You are on page 1of 12

Matrix metalloproteinases in kidney homeostasis and

diseases
Roderick J. Tan and Youhua Liu
Am J Physiol Renal Physiol 302:F1351-F1361, 2012. First published 4 April 2012;
doi: 10.1152/ajprenal.00037.2012

You might find this additional info useful...

This article cites 154 articles, 57 of which you can access for free at:
http://ajprenal.physiology.org/content/302/11/F1351.full#ref-list-1
This article has been cited by 1 other HighWire-hosted articles:

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


http://ajprenal.physiology.org/content/302/11/F1351#cited-by
Updated information and services including high resolution figures, can be found at:
http://ajprenal.physiology.org/content/302/11/F1351.full
Additional material and information about American Journal of Physiology - Renal Physiology can be
found at:
http://www.the-aps.org/publications/ajprenal

This information is current as of November 18, 2012.

American Journal of Physiology - Renal Physiology publishes original manuscripts on a broad range of subjects
relating to the kidney, urinary tract, and their respective cells and vasculature, as well as to the control of body fluid
volume and composition. It is published 24 times a year (twice monthly) by the American Physiological Society,
9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2012 the American Physiological Society. ESSN:
1522-1466. Visit our website at http://www.the-aps.org/.
Am J Physiol Renal Physiol 302: F1351–F1361, 2012.
First published April 4, 2012; doi:10.1152/ajprenal.00037.2012. Review

Matrix metalloproteinases in kidney homeostasis and diseases


Roderick J. Tan1 and Youhua Liu2
1
Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine and 2Department
of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
Submitted 20 January 2012; accepted in final form 3 April 2012

Tan RJ, Liu Y. Matrix metalloproteinases in kidney homeostasis and diseases.


Am J Physiol Renal Physiol 302: F1351–F1361, 2012. First published April 4,
2012; doi:10.1152/ajprenal.00037.2012.—Matrix metalloproteinases (MMPs) are a

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


family of zinc-dependent endopeptidases that have been increasingly linked to both
normal physiology and abnormal pathology in the kidney. Collectively able to
degrade all components of the extracellular matrix, MMPs were originally thought
to antagonize the development of fibrotic diseases solely through digestion of
excessive matrix. However, increasing evidence has shown that MMPs play a wide
variety of roles in regulating inflammation, epithelial-mesenchymal transition, cell
proliferation, angiogenesis, and apoptosis. We now have robust evidence for MMP
dysregulation in a multitude of renal diseases including acute kidney injury,
diabetic nephropathy, glomerulonephritis, inherited kidney disease, and chronic
allograft nephropathy. The goal of this review is to summarize current findings
regarding the role of MMPs in kidney diseases as well as the mechanisms of action
of this family of proteases.
MMP; fibrosis; epithelial-mesenchymal transition; inflammation; apoptosis

THE MATRIX METALLOPROTEINASES (MMPs) are a large family of proteolytic potential, MMPs are traditionally conceived as
zinc-dependent endopeptidases that are collectively capable of antifibrotic players in the development and progression of
proteolyzing all components of the extracellular matrix (ECM) chronic kidney diseases (CKD), in which tissue fibrosis is the
(15, 99). The first discovery of MMPs was originally reported common outcome. This assumption, however, has been chal-
by Gross and Lapiere (38) when they described collagenase lenged recently, and a new paradigm is emerging that MMPs
activity in metamorphosing tadpoles. Since then, the number of actually play a critical role in the initiation of fibrotic kidney
known MMPs as well as their characterized functions have disorders. Here, we review the biology of MMPs, with empha-
risen dramatically (83, 139). It is now accepted that MMPs sis on their role and potential mechanisms in the development
play a multitude of roles in regulating a diverse array of of kidney diseases.
biological processes such as embryonic development, tissue
homeostasis, tumorigenesis, and organ fibrogenesis (15, 97). Biology of MMPs
Initially thought to degrade only ECM proteins, MMPs are Structure and classification. All MMPs, with some modifi-
increasingly known to be able to cleave a wide variety of cations, share a core set of basic structural domains. As shown
substrates, which range from cell surface receptors and adhe- in Fig. 1, the prototype MMP consists of a prodomain, a
sion molecules to growth factors and cytokines. This broad catalytic domain, a hinge region, and a hemopexin-like domain
spectrum of substrates enables MMPs to be a critical player not (15, 87). As secreted proteins, MMPs are synthesized with a
only in regulating ECM remodeling but also in controlling signal peptide responsible for directing secretion out of the cell.
many cell behaviors such as cell proliferation, migration, The prodomain consists of a conserved cysteine residue that
differentiation, angiogenesis, and apoptosis (37, 83). Over the prevents binding and cleavage of the substrate, while the catalytic
last decade, thanks to the availability of many genetically domain contains a binding site comprised of three histidines that
modified mice (both knockout and transgenic) and specific coordinate with a single zinc and is responsible for the enzymatic
pharmacological inhibitors, our understanding of the biology activity of the protease. MMP-2 and -9 have a series of fibronectin
of MMPs in vivo has substantially evolved and improved. repeats in this domain as well. A flexible hinge region then
Many studies have provided significant insights into the exact separates the catalytic domain from a four-bladed propeller-
roles of specific MMPs in regulating physiological homeostasis shaped hemopexin domain. It is felt that substrate specificity as
and pathological disorders. well as endogenous inhibitor binding are influenced by speci-
MMPs are expressed in both developing and adult kidneys, ficities of each MMP’s catalytic and hemopexin domains (37,
and they are implicated in regulating nephron formation and 139). Some MMPs remain tethered to the cell via transmem-
the pathogenesis of kidney diseases, as nicely summarized brane and cytoplasmic domains at the C terminus. At least one
previously in a comprehensive review (15). In light of their MMP (MMP-23) has a type II transmembrane domain near the
N terminus and also possesses a cysteine-rich region and
Address for reprint requests and other correspondence: Y. Liu, Dept. of immunoglobulin-like domain (87, 139). The crystal structure
Pathology, Univ. of Pittsburgh, S-405 Biomedical Science Tower, 200 Lothrop of active human MMP-1 containing many of the domains
St., Pittsburgh, PA 15261 (e-mail: liuy@upmc.edu). described above is shown in Fig. 2.
http://www.ajprenal.org 1931-857X/12 Copyright © 2012 the American Physiological Society F1351
Review
F1352 MMPs IN KIDNEY DISEASES

Fig. 1. Domain structure of the matrix metalloproteinase (MMP)


family. The MMPs are structurally composed of similar modular
domains. The presence or absence of these domains, along with
their substrate specificity, has been used to classify the MMPs into
several subgroups. S, signal peptide; Pro, prodomain; Cat, catalytic
domain; h, hinge region; F, furin sensitive cleavage site; Fn,
fibronectin-like repeat; II, type II transmembrane domain; G,
glycosylphosphatidylinositol (GPI)-anchor.

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


MMPs are traditionally classified according to their structure epilysin (MMP-28) (83, 95). Substrate specificities are sum-
and/or ECM substrate specificity. The collagenases (MMP-1, marized in Table 1.
MMP-8, and MMP-13) can degrade native collagen and are Transcriptional regulation. Regulation of MMPs at the level
often hypothesized to be antifibrotic. Stromelysins (MMP-3, of gene expression has been reviewed elsewhere (29, 74), and
MMP-10, and MMP-11) share structural similarity with the it involves activating protein (AP)-1 and AP-2, PEA3, NF-␬B,
collagenases but are unable to degrade native collagen. As their Smad, and STAT transcription factors and their cognate bind-
name implies, the gelatinases (MMP-2, MMP-9) cleave dena- ing sites. Of note, some of the MMPs, including MMP-7 and
tured collagen (gelatin) as well as type IV collagen in basement MMP-9, are regulated through canonical Wnt/␤-catenin sig-
membranes. Matrilysins (MMP-7, MMP-26) degrade ECM naling (45, 140), a pathway that has been demonstrated to be
components such as laminin and entactin. The membrane-type associated with kidney development and injury (41, 44). Stud-
MMPs (MT-MMPs) are embedded in the plasma membrane of ies from our laboratory have also demonstrated that tissue
the cells via type I or II domains or glycosylphosphatidylinosi- plasminogen activator (tPA) leads to an enhanced transcription
tol (GPI)-anchors. Other MMPs do not conform easily to of MMP-9 in renal interstitial fibroblasts, through a pathway
classification and include macrophage elastase (MMP-12) and dependent on cell-surface LDL receptor-related protein-1
(LRP-1) and on Erk-1/2 activation. This activation is indepen-
dent of the enzymatic activity of tPA (49, 148).
Posttranslational activation. The MMPs are synthesized as
inactive zymogens maintained by a conserved cysteine residue
that interacts with the zinc in the active site and prevents a
catalytic water molecule from associating with the metal ion,
rendering the protease inactive (87). Activation requires disrup-
tion of this so-called “cysteine switch,” which can be accom-
plished through proteolytic cleavage of the propeptide by trypsin,
plasmin, or even other MMPs (137). In addition, several MMPs
contain a furin-sensitive cleavage site for the propeptide, indicat-
ing intracellular activation of these enzymes (87).
MMP-9 has a unique relationship with neutrophil gelatinase-
associated lipocalin (NGAL), as they are coexpressed by neu-
trophils and can interact with each other. This interaction
prevents the degradation of MMP-9, potentially prolonging its
lifespan and effects. As NGAL has long been recognized as a
potential biomarker for kidney injury, detectable in the urine of
affected individuals, it is conceivable that the interaction of
MMP-9 and NGAL could play a specific role in prolonging
MMP action in renal pathology (130), and this hypothesis
deserves further attention in the future.
An interesting story of MMPs with nitrosative and oxidative
stress has also emerged. The cysteine switch contains a thiol
residue that can be altered by reactive oxygen species (ROS)
and reactive nitrogen species (RNS), causing dissociation from
the catalytic site, leading to enzyme activation (96, 137). Such
Fig. 2. Crystal structure of MMP-1. Active human MMP-1 is shown as a regulation is tightly controlled, as it is shown that at lower
ribbon diagram. The structure was determined with the help of a single amino concentrations, MMP-7 is activated by hypochlorous acid
acid mutation to create an inhibitor-free structure that should closely represent
the active form. Four calcium ions and 2 zinc ions are part of the structure.
(from myeloperoxidase), whereas at higher concentrations
Helices (hA, hB, hC) and strands (s1–5) are labeled, as are the 4 propeller- MMP-7 is inactivated, apparently through oxidative cross-
shaped blades of the hemopexin domain (bI–IV). Published with permission (54). linking of amino acids affecting the active site (31, 32). These

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
MMPs IN KIDNEY DISEASES F1353
Table 1. MMPs and their substrates
MMP Alternate Names Selected Substrates Selected Reference(s)

MMP-1 Collagenase-1 Collagen I, II, III, entactin, perlectan, IGF-BP-2 and -3, pro-IL-1␤, IL-1␤ 38
MMP-2 Gelatinase A Gelatin, collagen IV, V, XI, laminin, aggrecan, pro-TGF-␤, pro-TNF-␣, 18, 131
IGFBP-3 and -5
MMP-3 Stromelysin-1 Aggrecan, laminin, fibronectin, fibrinogen, MCP-1 to -4, pro-MMP-1, -3, -7, 56, 126
-8, -9, -13
MMP-7 Matrilysin Plasminogen, pro-␣-defensin, FasL, pro-TNF-␣, E-cadherin, syndecan, 45, 78, 99, 140
pro-MMPs
MMP-8 Collagenase-2 Collagen I-III, VII, X, aggrecan, fibronectin, pro-TNF-␣, IGF-BP, MCP-1, 136
angiotensin
MMP-9 Gelatinase B Gelatin, collagen IV, V, XI, pro-IL-8, Pro-TNF-␣, pro-TGF-␤, pro-MMP-2, 49, 146, 153

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


-9, -13
MMP-10 Stromelysin-2 Gelatins, fibronectin, proteoglycan, pro-MMP-1, -8, -10 93, 106
MMP-11 Stromelysin-3 Fibronectin, laminin, aggrecan, IGFBP-1 84
MMP-12 Metalloelastase Elastin, fibronectin, laminin, plasminogen, pro-TNF-␣ 46
MMP-13 Collagenase-3 Collagen I, II, III, entactin, aggrecan, tenascin, pro-TNF-␣, pro-MMP-9, -13 62, 63
MMP-14 MT1-MMP Collagen I, II, III, laminin, fibronectin, pro-MMP-2, -13, CD44, tissue 87
transglutaminase
MMP-15 MT2-MMP Pro-MMP-2, pro-TNF-␣, tissue transglutaminase 1
MMP-16 MT3-MMP Collagen III, pro-MMP-2, pro-TNF-␣, tissue transglutaminase
MMP-17 MT4-MMP Gelatin, fibronectin, fibrin, pro-MMP-2, ADAMTS-4, TIMPs, pro-TNF-␣ 118
MMP-18 Collagenase-4 Collagen I, II, III 129
MMP-19 Stromelysin-4 Collagen IV, gelatin, laminin 10, 85
MMP-20 Enamelysin Amelogenin, aggrecan, cartilage oligomeric matrix protein (COMP) 73
MMP-21 Gelatin, ␣-1-antitrypsin 76
MMP-23 May be similar to stromelysins and collagenases 138
MMP-24 MT5-MMP Pro-MMP-2
MMP-25 MT6-MMP Collagen IV, gelatin, fibrin, fironectin, pro-MMP-2 and -9, TIMPs, uPAR 118
MMP-26 Matrilysin-2 Collagen IV, fibronectin, fibrin, fibrinogen, pro-MMP-9 98, 115
MMP-27 Gelatin, casein
MMP-28 Epilysin Neural cell adhesion molecule (NCAM), casein 53
MMP, matrix metalloproteinase. Data were compiled with the noted references as well as previous reviews (37, 95). uPAR, urokinase plasminogen activator
receptor; IGF-BP, insulin-like growth factor binding protein.

findings suggest that although the cysteine switch thiol is the kidney disease. Table 2 summarizes the effect of these MMP-
preferred site of oxidative alteration, at times of greater oxidant specific interventions in renal pathology.
production, the enzyme is inactivated, possibly preventing Acute kidney injury. Acute kidney injury (AKI) is usually
overactivity. Such fine-tuning of MMP activity by oxidative caused by exposure to ischemia-reperfusion injury or nephro-
stress underscores the complexity of MMP regulation. Whether toxins and is a major cause of morbidity and mortality (142).
such mechanisms play a role in renal pathology remains to be Urinary MMP-9, in combination with other markers, could
determined. serve as a biomarker of AKI (40). A wide variety of AKI
Natural inhibitors. MMP activity is regulated by a family of models have characterized an increase in the expression of
naturally occurring, endogenous inhibitors known as tissue inhib- MMP-2 and MMP-9 as well as MMP-7 (8, 11, 91). Although
itors of metalloproteinases (TIMPs). The four known TIMPs have methods to decrease MMP activity generally reduce injury in
varying specificities for different MMPs but collectively can the majority of experimental models, at least one study shows
inhibit all of them. Generally, TIMPs combine in 1:1 stoichiom- that MMP-9 knockouts did worse, due to a diminished anti-
etry with MMPs and associate with the catalytic and hemopexin apoptotic signaling through stem cell factor (Table 2).
domains to cause inhibition (87). However, TIMPs also partici- Diabetic nephropathy. Diabetic nephropathy is the most
pate in metalloproteinase activation, as in the case of TIMP-2- common cause of end-stage kidney disease in the industrialized
associated activation of MMP-2 in the presence of MT1-MMP. world and is characterized clinically by albuminuria and pro-
There is now evidence that TIMPs can participate in other non- gressive renal failure, and pathologically by mesangial expan-
traditional functions such as cell proliferation, apoptosis, and sion, glomerulosclerosis, tubular atrophy, and interstitial fibro-
angiogenesis, and that a role of MMPs is to modulate TIMP sis (87). Collectively, the majority of studies show increased
signaling via sequestering a limited pool of TIMPs (82, 87). MMP-2, MMP-8, and MMP-9 in the serum and urine from
diabetic patients (36, 60, 65, 108, 125, 126, 135). In fact, a
MMPs in Renal Pathology
positive correlation exists between degree of albuminuria and
While the temporal and spatial expression of MMPs in the levels of urinary MMP-9 (125). In addition, urinary excretion
kidney has not been completely characterized, numerous stud- of NGAL is concomitantly increased with MMP-9, which is
ies have demonstrated dysregulation of MMPs in a wide particularly intriguing since NGAL is not only a proposed
variety of kidney disorders. Figure 3 compiles the distribution biomarker of kidney injury but can aid in sustained activation
and expression profile of MMPs along the nephron in normal of MMPs (127). An elevated serum MMP-7 level may also be
and pathological conditions. Ample evidence also suggests that a biomarker, as it is inversely correlated with renal function in
MMPs could be potential targets for therapeutic intervention in proteinuric diabetic patients (5). Tubular expression of MMP-7

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
F1354 MMPs IN KIDNEY DISEASES

Fig. 3. Expression profile of MMP/tissue inhibitors of metallo-


proteinases (TIMPs) along the nephron. The various MMPs and
their inhibitors have unique expression profiles along the
nephron. Currently known localizations are shown. An asterisk
(*) denotes upregulation in renal pathological conditions, while
a dagger (†) denotes downregulation in disease states. PCT,

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


proximal convoluted tubule; LOH, loop of Henle; DCT, distal
convoluted tubule; CD, collecting duct. Data are compiled from
many original studies.

is induced in human kidney biopsies from patients with dia- MMP activity, particularly of MMP-2 and MMP-9, ameliorate
betic nephropathy, and its level is closely correlated with kidney lesions (107, 114, 144, 150), whereas in other models
␤-catenin abundance (45). Observations have also described maintenance of MMPs, namely, MMP-1 and in some cases
downregulation of MMP-1 and TIMPs, with upregulation of even MMP-9, is associated with improvement (see Table 2) (4,
membrane-type MMPs (21, 60, 108, 109). 34). Similarly contradictory, TIMP-3 was elevated in diabetes,
The role of MMPs in the pathogenesis of diabetic nephrop- but TIMP-3 null mice actually had worsened fibrosis and
athy is controversial and inconsistent. In addition, experimental increased activation of MMP-2 (55, 57). Collectively, these
models do not always recapitulate clinical findings, as recently data highlight the complexities of MMP pathobiology and the
reviewed (126). These contradictions could be caused by continued need for mechanistic studies.
differences in animal strains, samples obtained, and cell types Nondiabetic glomerular disease. MMP levels are altered in
examined, as well as the fact that diabetes treatment affects other nondiabetic glomerular diseases. Single nucleotide poly-
MMP expression (126). Some models show that decreased morphisms in MMP-14 confer risk for developing focal seg-

Table 2. Effect of MMP manipulations in kidney diseases


Intervention Model System Overall Effect Specific Findings Reference(s)

Acute kidney injury


MMP-2 null Mouse, I/R Improved Reduced incidence of AKI 59, 61
MMP-9 null Mouse, I/R Improved Prevented loss of microvascular density 61
MMP-9 null Mouse, I/R Worsened Tubular apoptosis, delayed recovery 11
MMP inhibition Rat, I/R Improved Decreased AKI, apoptosis, and cytokine release 51
MMP inhibition Mouse, I/R Improved Decreased histological damage 91
Diabetic nephropathy
MMP inhibition Human, diabetes Improved Decreased proteinuria at 3 mo 2
MMP-1 transgenic Mouse, diabetes Improved Decreased fibrosis 4
MMP inhibition Rat, diabetes Improved Reduced proteinuria/glomerulosclerosis/fibrosis 144
Interstitial fibrosis
TIMP-1 null Mouse, UUO No change Compensatory TIMP-3 upregulation 57
TIMP-1 overexpression Mouse, UUO Worsened Increased fibrosis and inflammation 13
TIMP-3 null Mouse, aging, UUO Worsened Increased fibrosis 55
MMP-9 null Mouse, UUO Improved Reduced fibrosis and EMT 141
MMP-2 inhibition Mouse, UUO Worsened Reduced inflammation, accelerated fibrosis 90
Inherited diseases
MMP inhibition Rat, PKD Improved Decreased cyst number 92
MMP gene deletion or inhibition Mouse, Alport’s Stage-specific Early inhibition attenuated disease, late inhibition 153
worsened disease
Glomerulonephritis
Doxycycline Rat, IC nephritis Improved Decreased proliferative lesions, IgG/C3 deposits 110
MMP inhibition Rats, MPGN Improved Decreased mesangial proliferation, fibrosis 119
MMP inhibition Rats, MPGN Improved Decreased collagen, ␣-SMA expression 81
MMP-9 or -13 null FSGS Improved Decreased collagen IV and proteinuria 111
TIMP, tissue inhibitor of metalloproteinase; I/R, ischemia-reperfusion injury; AKI, acute kidney injury; UUO, unilateral ureteral obstruction; PKD, polycystic
kidney disease; EMT, epithelial-mesenchymal transition; MPGN, membranoproliferative glomerulonephritis; FSGS, focal segmental glomerulosclerosis;
␣-SMA, ␣-smooth muscle actin.

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
MMPs IN KIDNEY DISEASES F1355
mental glomerulosclerosis (FSGS), although the underlying evidence may indicate a role for MMPs. Numerous studies
mechanism is unknown (86). Patients with FSGS, minimal have collectively identified increases in MMP-2, MMP-7,
change disease, membranous nephropathy, human immunode- MMP-8, and MMP-9, as well as TIMP-1 and -2 in cases of
ficiency virus (HIV)-associated nephropathy, as well as anti- allograft rejection (16, 27, 68, 80, 104, 105, 145). Interestingly,
neutrophil cytoplasmic antibody (ANCA)-associated vasculi- polymorphisms in MMP-2 and -9 have been associated with
tis, each had unique expression patterns of MMP-2 and increased allograft survival (116), and the immunosuppressive
MMP-9 (3, 9, 24, 112, 113). Increased glomerular MMP-9 was drug rapamycin inhibits MMP-9 expression while increasing
found in lupus nephritis, IgA nephropathy, Henoch-Schoenlein TIMP-1 (94). The potential mechanism of MMPs in rejection
Purpura (HSP), and postinfectious glomerulonephritis (134). remains an area of active investigation.
MMP-7 is induced in renal tubular epithelium of the kidneys in
FSGS and IgA nephropathy (45). MMP-3 is upregulated in Mechanisms of MMP Action in Kidney Disease

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


HSP and ANCA vasculitis (113, 115).
Experimentally, MMP-9 is upregulated in models of FSGS, As renal fibrosis is the common final outcome of CKD, it
lupus nephritis, and Thy1.1 nephritis, a model for mem- was initially thought that MMPs may be globally protective
branoproliferative glomerulonephritis (MPGN) (69, 81, 128, through antagonism of ECM accumulation. With an increas-
132). However, impaired resolution of disease appears to be ingly expanding list of new MMP substrates, one has to
associated with a paradoxical decrease in MMP-9 late in the concede that MMP roles are not limited to digestion of matrix.
course of Thy1.1 nephritis (128). Meanwhile, MMP-2 is in- We have recently proposed that renal fibrosis appears to
creased in both lupus and MPGN (81, 119, 133). A few studies progress through a series of phases including inflammation,
utilizing both selective and nonselective MMP inhibitors in fibroblast/myofibroblast activation, matrix deposition, and fi-
these models appear to ameliorate renal injury (Table 2). brosis (70). It is conceivable that MMPs act at all levels of this
Inherited kidney disease. Polycystic kidney disease (PKD) is pathologic process, as well as in regulation of apoptosis and
the most common inherited kidney disorder and has been angiogenesis (Figs. 4 and 5).
associated with MMP dysregulation. In serum of PKD patients, Inflammation. MMPs have critical roles in inflammatory cell
there is an increase in TIMP-1, MMP-1, MMP-9, and type IV recruitment and chemotaxis, thereby regulating the inflamma-
collagen (a target of MMP-9) compared with controls (88). tory response (Fig. 4). ICAM-1, a cell surface molecule pro-
There is also dramatic upregulation of MMP-7 associated with moting leukocyte transmigration into tissues, is cleaved by
expression of Wnt in PKD (120). As Wnt/␤-catenin signaling MMP-13 and MMP-14, suggesting an anti-inflammatory effect
is activated in tubular injury, it now appears that MMP-7 is a (28, 124). Consistent with this, transgenic TIMP-1 overexpres-
target of this pathological signaling. MMP-14 is also noted in sion does increase ICAM-1 in proximal tubular epithelium
cyst-lining epithelia in experimental PKD, and MMP inhibitor (13). MMP-9 appears to play a role in self-propagating inflam-
therapy decreases cyst formation (92). mation by creating collagen fragments which are both chemot-
Alport syndrome (AS) is characterized by a mutation in colla- actic for neutrophils and stimulate them to release more
gen IV, which may lead to altered basement membrane proteol- MMP-9 (146). Furthermore, MMP-9 mediates dendritic cell
ysis. Upregulation of MMP-2, -3, and -9 occurred in both human (DC) migratory capacity and may also bind to cell surface
AS and in animal models, and each MMP could digest AS- costimulatory molecules (50, 151). MMP-14 similarly appears
derived basement membrane in vitro. Interestingly, early MMP to have stimulatory roles in DC migration (35, 149). MMP-7
inhibition before onset of proteinuria attenuates renal lesions cleaves syndecans and their attached neutrophilic chemokines,
while inhibition after proteinuria accelerates disease progression increasing inflammation by generating a chemotactic gradient
(153). Numerous other MMPs are upregulated in AS, but their (64, 122). In addition, MMP-7 may promote inflammation by
roles have yet to be determined (22, 79, 103). facilitating the release of TNF-␣ from macrophages (43). This
Chronic allograft nephropathy. Chronic rejection remains MMP also cleaves E-cadherin and can regulate migration of DCs
the major limitation of renal transplantation, and circumstantial containing the E-cadherin receptor, CD103 (␣E␤7-integrin) (75).

Fig. 4. Roles of MMPs in renal inflammation. MMPs, together


with TIMPs, regulate the activation, cytokine release, che-
motaxis, proliferation, and apoptosis of multiple inflammatory
cells.

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
F1356 MMPs IN KIDNEY DISEASES

Fig. 5. Roles of MMPs in regulating epithelial-mesenchymal


transition (EMT) and renal fibrosis. MMPs are instrumental in
mediating tubular EMT via E-cadherin shedding and degrada-
tion of tubular basement membrane (TBM). MMPs, together
with TIMPs, also regulate the proliferation/apoptosis of inter-
stitial fibroblasts and glomerular mesangial cells. The proteo-
lytic products of matrix by MMPs also play a role in regulating
cellular activities such as endothelial cell growth and angiogenesis.

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


After chemotaxis, MMPs also regulate inflammatory cell quently bind to the EGF receptor (100). Similarly, MMPs may
function. Gelatinase inhibition depresses lymphocyte prolifer- sequester TIMP-1, which could lead to inhibition of TIMP-1-
ation in vitro and in vivo in experimental lung disease (12). mediated fibroblast proliferation (72).
MMP-8 and -9 promote neutrophil survival (23, 58). Although General MMP inhibition induces cell cycle arrest and apop-
unproven, it has been proposed that MMPs could play a role in tosis in mesangial cells in anti-Thy1.1 nephritis, while TIMP-1
exposing immunoreactive epitopes through cleavage of sub- similarly reduces apoptosis in mesangial cells under stress in
strates, leading to immune complex deposition (132). Regula- vitro (25, 67). MMP-7 can stimulate apoptosis through cleav-
tion can occur in both ways, as inflammatory cytokines mod- ing and generating active soluble Fas ligand (101). On the
ulate MMP production (89). other hand, MMP-9 protects against apoptosis in acute kidney
Inflammation mediates transplant rejection, but we can only injury through release of soluble stem cell factor (SCF), which
infer from nonrenal organ transplants the role of MMPs in the is the ligand for the c-kit tyrosine kinase receptor (11).
kidney allografts. MMP-2 null mice have decreased T cell MMPs also modulate apoptosis via surprising mechanisms
alloreactivity and inflammatory cytokine release, while MMP-9 separate from ectodomain shedding. MMP-8 null mice have
null mice have increased alloreactivity and cytokine release (14). reduced neutrophil apoptosis via downregulation of caspases
Broad-spectrum inhibition of MMPs leads to decreased inflam- (23). Interestingly, MMP-3 could actually translocate into the
matory cell chemotaxis and improves cardiac graft survival (26). nucleus of mammalian cells and stimulate apoptosis, and this
However, these results may be tissue specific, as MMP-9 null effect is dependent on retention of its catalytic activity (117).
mice have improvement in survival of tracheal allografts (30). Similarly, MMP-1 can localize to mitochondria and the nu-
Epithelial-mesenchymal transition. Epithelial-mesenchymal cleus of various cell types, and knockdown of MMP-1 by small
transition (EMT) is the conversion of epithelium to a fibro- interfering RNA (siRNA) sensitizes cells to apoptosis (66).
blastic or myofibroblastic phenotype (71). Although the role of Further investigation is necessary to determine whether these
EMT in renal fibrosis is controversial (102, 152), it is clear that mechanisms impact the development of renal diseases.
MMPs are a part of its regulation under experimental condi- ECM accumulation. Mesangial cell activation is believed to
tions. MMP-2 localizes to areas of purported EMT, and, when be responsible for glomerulosclerosis development, and evi-
added to epithelial cultures, is sufficient to induce the expres- dence supports a pathogenic role for MMP-2, which is upregu-
sion of ␣-smooth muscle actin (␣-SMA), a marker of myofi- lated in kidney fibrosis (18). MMP-2 production is increased in
broblasts (17). Furthermore, mice overexpressing MMP-2 in mesangial cell cultures when exposed to components of the
proximal tubular epithelium induces EMT in a process char- ECM, including collagen 1, vitronectin, and fibronectin (77).
acterized by basement membrane disruption, leading to spon- Cultured mesangial cells with high expression of MMP-2 have
taneous tubular atrophy consistent with chronic kidney injury greater proliferative capacity, are more motile, and produce
(18). MMP-3 induces EMT in lung epithelia and promotes more ␣-SMA and collagen. This phenotype is reversed with
fibrosis in vivo (147). MMP-9 could mediate EMT and in- MMP-2-silencing RNA (131). Therefore, somewhat surpris-
crease the migratory capacity of kidney epithelial cells (52, ingly, MMP-2 can promote ECM production and accumulation
123). Both MMP-3 and MMP-9 could individually induce in kidney cells. Whether MMP-2 exerts its action via a direct
EMT in renal tubular cell cultures through cleavage of the mechanism or an indirect one, such as promoting the release of
epithelial cell marker E-cadherin, which leads to activation of fibrogenic cytokines, remains to be determined.
␤-catenin (Fig. 5) (154). Consistent with these results, MMP-9 ECM degradation. The excessive accumulation of ECM in
null mice have decreased EMT as measured by ␣-SMA and CKD is likely to involve aberrant levels of MMPs/TIMPs.
decreased fibrosis after obstructive injury (141). MMP-1 is downregulated in aging kidneys that have collagen
Cell proliferation and apoptosis. One of the interesting roles deposition (33). TIMP-1 overexpression occurs in fibrosis and can
discovered for MMPs is their involvement in the regulation of promote fibroblast proliferation independently of MMP inhibition
cell proliferation and apoptosis (Fig. 5) (72, 100). MMPs (72). TIMP-1 deficiency does not prevent fibrosis, perhaps due to
modulate cell proliferation primarily by indirect mechanisms. the compensatory upregulation of other TIMPs (57).
For instance, MMP-9 is known to cleave membrane-bound While the degradation of ECM proteins by MMPs is beneficial
precursor proteins and release EGF-like ligands that subse- in that it reduces matrix accumulation, products of the degraded

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
MMPs IN KIDNEY DISEASES F1357
ECM components may be capable of exerting some biological sufficient to induce EMT, a potentially significant step in fibrosis.
actions. In this regard, a recent study shows that MMP-9 induced Surprising intracellular localization of MMPs and their roles in
EMT in vitro only when tubular cells were cultured on Matrigel regulating various cellular activities are just beginning to emerge.
(141), suggesting that the proteolytic products of Matrigel by The discovery of the specific roles for various MMPs does
MMP-9 are instrumental in promoting EMT. Therefore, increased suggest possible therapeutic options for patients with renal
ECM degradation by MMPs, per se, may not necessarily translate diseases. However, as described in this review, the roles of
into a reduced fibrosis in CKD. MMPs in kidney diseases are extremely complex, necessitating
Degradation of the basement membrane, which is mainly that specific inhibitors target a single pathological MMP,
composed of type IV collagen and laminin, by MMPs such as delivered at a particular point in time when that MMP is active,
MMP-2, -7 and -9 could have a detrimental effect on the without affecting MMPs important for normal tissue homeo-
integrity of renal parenchyma, thereby promoting the progres- stasis and function. Such therapy is challenging, even concep-

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


sion of CKD (Fig. 5). We have shown that although tPA can tually, and suggests that clinically useful MMP inhibitors are
lead to myofibroblast activation, serve as a mitogen for renal still a distant reality.
fibroblasts, and promote their survival (42, 47, 48), its ability to Nonetheless, continued research is needed to further refine
induce MMP-9, which in turn impairs tubular basement mem- our understanding of MMPs in specific renal pathologies.
brane, probably plays a critical role in renal fibrogenesis (148). Identification of the varying mechanisms of action of MMPs
Taken together, it is conceivable that the impact of ECM will undoubtedly aid in our basic understanding of the patho-
degradation by MMPs in CKD is very complex and could be genesis of various kidney disorders. Utilizing this knowledge
either positive or negative to disease progression, depending on should renew the hope for targeted therapies that can be
MMP substrate specificities, disease models, and stages. beneficial for patients with kidney diseases in the future.
Vascular remodeling. MMPs also play an important role in
regulating endothelial cell behavior and vascular wall stiffness/ GRANTS
elasticity, thereby affecting vascular physiology and pathology. The work was supported by National Institute of Diabetes and Digestive
It is well known that the proteolytic products of collagens by and Kidney Disease Grants DK064005 and DK091239.
MMPs, such as endostatin, are critical mediators in regulating DISCLOSURES
endothelial cell biology and angiogenesis (39, 143) (Fig. 5).
Angiostatin, generated by MMP-mediated degradation of col- No conflicts of interest, financial or otherwise, are declared by the authors.
lagens, is antiangiogenic and is increased in ischemic injury REFERENCES
(8), consistent with a finding of reduced peritubular capillary
density in the outer medulla after acute renal failure (6). This 1. Abraham R, Schafer J, Rothe M, Bange J, Knyazev P, Ullrich A.
Identification of MMP-15 as an anti-apoptotic factor in cancer cells. J
is accompanied by persistent hypoxia that can be detected up to Biol Chem 280: 34123–34132, 2005.
5 wk after ischemia-reperfusion injury (7). These changes in 2. Aggarwal HK, Jain D, Talapatra P, Yadav RK, Gupta T, Kathuria
renal vasculature could portend worsened renal prognosis and KL. Evaluation of role of doxycycline (a matrix metalloproteinase
increased vulnerability to future injuries. inhibitor) on renal functions in patients of diabetic nephropathy. Ren Fail
More recent studies confirm increased perivascular MMP-2 32: 941–946, 2010.
3. Ahuja TS, Gopalani A, Davies P, Ahuja H. Matrix metalloproteinase-9
and -9 after acute ischemic injury, and MMP-9 null mice expression in renal biopsies of patients with HIV-associated nephropa-
display reduced microvascular losses after ischemia, associated thy. Nephron Clin Pract 95: c100 –c104, 2003.
with preserved VEGF levels (61). Vascular permeability is 4. Aoyama T, Yamamoto S, Kanematsu A, Ogawa O, Tabata Y. Local
reduced with inhibition of MMPs (121). Systemic effects are delivery of matrix metalloproteinase gene prevents the onset of renal
sclerosis in streptozotocin-induced diabetic mice. Tissue Eng 9: 1289 –
also present, as CKD and dialysis patients have higher MMP-2 1299, 2003.
and -9 levels in extrarenal arteries, associated with calcification 5. Ban CR, Twigg SM, Franjic B, Brooks BA, Celermajer D, Yue DK,
and loss of elasticity. MMP inhibitors are capable of improving McLennan SV. Serum MMP-7 is increased in diabetic renal disease and
vessel relaxation (19, 20). diabetic diastolic dysfunction. Diabetes Res Clin Pract 87: 335–341,
2010.
6. Basile DP, Donohoe D, Roethe K, Osborn JL. Renal ischemic injury
Conclusion and Perspectives results in permanent damage to peritubular capillaries and influences
long-term function. Am J Physiol Renal Physiol 281: F887–F899, 2001.
Numerous inherited and acquired kidney diseases are char- 7. Basile DP, Donohoe DL, Roethe K, Mattson DL. Chronic renal
acterized by dysregulation of MMPs. In disorders ranging from hypoxia after acute ischemic injury: effects of L-arginine on hypoxia and
PKD to diabetic nephropathy to glomerulonephritis, MMPs are secondary damage. Am J Physiol Renal Physiol 284: F338 –F348, 2003.
8. Basile DP, Fredrich K, Weihrauch D, Hattan N, Chilian WM.
upregulated (MMP-2, -7, and -9) as well as downregulated Angiostatin and matrix metalloprotease expression following ischemic
(MMP-1). With the availability of MMP null animal models as acute renal failure. Am J Physiol Renal Physiol 286: F893–F902, 2004.
well as MMP-specific pharmacological inhibitors, we now 9. Bauvois B, Mothu N, Nguyen J, Nguyen-Khoa T, Noel LH, Jungers
have the tools to carry out sophisticated mechanistic studies P. Specific changes in plasma concentrations of matrix metalloprotei-
that promise to provide greater insights into their exact roles in nase-2 and -9, TIMP-1 and TGF-beta1 in patients with distinct types of
primary glomerulonephritis. Nephrol Dial Transplant 22: 1115–1122,
renal pathophysiology. 2007.
Far from being simple enzymes whose only role is to maintain 10. Beck IM, Ruckert R, Brandt K, Mueller MS, Sadowski T, Brauer R,
ECM homeostasis, MMPs and their associated TIMPs are now Schirmacher P, Mentlein R, Sedlacek R. MMP19 is essential for T cell
implicated in a diverse array of biological processes and cellular development and T cell-mediated cutaneous immune responses. PLoS
One 3: e2343, 2008.
events. MMPs can both promote and inhibit inflammation, acting 11. Bengatta S, Arnould C, Letavernier E, Monge M, de Preneuf HM,
on events from chemotaxis to lymphocyte proliferation. Some Werb Z, Ronco P, Lelongt B. MMP9 and SCF protect from apoptosis
MMPs such as MMP-2, MMP-7, and MMP-9 seem necessary and in acute kidney injury. J Am Soc Nephrol 20: 787–797, 2009.

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
F1358 MMPs IN KIDNEY DISEASES

12. Benson HL, Mobashery S, Chang M, Kheradmand F, Hong JS, 31. Fu X, Kao JL, Bergt C, Kassim SY, Huq NP, d’Avignon A, Parks
Smith GN, Shilling RA, Wilkes DS. Endogenous matrix metalloprotei- WC, Mecham RP, Heinecke JW. Oxidative cross-linking of tryptophan
nases 2 and 9 regulate activation of CD4⫹ and CD8⫹ T cells. Am J to glycine restrains matrix metalloproteinase activity: specific structural
Respir Cell Mol Biol 44: 700 –708, 2011. motifs control protein oxidation. J Biol Chem 279: 6209 –6212, 2004.
13. Cai G, Zhang X, Hong Q, Shao F, Shang X, Fu B, Feng Z, Lin H, 32. Fu X, Kassim SY, Parks WC, Heinecke JW. Hypochlorous acid
Wang J, Shi S, Yin Z, Chen X. Tissue inhibitor of metalloproteinase-1 generated by myeloperoxidase modifies adjacent tryptophan and glycine
exacerbated renal interstitial fibrosis through enhancing inflammation. residues in the catalytic domain of matrix metalloproteinase-7 (matrily-
Nephrol Dial Transplant 23: 1861–1875, 2008. sin): an oxidative mechanism for restraining proteolytic activity during
14. Campbell LG, Ramachandran S, Liu W, Shipley JM, Itohara S, inflammation. J Biol Chem 278: 28403–28409, 2003.
Rogers JG, Moazami N, Senior RM, Jaramillo A. Different roles for 33. Gagliano N, Arosio B, Santambrogio D, Balestrieri MR, Padoani G,
matrix metalloproteinase-2 and matrix metalloproteinase-9 in the patho- Tagliabue J, Masson S, Vergani C, Annoni G. Age-dependent expres-
genesis of cardiac allograft rejection. Am J Transplant 5: 517–528, 2005. sion of fibrosis-related genes and collagen deposition in rat kidney
15. Catania JM, Chen G, Parrish AR. Role of matrix metalloproteinases in cortex. J Gerontol A Biol Sci Med Sci 55: B365–B372, 2000.

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


renal pathophysiologies. Am J Physiol Renal Physiol 292: F905–F911, 34. Gagliardini E, Corna D, Zoja C, Sangalli F, Carrara F, Rossi M,
2007. Conti S, Rottoli D, Longaretti L, Remuzzi A, Remuzzi G, Benigni A.
16. Chang HR, Kuo WH, Hsieh YS, Yang SF, Lin CC, Lee ML, Lian JD, Unlike each drug alone, lisinopril if combined with avosentan promotes
Chu SC. Circulating matrix metalloproteinase-2 is associated with cys- regression of renal lesions in experimental diabetes. Am J Physiol Renal
tatin C level, posttransplant duration, and diabetes mellitus in kidney Physiol 297: F1448 –F1456, 2009.
transplant recipients. Transl Res 151: 217–223, 2008. 35. Gawden-Bone C, Zhou Z, King E, Prescott A, Watts C, Lucocq J.
17. Cheng S, Lovett DH. Gelatinase A (MMP-2) is necessary and sufficient Dendritic cell podosomes are protrusive and invade the extracellular
for renal tubular cell epithelial-mesenchymal transformation. Am J matrix using metalloproteinase MMP-14. J Cell Sci 123: 1427–1437,
Pathol 162: 1937–1949, 2003. 2010.
18. Cheng S, Pollock AS, Mahimkar R, Olson JL, Lovett DH. Matrix 36. Gharagozlian S, Svennevig K, Bangstad HJ, Winberg JO, Kolset SO.
metalloproteinase 2 and basement membrane integrity: a unifying mech- Matrix metalloproteinases in subjects with type 1 diabetes. BMC Clin
anism for progressive renal injury. FASEB J 20: 1898 –1900, 2006. Pathol 9: 7, 2009.
19. Chung AW, Yang HH, Kim JM, Sigrist MK, Chum E, Gourlay WA, 37. Gialeli C, Theocharis AD, Karamanos NK. Roles of matrix metallo-
Levin A. Upregulation of matrix metalloproteinase-2 in the arterial proteinases in cancer progression and their pharmacological targeting.
vasculature contributes to stiffening and vasomotor dysfunction in pa- FEBS J 278: 16 –27, 2011.
tients with chronic kidney disease. Circulation 120: 792–801, 2009. 38. Gross J. How tadpoles lose their tails: path to discovery of the first
20. Chung AW, Yang HH, Sigrist MK, Brin G, Chum E, Gourlay WA, matrix metalloproteinase. Matrix Biol 23: 3–13, 2004.
Levin A. Matrix metalloproteinase-2 and -9 exacerbate arterial stiffening 39. Hamano Y, Kalluri R. Tumstatin, the NC1 domain of alpha3 chain of
and angiogenesis in diabetes and chronic kidney disease. Cardiovasc Res type IV collagen, is an endogenous inhibitor of pathological angiogenesis
84: 494 –504, 2009. and suppresses tumor growth. Biochem Biophys Res Commun 333:
21. Cornish TC, Bagnasco SM, Macgregor AM, Lu J, Selvin E, Ha- 292–298, 2005.
lushka MK. Glomerular protein levels of matrix metalloproteinase-1 and 40. Han WK, Waikar SS, Johnson A, Betensky RA, Dent CL, Devarajan
tissue inhibitor of metalloproteinase-1 are lower in diabetic subjects. J P, Bonventre JV. Urinary biomarkers in the early diagnosis of acute
Histochem Cytochem 57: 995–1001, 2009. kidney injury. Kidney Int 73: 863–869, 2008.
22. Cosgrove D, Meehan DT, Delimont D, Pozzi A, Chen X, Rodgers KD, 41. Hao S, He W, Li Y, Ding H, Hou Y, Nie J, Hou FF, Kahn M, Liu Y.
Tempero RM, Zallocchi M, Rao VH. Integrin alpha1beta1 regulates Targeted inhibition of ␤-catenin/CBP signaling ameliorates renal inter-
matrix metalloproteinases via P38 mitogen-activated protein kinase in stitial fibrosis. J Am Soc Nephrol 22: 1642–1653, 2011.
mesangial cells: implications for Alport syndrome. Am J Pathol 172: 42. Hao S, Shen H, Hou Y, Mars WM, Liu Y. tPA is a potent mitogen for
761–773, 2008. renal interstitial fibroblasts: role of beta1 integrin/focal adhesion kinase
23. Cox JH, Starr AE, Kappelhoff R, Yan R, Roberts CR, Overall CM. signaling. Am J Pathol 177: 1164 –1175, 2010.
Matrix metalloproteinase 8 deficiency in mice exacerbates inflammatory 43. Haro H, Crawford HC, Fingleton B, Shinomiya K, Spengler DM,
arthritis through delayed neutrophil apoptosis and reduced caspase 11 Matrisian LM. Matrix metalloproteinase-7-dependent release of tumor
expression. Arthritis Rheum 62: 3645–3655, 2010. necrosis factor-alpha in a model of herniated disc resorption. J Clin
24. Czech KA, Bennett M, Devarajan P. Distinct metalloproteinase excre- Invest 105: 143–150, 2000.
tion patterns in focal segmental glomerulosclerosis. Pediatr Nephrol 26: 44. He W, Dai C, Li Y, Zeng G, Monga SP, Liu Y. Wnt/␤-catenin
2179 –2184, 2011. signaling promotes renal interstitial fibrosis. J Am Soc Nephrol 20:
25. Daniel C, Duffield J, Brunner T, Steinmann-Niggli K, Lods N, Marti 765–776, 2009.
HP. Matrix metalloproteinase inhibitors cause cell cycle arrest and 45. He W, Tan RJ, Li Y, Wang D, Nie J, Hou FF, Liu Y. Matrix
apoptosis in glomerular mesangial cells. J Pharmacol Exp Ther 297: metalloproteinase-7 as a surrogate marker predicts renal Wnt/␤-catenin
57–68, 2001. activity in CKD. J Am Soc Nephrol 23: 294 –304, 2012.
26. Eaton VL, Lerret NM, Velasquez-Lopera MM, John R, Caicedo M, 46. Houghton AM, Quintero PA, Perkins DL, Kobayashi DK, Kelley
DeCresce RP, Jaramillo A. Enhanced allograft survival and modulation DG, Marconcini LA, Mecham RP, Senior RM, Shapiro SD. Elastin
of T-cell alloreactivity induced by inhibition of MMP/ADAM enzymatic fragments drive disease progression in a murine model of emphysema. J
activity. Am J Transplant 8: 507–516, 2008. Clin Invest 116: 753–759, 2006.
27. Elster EA, Hawksworth JS, Cheng O, Leeser DB, Ring M, Tadaki 47. Hu K, Lin L, Tan X, Yang J, Bu G, Mars WM, Liu Y. tPA protects
DK, Kleiner DE, Eberhardt JS, 3rd Brown TS, Mannon RB. Prob- renal interstitial fibroblasts and myofibroblasts from apoptosis. J Am Soc
abilistic (Bayesian) modeling of gene expression in transplant glomeru- Nephrol 19: 503–514, 2008.
lopathy. J Mol Diagn 12: 653–663, 2010. 48. Hu K, Wu C, Mars WM, Liu Y. Tissue-type plasminogen activator
28. Essick E, Sithu S, Dean W, D’Souza S. Pervanadate-induced shedding promotes murine myofibroblast activation through LDL receptor-related
of the intercellular adhesion molecule (ICAM)-1 ectodomain is mediated protein 1-mediated integrin signaling. J Clin Invest 117: 3821–3832,
by membrane type-1 matrix metalloproteinase (MT1-MMP). Mol Cell 2007.
Biochem 314: 151–159, 2008. 49. Hu K, Yang J, Tanaka S, Gonias SL, Mars WM, Liu Y. Tissue-type
29. Fanjul-Fernandez M, Folgueras AR, Cabrera S, Lopez-Otin C. plasminogen activator acts as a cytokine that triggers intracellular signal
Matrix metalloproteinases: evolution, gene regulation and functional transduction and induces matrix metalloproteinase-9 gene expression. J
analysis in mouse models. Biochim Biophys Acta 1803: 3–19, 2010. Biol Chem 281: 2120 –2127, 2006.
30. Fernandez FG, Campbell LG, Liu W, Shipley JM, Itohara S, Pat- 50. Hu Y, Ivashkiv LB. Costimulation of chemokine receptor signaling by
terson GA, Senior RM, Mohanakumar T, Jaramillo A. Inhibition of matrix metalloproteinase-9 mediates enhanced migration of IFN-alpha
obliterative airway disease development in murine tracheal allografts by dendritic cells. J Immunol 176: 6022–6033, 2006.
matrix metalloproteinase-9 deficiency. Am J Transplant 5: 671–683, 51. Ihtiyar E, Yasar NF, Erkasap N, Koken T, Tosun M, Oner S,
2005. Erkasap S. Effects of doxycycline on renal ischemia reperfusion injury

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
MMPs IN KIDNEY DISEASES F1359
induced by abdominal compartment syndrome. J Surg Res 167: 113–120, 75. Manicone AM, Huizar I, McGuire JK. Matrilysin (Matrix Metallopro-
2011. teinase-7) regulates anti-inflammatory and antifibrotic pulmonary den-
52. Ikari A, Takiguchi A, Atomi K, Sato T, Sugatani J. Decrease in dritic cells that express CD103 (alpha(E)beta(7)-integrin). Am J Pathol
claudin-2 expression enhances cell migration in renal epithelial Madin- 175: 2319 –2331, 2009.
Darby canine kidney cells. J Cell Physiol 226: 1471–1478, 2011. 76. Marchenko GN, Marchenko ND, Strongin AY. The structure and
53. Illman SA, Lohi J, Keski-Oja J. Epilysin (MMP-28)–structure, expres- regulation of the human and mouse matrix metalloproteinase-21 gene and
sion and potential functions. Exp Dermatol 17: 897–907, 2008. protein. Biochem J 372: 503–515, 2003.
54. Iyer S, Visse R, Nagase H, Acharya KR. Crystal structure of an active 77. Martin J, Eynstone L, Davies M, Steadman R. Induction of metallo-
form of human MMP-1. J Mol Biol 362: 78 –88, 2006. proteinases by glomerular mesangial cells stimulated by proteins of the
55. Kassiri Z, Oudit GY, Kandalam V, Awad A, Wang X, Ziou X, Maeda extracellular matrix. J Am Soc Nephrol 12: 88 –96, 2001.
N, Herzenberg AM, Scholey JW. Loss of TIMP3 enhances interstitial 78. McGuire JK, Li Q, Parks WC. Matrilysin (matrix metalloproteinase-7)
nephritis and fibrosis. J Am Soc Nephrol 20: 1223–1235, 2009. mediates E-cadherin ectodomain shedding in injured lung epithelium. Am
56. Kim EM, Hwang O. Role of matrix metalloproteinase-3 in neurodegen- J Pathol 162: 1831–1843, 2003.
eration. J Neurochem 116: 22–32, 2011.

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


79. Meehan DT, Delimont D, Cheung L, Zallocchi M, Sansom SC,
57. Kim H, Oda T, Lopez-Guisa J, Wing D, Edwards DR, Soloway PD, Holzclaw JD, Rao V, Cosgrove D. Biomechanical strain causes mal-
Eddy AA. TIMP-1 deficiency does not attenuate interstitial fibrosis in adaptive gene regulation, contributing to Alport glomerular disease.
obstructive nephropathy. J Am Soc Nephrol 12: 736 –748, 2001. Kidney Int 76: 968 –976, 2009.
58. Kolaczkowska E, Koziol A, Plytycz B, Arnold B, Opdenakker G.
80. Metzger J, Chatzikyrkou C, Broecker V, Schiffer E, Jaensch L,
Altered apoptosis of inflammatory neutrophils in MMP-9-deficient mice
Iphoefer A, Mengel M, Mullen W, Mischak H, Haller H, Gwinner W.
is due to lower expression and activity of caspase-3. Immunol Lett 126:
Diagnosis of subclinical and clinical acute T-cell-mediated rejection in
73–82, 2009.
renal transplant patients by urinary proteome analysis. Proteomics Clin
59. Kunugi S, Shimizu A, Kuwahara N, Du X, Takahashi M, Terasaki Y,
Fujita E, Mii A, Nagasaka S, Akimoto T, Masuda Y, Fukuda Y. Appl 5: 322–333, 2011.
Inhibition of matrix metalloproteinases reduces ischemia-reperfusion 81. Mitani O, Katoh M, Shigematsu H. Participation of the matrix metal-
acute kidney injury. Lab Invest 91: 170 –180, 2011. loproteinase inhibitor in Thy-1 nephritis. Pathol Int 54: 241–250, 2004.
60. Lauhio A, Sorsa T, Srinivas R, Stenman M, Tervahartiala T, Sten- 82. Moore CS, Crocker SJ. An alternate perspective on the roles of TIMPs
man UH, Gronhagen-Riska C, Honkanen E. Urinary matrix metallo- and MMPs in Pathology. Am J Pathol 180: 12–16, 2012.
proteinase -8, -9, -14 and their regulators (TRY-1, TRY-2, TATI) in 83. Morrison CJ, Butler GS, Rodriguez D, Overall CM. Matrix metallo-
patients with diabetic nephropathy. Ann Med 40: 312–320, 2008. proteinase proteomics: substrates, targets, and therapy. Curr Opin Cell
61. Lee SY, Horbelt M, Mang HE, Knipe NL, Bacallao RL, Sado Y, Biol 21: 645–653, 2009.
Sutton TA. MMP-9 gene deletion mitigates microvascular loss in a 84. Motrescu ER, Rio MC. Cancer cells, adipocytes and matrix metallo-
model of ischemic acute kidney injury. Am J Physiol Renal Physiol 301: proteinase 11: a vicious tumor progression cycle. Biol Chem 389:
F101–F109, 2011. 1037–1041, 2008.
62. Leeman MF, Curran S, Murray GI. The structure, regulation, and 85. Muller M, Beck IM, Gadesmann J, Karschuk N, Paschen A, Proksch
function of human matrix metalloproteinase-13. Crit Rev Biochem Mol E, Djonov V, Reiss K, Sedlacek R. MMP19 is upregulated during
Biol 37: 149 –166, 2002. melanoma progression and increases invasion of melanoma cells. Mod
63. Li NG, Shi ZH, Tang YP, Wang ZJ, Song SL, Qian LH, Qian DW, Pathol 23: 511–521, 2010.
Duan JA. New hope for the treatment of osteoarthritis through selective 86. Munkert A, Helmchen U, Kemper MJ, Bubenheim M, Stahl RA,
inhibition of MMP-13. Curr Med Chem 18: 977–1001, 2011. Harendza S. Characterization of the transcriptional regulation of the
64. Li Q, Park PW, Wilson CL, Parks WC. Matrilysin shedding of human MT1-MMP gene and association of risk reduction for focal-
syndecan-1 regulates chemokine mobilization and transepithelial efflux segmental glomerulosclerosis with two functional promoter SNPs. Neph-
of neutrophils in acute lung injury. Cell 111: 635–646, 2002. rol Dial Transplant 24: 735–742, 2009.
65. Li Y, Kang YS, Dai C, Kiss LP, Wen X, Liu Y. Epithelial-to- 87. Nagase H, Visse R, Murphy G. Structure and function of matrix
mesenchymal transition is a potential pathway leading to podocyte metalloproteinases and TIMPs. Cardiovasc Res 69: 562–573, 2006.
dysfunction and proteinuria. Am J Pathol 172: 299 –308, 2008. 88. Nakamura T, Ushiyama C, Suzuki S, Ebihara I, Shimada N, Koide
66. Limb GA, Matter K, Murphy G, Cambrey AD, Bishop PN, Morris H. Elevation of serum levels of metalloproteinase-1, tissue inhibitor of
GE, Khaw PT. Matrix metalloproteinase-1 associates with intracellular metalloproteinase-1 and type IV collagen, and plasma levels of metallo-
organelles and confers resistance to lamin A/C degradation during proteinase-9 in polycystic kidney disease. Am J Nephrol 20: 32–36, 2000.
apoptosis. Am J Pathol 166: 1555–1563, 2005. 89. Nee LE, McMorrow T, Campbell E, Slattery C, Ryan MP. TNF-alpha
67. Lin H, Chen X, Wang J, Yu Z. Inhibition of apoptosis in rat mesangial and IL-1beta-mediated regulation of MMP-9 and TIMP-1 in renal prox-
cells by tissue inhibitor of metalloproteinase-1. Kidney Int 62: 60 –69, imal tubular cells. Kidney Int 66: 1376 –1386, 2004.
2002. 90. Nishida M, Okumura Y, Ozawa S, Shiraishi I, Itoi T, Hamaoka K.
68. Ling XB, Sigdel TK, Lau K, Ying L, Lau I, Schilling J, Sarwal MM. MMP-2 inhibition reduces renal macrophage infiltration with increased
Integrative urinary peptidomics in renal transplantation identifies bio-
fibrosis in UUO. Biochem Biophys Res Commun 354: 133–139, 2007.
markers for acute rejection. J Am Soc Nephrol 21: 646 –653, 2010.
91. Novak KB, Le HD, Christison-Lagay ER, Nose V, Doiron RJ, Moses
69. Liu S, Li Y, Zhao H, Chen D, Huang Q, Wang S, Zou W, Zhang Y,
MA, Puder M. Effects of metalloproteinase inhibition in a murine model
Li X, Huang H. Increase in extracellular cross-linking by tissue trans-
of renal ischemia-reperfusion injury. Pediatr Res 67: 257–262, 2010.
glutaminase and reduction in expression of MMP-9 contribute differen-
tially to focal segmental glomerulosclerosis in rats. Mol Cell Biochem 92. Obermuller N, Morente N, Kranzlin B, Gretz N, Witzgall R. A
284: 9 –17, 2006. possible role for metalloproteinases in renal cyst development. Am J
70. Liu Y. Cellular and molecular mechanisms of renal fibrosis. Nat Rev Physiol Renal Physiol 280: F540 –F550, 2001.
Nephrol 7: 684 –696, 2011. 93. Orbe J, Barrenetxe J, Rodriguez JA, Vivien D, Orset C, Parks WC,
71. Liu Y. New insights into epithelial-mesenchymal transition in kidney Birkland TP, Serrano R, Purroy A, Martinez de Lizarrondo S,
fibrosis. J Am Soc Nephrol 21: 212–222, 2010. Angles-Cano E, Paramo JA. Matrix metalloproteinase-10 effectively
72. Lu Y, Liu S, Zhang S, Cai G, Jiang H, Su H, Li X, Hong Q, Zhang reduces infarct size in experimental stroke by enhancing fibrinolysis via
X, Chen X. Tissue inhibitor of metalloproteinase-1 promotes NIH3T3 a thrombin-activatable fibrinolysis inhibitor-mediated mechanism. Cir-
fibroblast proliferation by activating p-Akt and cell cycle progression. culation 124: 2909 –2919, 2011.
Mol Cells 31: 225–230, 2011. 94. Osman B, Doller A, Akool el S, Holdener M, Hintermann E,
73. Lu Y, Papagerakis P, Yamakoshi Y, Hu JC, Bartlett JD, Simmer JP. Pfeilschifter J, Eberhardt W. Rapamycin induces the TGFbeta1/Smad
Functions of KLK4 and MMP-20 in dental enamel formation. Biol Chem signaling cascade in renal mesangial cells upstream of mTOR. Cell
389: 695–700, 2008. Signal 21: 1806 –1817, 2009.
74. Mancini A, Di Battista JA. Transcriptional regulation of matrix metal- 95. Overall CM. Molecular determinants of metalloproteinase substrate
loprotease gene expression in health and disease. Front Biosci 11: specificity: matrix metalloproteinase substrate binding domains, mod-
423–446, 2006. ules, and exosites. Mol Biotechnol 22: 51–86, 2002.

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
F1360 MMPs IN KIDNEY DISEASES

96. Owens MW, Milligan SA, Jourd’heuil D, Grisham MB. Effects of 116. Singh R, Srivastava P, Srivastava A, Mittal RD. Matrix metallopro-
reactive metabolites of oxygen and nitrogen on gelatinase A activity. Am teinase (MMP-9 and MMP-2) gene polymorphisms influence allograft
J Physiol Lung Cell Mol Physiol 273: L445–L450, 1997. survival in renal transplant recipients. Nephrol Dial Transplant 25:
97. Pardo A, Selman M. Matrix metalloproteases in aberrant fibrotic tissue 3393–3401, 2010.
remodeling. Proc Am Thorac Soc 3: 383–388, 2006. 117. Si-Tayeb K, Monvoisin A, Mazzocco C, Lepreux S, Decossas M,
98. Park HI, Lee S, Ullah A, Cao Q, Sang QX. Effects of detergents on Cubel G, Taras D, Blanc JF, Robinson DR, Rosenbaum J. Matrix
catalytic activity of human endometase/matrilysin 2, a putative cancer metalloproteinase 3 is present in the cell nucleus and is involved in
biomarker. Anal Biochem 396: 262–268, 2010. apoptosis. Am J Pathol 169: 1390 –1401, 2006.
99. Parks WC, Wilson CL, Lopez-Boado YS. Matrix metalloproteinases as 118. Sohail A, Sun Q, Zhao H, Bernardo MM, Cho JA, Fridman R.
modulators of inflammation and innate immunity. Nat Rev Immunol 4: MT4-(MMP17) and MT6-MMP (MMP25), A unique set of membrane-
617–629, 2004. anchored matrix metalloproteinases: properties and expression in cancer.
100. Perng DW, Chang KT, Su KC, Wu YC, Chen CS, Hsu WH, Tsai Cancer Metastasis Rev 27: 289 –302, 2008.
CM, Lee YC. Matrix metalloprotease-9 induces transforming growth 119. Steinmann-Niggli K, Ziswiler R, Kung M, Marti HP. Inhibition of
factor-beta(1) production in airway epithelium via activation of epider-

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


matrix metalloproteinases attenuates anti-Thy1.1 nephritis. J Am Soc
mal growth factor receptors. Life Sci 89: 204 –212, 2011. Nephrol 9: 397–407, 1998.
101. Powell WC, Fingleton B, Wilson CL, Boothby M, Matrisian LM. The 120. Surendran K, Simon TC, Liapis H, McGuire JK. Matrilysin (MMP-7)
metalloproteinase matrilysin proteolytically generates active soluble Fas expression in renal tubular damage: association with Wnt4. Kidney Int
ligand and potentiates epithelial cell apoptosis. Curr Biol 9: 1441–1447, 65: 2212–2222, 2004.
1999. 121. Sutton TA, Kelly KJ, Mang HE, Plotkin Z, Sandoval RM, Dagher
102. Quaggin SE, Kapus A. Scar wars: mapping the fate of epithelial- PC. Minocycline reduces renal microvascular leakage in a rat model of
mesenchymal-myofibroblast transition. Kidney Int 80: 41–50, 2011. ischemic renal injury. Am J Physiol Renal Physiol 288: F91–F97, 2005.
103. Rao VH, Meehan DT, Delimont D, Nakajima M, Wada T, Gratton 122. Swee M, Wilson CL, Wang Y, McGuire JK, Parks WC. Matrix
MA, Cosgrove D. Role for macrophage metalloelastase in glomerular metalloproteinase-7 (matrilysin) controls neutrophil egress by generating
basement membrane damage associated with Alport syndrome. Am J chemokine gradients. J Leukoc Biol 83: 1404 –1412, 2008.
Pathol 169: 32–46, 2006. 123. Tan TK, Zheng G, Hsu TT, Wang Y, Lee VW, Tian X, Wang Y, Cao
104. Rodder S, Scherer A, Korner M, Eisenberger U, Hertig A, Raulf F, Q, Wang Y, Harris DC. Macrophage matrix metalloproteinase-9 me-
Rondeau E, Marti HP. Meta-analyses qualify metzincins and related diates epithelial-mesenchymal transition in vitro in murine renal tubular
genes as acute rejection markers in renal transplant patients. Am J cells. Am J Pathol 176: 1256 –1270, 2010.
Transplant 10: 286 –297, 2010. 124. Tarin C, Gomez M, Calvo E, Lopez JA, Zaragoza C. Endothelial
105. Rodder S, Scherer A, Raulf F, Berthier CC, Hertig A, Couzi L, nitric oxide deficiency reduces MMP-13-mediated cleavage of ICAM-1
Durrbach A, Rondeau E, Marti HP. Renal allografts with IF/TA in vascular endothelium: a role in atherosclerosis. Arterioscler Thromb
display distinct expression profiles of metzincins and related genes. Am Vasc Biol 29: 27–32, 2009.
J Transplant 9: 517–526, 2009.
125. Tashiro K, Koyanagi I, Ohara I, Ito T, Saitoh A, Horikoshi S,
106. Rodriguez JA, Orbe J, Martinez de Lizarrondo S, Calvayrac O,
Tomino Y. Levels of urinary matrix metalloproteinase-9 (MMP-9) and
Rodriguez C, Martinez-Gonzalez J, Paramo JA. Metalloproteinases
renal injuries in patients with type 2 diabetic nephropathy. J Clin Lab
and atherothrombosis: MMP-10 mediates vascular remodeling promoted
Anal 18: 206 –210, 2004.
by inflammatory stimuli. Front Biosci 13: 2916 –2921, 2008.
126. Thrailkill KM, Clay Bunn R, Fowlkes JL. Matrix metalloproteinases:
107. Rodriguez WE, Tyagi N, Joshua IG, Passmore JC, Fleming JT,
their potential role in the pathogenesis of diabetic nephropathy. Endo-
Falcone JC, Tyagi SC. Pioglitazone mitigates renal glomerular vascular
crine 35: 1–10, 2009.
changes in high-fat, high-calorie-induced type 2 diabetes mellitus. Am J
127. Thrailkill KM, Moreau CS, Cockrell GE, Jo CH, Bunn RC, Morales-
Physiol Renal Physiol 291: F694 –F701, 2006.
Pozzo AE, Lumpkin CK, Fowlkes JL. Disease and gender-specific
108. Romanic AM, Burns-Kurtis CL, Ao Z, Arleth AJ, Ohlstein EH.
Upregulated expression of human membrane type-5 matrix metallopro- dysregulation of NGAL and MMP-9 in type 1 diabetes mellitus. Endo-
teinase in kidneys from diabetic patients. Am J Physiol Renal Physiol crine 37: 336 –343, 2010.
281: F309 –F317, 2001. 128. Tomita M, Koike H, Han GD, Shimizu F, Kawachi H. Decreased
109. Rysz J, Banach M, Stolarek RA, Pasnik J, Cialkowska-Rysz A, collagen-degrading activity could be a marker of prolonged mesangial
Koktysz R, Piechota M, Baj Z. Serum matrix metalloproteinases matrix expansion. Clin Exp Nephrol 8: 17–26, 2004.
MMP-2 and MMP-9 and metalloproteinase tissue inhibitors TIMP-1 and 129. Tomlinson ML, Garcia-Morales C, Abu-Elmagd M, Wheeler GN.
TIMP-2 in diabetic nephropathy. J Nephrol 20: 444 –452, 2007. Three matrix metalloproteinases are required in vivo for macrophage
110. Saglam F, Celik A, Tayfur D, Cavdar Z, Yilmaz O, Sarioglu S, migration during embryonic development. Mech Dev 125: 1059 –1070,
Kolatan E, Oktay G, Camsari T. Decrease in cell proliferation by an 2008.
matrix metalloproteinase inhibitor, doxycycline, in a model of immune- 130. Tschesche H, Zolzer V, Triebel S, Bartsch S. The human neutrophil
complex nephritis. Nephrology (Carlton) 15: 560 –567, 2010. lipocalin supports the allosteric activation of matrix metalloproteinases.
111. Sakamaki Y, Sasamura H, Hayashi K, Ishiguro K, Takaishi H, Eur J Biochem 268: 1918 –1928, 2001.
Okada Y, D’Armiento JM, Saruta T, Itoh H. Absence of gelatinase 131. Turck J, Pollock AS, Lee LK, Marti HP, Lovett DH. Matrix metal-
(MMP-9) or collagenase (MMP-13) attenuates adriamycin-induced albu- loproteinase 2 (gelatinase A) regulates glomerular mesangial cell prolif-
minuria and glomerulosclerosis. Nephron Exp Nephrol 115: e22–e32, eration and differentiation. J Biol Chem 271: 15074 –15083, 1996.
2010. 132. Tveita A, Rekvig OP, Zykova SN. Glomerular matrix metalloprotei-
112. Sanders JS, Huitema MG, Hanemaaijer R, van Goor H, Kallenberg nases and their regulators in the pathogenesis of lupus nephritis. Arthritis
CG, Stegeman CA. Urinary matrix metalloproteinases reflect renal Res Ther 10: 229, 2008.
damage in anti-neutrophil cytoplasm autoantibody-associated vasculitis. 133. Tveita AA, Rekvig OP, Zykova SN. Increased glomerular matrix
Am J Physiol Renal Physiol 293: F1927–F1934, 2007. metalloproteinase activity in murine lupus nephritis. Kidney Int 74:
113. Sanders JS, van Goor H, Hanemaaijer R, Kallenberg CG, Stegeman 1150 –1158, 2008.
CA. Renal expression of matrix metalloproteinases in human ANCA- 134. Urushihara M, Kagami S, Kuhara T, Tamaki T, Kuroda Y. Glomer-
associated glomerulonephritis. Nephrol Dial Transplant 19: 1412–1419, ular distribution and gelatinolytic activity of matrix metalloproteinases in
2004. human glomerulonephritis. Nephrol Dial Transplant 17: 1189 –1196,
114. Sen U, Rodriguez WE, Tyagi N, Kumar M, Kundu S, Tyagi SC. 2002.
Ciglitazone, a PPARgamma agonist, ameliorates diabetic nephropathy in 135. van der Zijl NJ, Hanemaaijer R, Tushuizen ME, Schindhelm RK,
part through homocysteine clearance. Am J Physiol Endocrinol Metab Boerop J, Rustemeijer C, Bilo HJ, Verheijen JH, Diamant M.
295: E1205–E1212, 2008. Urinary matrix metalloproteinase-8 and -9 activities in type 2 diabetic
115. Shin JI, Song KS, Kim H, Cho NH, Kim J, Kim HS, Lee JS. The gene subjects: A marker of incipient diabetic nephropathy? Clin Biochem 43:
expression profile of matrix metalloproteinases and their inhibitors in 635–639, 2010.
children with Henoch-Schonlein purpura. Br J Dermatol 164: 1348 – 136. Van Lint P, Libert C. Matrix metalloproteinase-8: cleavage can be
1355, 2011. decisive. Cytokine Growth Factor Rev 17: 217–223, 2006.

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org


Review
MMPs IN KIDNEY DISEASES F1361
137. Van Wart HE, Birkedal-Hansen H. The cysteine switch: a principle of 146. Xu X, Jackson PL, Tanner S, Hardison MT, Abdul Roda M, Blalock
regulation of metalloproteinase activity with potential applicability to the JE, Gaggar A. A self-propagating matrix metalloprotease-9 (MMP-9)
entire matrix metalloproteinase gene family. Proc Natl Acad Sci USA 87: dependent cycle of chronic neutrophilic inflammation. PLoS One 6:
5578 –5582, 1990. e15781, 2011.
138. Velasco G, Pendas AM, Fueyo A, Knauper V, Murphy G, Lopez- 147. Yamashita CM, Dolgonos L, Zemans RL, Young SK, Robertson J,
Otin C. Cloning and characterization of human MMP-23, a new matrix Briones N, Suzuki T, Campbell MN, Gauldie J, Radisky DC, Riches
metalloproteinase predominantly expressed in reproductive tissues and DW, Yu G, Kaminski N, McCulloch CA, Downey GP. Matrix metal-
lacking conserved domains in other family members. J Biol Chem 274: loproteinase 3 is a mediator of pulmonary fibrosis. Am J Pathol 179:
4570 –4576, 1999. 1733–1745, 2011.
139. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of 148. Yang J, Shultz RW, Mars WM, Wegner RE, Li Y, Dai C, Nejak K,
metalloproteinases: structure, function, and biochemistry. Circ Res 92: Liu Y. Disruption of tissue-type plasminogen activator gene in mice
827–839, 2003. reduces renal interstitial fibrosis in obstructive nephropathy. J Clin Invest
140. Wang D, Dai C, Li Y, Liu Y. Canonical Wnt ␤-catenin signaling 110: 1525–1538, 2002.
mediates transforming growth factor-␤1-driven podocyte injury and 149. Yang MX, Qu X, Kong BH, Lam QL, Shao QQ, Deng BP, Ko KH,

Downloaded from http://ajprenal.physiology.org/ at University of South Dakota on November 18, 2012


proteinuria. Kidney Int 80: 1159 –1169, 2011. Lu L. Membrane type 1-matrix metalloproteinase is involved in the
141. Wang X, Zhou Y, Tan R, Xiong M, He W, Fang L, Wen P, Jiang L, migration of human monocyte-derived dendritic cells. Immunol Cell Biol
Yang J. Mice lacking the matrix metalloproteinase-9 gene reduce renal 84: 557–562, 2006.
interstitial fibrosis in obstructive nephropathy. Am J Physiol Renal 150. Yao XM, Ye SD, Zai Z, Chen Y, Li XC, Yang GW, Wang YX, Chen
Physiol 299: F973–F982, 2010. K. Simvastatin protects diabetic rats against kidney injury through the
142. Weisbord SD, Chen H, Stone RA, Kip KE, Fine MJ, Saul MI, suppression of renal matrix metalloproteinase-9 expression. J Endocrinol
Palevsky PM. Associations of increases in serum creatinine with mor- Invest 33: 292–296, 2010.
tality and length of hospital stay after coronary angiography. J Am Soc 151. Yen JH, Khayrullina T, Ganea D. PGE2-induced metalloproteinase-9
Nephrol 17: 2871–2877, 2006. is essential for dendritic cell migration. Blood 111: 260 –270, 2008.
143. Wickstrom SA, Alitalo K, Keski-Oja J. Endostatin signaling and 152. Zeisberg M, Duffield JS. Resolved: EMT produces fibroblasts in the
regulation of endothelial cell-matrix interactions. Adv Cancer Res 94: kidney. J Am Soc Nephrol 21: 1247–1253, 2010.
197–229, 2005. 153. Zeisberg M, Khurana M, Rao VH, Cosgrove D, Rougier JP, Werner
144. Williams JM, Zhang J, North P, Lacy S, Yakes M, Dahly-Vernon A, MC, Shield CF 3rd, Werb Z, Kalluri R. Stage-specific action of matrix
Roman RJ. Evaluation of metalloprotease inhibitors on hypertension metalloproteinases influences progressive hereditary kidney disease.
and diabetic nephropathy. Am J Physiol Renal Physiol 300: F983–F998, PLoS Med 3: e100, 2006.
2011. 154. Zheng G, Lyons JG, Tan TK, Wang Y, Hsu TT, Min D, Succar L,
145. Wong W, DeVito J, Nguyen H, Sarracino D, Porcheray F, Dargon I, Rangan GK, Hu M, Henderson BR, Alexander SI, Harris DC.
Pelle PD, Collins AB, Tolkoff-Rubin N, Smith RN, Colvin R, Zorn E. Disruption of E-cadherin by matrix metalloproteinase directly mediates
Chronic humoral rejection of human kidney allografts is associated with epithelial-mesenchymal transition downstream of transforming growth
MMP-2 accumulation in podocytes and its release in the urine. Am J factor-beta1 in renal tubular epithelial cells. Am J Pathol 175: 580 –591,
Transplant 10: 2463–2471, 2010. 2009.

AJP-Renal Physiol • doi:10.1152/ajprenal.00037.2012 • www.ajprenal.org

You might also like