Plasminogen activator inhibitor1 19251937 (2015). transplantation. Xenotransplantation 21, 367375
production is pathogenetic in experimental murine 195. Bruno, N.E. etal. Immune complex-mediated (2014).
diabetic renal disease. Diabetologia 50, 13151326 glomerulonephritis is ameliorated by thrombin-
(2007). activatable fibrinolysis inhibitor deficiency. Acknowledgements
188. Nicholas, S.B. etal. Plasminogen activator inhibitor1 Thromb.Haemost. 100, 90100 (2008). This work was supported by grants from the Deutsche
deficiency retards diabetic nephropathy. Kidney Int. 196. Grandaliano, G. etal. Protease-activated receptor2 Forschungsgemeinschaft (IS 67/24, IS67/52, SFB 854 to
67, 12971307 (2005). expression in IgA nephropathy: a potential role in B.I.; TH 1789/11 to T.M.), the Stiftung Pathobiochemie und
189. Kenichi, M. etal. Renal synthesis of urokinase type- thepathogenesis of interstitial fibrosis. J.Am. Soc. Molekulare Diagnostik (B.I. and T.M.) and from the National
plasminogen activator, its receptor, and plasminogen Nephrol. 14, 20722083 (2003). I n s t i t u t e s o f H e a l t h ( U 5 4 D K 0 8 3 91 2 0 5 S 1 a n d
activator inhibitor1 in diabetic nephropathy in rats: 197. Chung, H., Ramachandran, R., Hollenberg, M.D. L40DK103299m to B.A.K.).
modulation by angiotensinconvertingenzyme &Muruve, D.A. Proteinase-activated receptor2
inhibitor. J.Lab. Clin. Med. 144, 6977 (2004). transactivation of epidermal growth factor receptor
190. Cheng, H., Chen, C. & Wang, S. Effects of uPA on and transforming growth factor- receptor signaling Author contributions
mesangial matrix changes in the kidney of diabetic pathways contributes to renal fibrosis. J.Biol. Chem. All authors researched the data, made substantial contribu-
rats. Ren. Fail. 36, 13221327 (2014). 288, 3731937331 (2013). tions to discussion of the content, wrote the text and
191. Wang, H. etal. The lectin-like domain of 198. Pontrelli, P. etal. Rapamycin inhibits PAI1 expression reviewed and edited the manuscript before submission.
thrombomodulin ameliorates diabetic glomerulopathy and reduces interstitial fibrosis and glomerulosclerosis
via complement inhibition. Thromb. Haemost. 108, in chronic allograft nephropathy. Transplantation 85, Competing interests statement
11411153 (2012). 125134 (2008). The authors declare no competing interests.

NATURE REVIEWS | NEPHROLOGY VOLUME 12 | FEBRUARY 2016 | 109

2016 Macmillan Publishers Limited. All rights reserved

You might also like