You are on page 1of 9

Christos Chatziantoniou and Jean-Claude Dussaule

Am J Physiol Renal Physiol 289:227-234, 2005. doi:10.1152/ajprenal.00453.2004

You might find this additional information useful...

This article cites 78 articles, 41 of which you can access free at:
http://ajprenal.physiology.org/cgi/content/full/289/2/F227#BIBL

This article has been cited by 9 other HighWire hosted articles, the first 5 are:
Pharmacological blockade of B2-kinin receptor reduces renal protective effect of
angiotensin-converting enzyme inhibition in db/db mice model
M. Buleon, J. Allard, A. Jaafar, F. Praddaude, Z. Dickson, M.-T. Ranera, C. Pecher, J.-P.
Girolami and I. Tack
Am J Physiol Renal Physiol, May 1, 2008; 294 (5): F1249-F1256.
[Abstract] [Full Text] [PDF]
Immune suppression blocks sodium-sensitive hypertension following recovery from
ischemic acute renal failure
K. R. Pechman, D. P. Basile, H. Lund and D. L. Mattson
Am J Physiol Regulatory Integrative Comp Physiol, April 1, 2008; 294 (4): R1234-R1239.
[Abstract] [Full Text] [PDF]

Downloaded from ajprenal.physiology.org on August 4, 2010


Molecular mechanisms linking wound inflammation and fibrosis: knockdown of
osteopontin leads to rapid repair and reduced scarring
R. Mori, T. J. Shaw and P. Martin
J. Exp. Med., January 21, 2008; 205 (1): 43-51.
[Abstract] [Full Text] [PDF]

Treatment targets in renal fibrosis


P. Boor, K. Sebekova, T. Ostendorf and J. Floege
Nephrol. Dial. Transplant., December 1, 2007; 22 (12): 3391-3407.
[Full Text] [PDF]

Oncostatin M-induced effects on EMT in human proximal tubular cells: differential role of
ERK signaling
V. Pollack, R. Sarkozi, Z. Banki, E. Feifel, S. Wehn, G. Gstraunthaler, H. Stoiber, G. Mayer, R.
Montesano, F. Strutz and H. Schramek
Am J Physiol Renal Physiol, November 1, 2007; 293 (5): F1714-F1726.
[Abstract] [Full Text] [PDF]

Medline items on this article's topics can be found at http://highwire.stanford.edu/lists/artbytopic.dtl


on the following topics:
Biochemistry .. Angiotensin II
Biochemistry .. Vasoactive Peptide
Oncology .. Growth Factors
Pharmacology .. Growth Substances
Medicine .. Fibrosis
Physiology .. Humans
Updated information and services including high-resolution figures, can be found at:
http://ajprenal.physiology.org/cgi/content/full/289/2/F227

Additional material and information about AJP - Renal Physiology can be found at:
http://www.the-aps.org/publications/ajprenal

This information is current as of August 4, 2010 .

AJP - 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 12 times a year
(monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2005 by the
American Physiological Society. ISSN: 0363-6127, ESSN: 1522-1466. Visit our website at http://www.the-aps.org/.
Am J Physiol Renal Physiol 289: F227–F234, 2005;
doi:10.1152/ajprenal.00453.2004. Invited Review

Insights into the mechanisms of renal fibrosis: is it possible


to achieve regression?
Christos Chatziantoniou and Jean-Claude Dussaule
Institut National de la Santé et de la Recherche Médicale Unité 702, Hôpital Tenon,
and AP-HP, Laboratoire de Physiologie, Faculté de Médecine St. Antoine, Paris, France

Chatziantoniou, Christos, and Jean-Claude Dussaule. Insights into the mech-


anisms of renal fibrosis: is it possible to achieve regression?. Am J Physiol Renal
Physiol 289: F227–F234, 2005; doi:10.1152/ajprenal.00453.2004.—Recent evi-
dence suggests that the progression of renal fibrosis is a reversible process, at least
in experimental models. The present review summarizes the new insights concern-
ing the mechanisms of progression and regression of renal disease and examines
this novel evidence under the light of feasibility and transfer to human nephropa-
thies. The involved mechanisms are discussed with particular emphasis on the
fibrotic role of vasoactive peptides such as angiotensin II and endothelin and
growth factors such as transforming growth factor (TGF)-␤. The possibility of

Downloaded from ajprenal.physiology.org on August 4, 2010


regression is introduced by presenting the in vivo efficiency of antihypertensive
treatments and of systems that antagonize the fibrogenic action of TGF-␤ such as
bone morphogenic protein-7 and HGF. Finally, we provide a brief description of
the promising future directions and clinical considerations about the applications of
the experimental data to humans.
chronic renal failure; collagen; angiotensin; growth factors; metalloproteinases

RENAL FIBROSIS IS DEFINED by the abnormal accumulation of slowing down the progression and to try to achieve regression
extracellular matrix that replaces normal kidney structures. of renal fibrosis and restoration of renal structure. The forma-
Contrary to the kidney, regression of fibrosis is well estab- tion of extracellular matrix follows schematically three distinct
lished in other tissues, such as heart, liver, and skin (19, 20, steps: synthesis of the mature protein (a complex process
23). Because renal fibrosis appears irrespective of the under- starting at synthesis of propeptides, followed by cleavage and
lying disease (hypertension, diabetes, infection, inflammation assembly of the peptidic chains to form the mature protein);
of renal blood vessels and glomeruli, kidney stones, and cysts) extracellular stabilization with the aid of matrix “receptors”
and originating renal compartment (renal vessels, glomeruli, and cross-linking enzymes; and catabolism by specific protein-
tubules), it is generally believed that is under the control of a ases. Fibrosis is developed when the rate of synthesis and
common final physiopathological pathway independent of the stabilization of matrix is exaggerated and/or when the capacity
primary cause. Thus identifying and targeting the systems of degradation is decreased. In this review, we will present data
participating in this pathway may lead to efficient treatments obtained mainly in experimental rodent models that provided
against renal fibrosis and failure regardless of the initiating novel insights into the cellular mechanisms participating in the
pathology. Over the last few years, studies in humans gave progression and identified possible targets to achieve regres-
hope by showing that antihypertensive treatments can indeed sion of renal fibrosis.
induce relative renoprotection. After an initial observation that
blockade of the angiotensin II action protects against the INHIBITION OF INFLAMMATION-OXIDATIVE STRESS
progression of renal insufficiency in patients with various renal Glomerular or interstitial inflammation is thought to play an
diseases (such as glomerulopathies, interstitial nephritis, ne- important role in the initiation of renal fibrosis. Accordingly,
phrosclerosis, polycystic kidney disease, or diabetic nephrop- the strategies used were either against inflammation as a
athy) (54), large-scale studies concerning hypertensive patients general pathology (corticosteroid treatment) or, more specifi-
with type 2 diabetes demonstrated the renoprotective efficiency cally, against cellular differentiation (fibroblasts, activated
of angiotensin II antagonism (13, 51, 60). In these studies, mesangial cells) and/or the proinflammatory agents released
angiotensin II receptor antagonism reduced microalbuminuria during inflammation (cytokines or chemokines). Treatment
and slowed the rate of decline of the filtration rate; these with corticosteroids has been used in some fibrotic nephropa-
renoprotective effects were independent of the blood pressure thies (IgA) with relative success (3, 39, 64), whereas steroid
decrease, supporting the notion of dissociation between sys- treatment had no beneficial effect on patients of African Amer-
temic and local actions of angiotensin II in kidneys. Despite ican genetic background with focal segmental glomeruloscle-
these promising results, the challenge remains to go beyond rosis (18).
The blockade of fibroblast activation was mainly tested in
Address for reprint requests and other correspondence: C. Chatziantoniou,
experimental forms of nephropathy. Preventive treatment with
INSERM U702, 4 Rue de la Chine, Hôpital Tenon, 75020 Paris, France an interleukin-1 receptor antagonist or with an anti-intercellu-
(e-mail: christos.chatziantoniou@tnn.ap-hop-paris.fr). lar adhesion molecule-1 (ICAM-1) antibody protected rat kid-
http://www.ajprenal.org 0363-6127/05 $8.00 Copyright © 2005 the American Physiological Society F227
Invited Review
F228 REGRESSION OF RENAL FIBROSIS

neys from the development of fibrosis in the anti-glomerular actions of vasoconstrictors indicated that these peptides could
basement membrane model of nephropathy (48, 56). induce, stimulate, or control a variety of cellular pathways in
However, when the treatment with anti-ICAM-1 started after parallel and/or independently of their constrictor action (such
the development of the fibrosis, protection of the renal struc- as cell migration, hypertrophy, proliferation, inflammation,
ture was rather limited. More recently, an antisense oligodes- generation of reactive oxygen species, transactivation of
oxynucleotide strategy against ICAM-1 was used to prevent growth factor receptor) that are directly involved in protein
ischemia-reperfusion injury and to prolong renal isograft sur- synthesis and extracellular matrix formation. Two of the most
vival in rats (16, 21). Although promising as an approach, there studied peptides in this field are angiotensin II and endothelin.
are several technical problems to be solved before testing of its
applicability to other forms of renal injury and making this The Renin-Angiotensin System
strategy available in humans.
The role of tumor necrosis factor-␣ (TNF-␣) in the progres- We have presented above the studies in diabetic patients that
sion of renal scarring (especially in the model of ureteral gave promising results regarding relative renoprotection by
obstruction) is well documented (46). Knockout mice for either blocking the renin-angiotensin system. A slight regression of
the TNFR1 or TNFR2 receptors displayed decreased scores of sclerosis was also observed after long-term treatment with
collagen IV and matrix expansion compared with wild-type angiotensin receptor antagonists in the aging rat kidney (53).
controls 5 days after unilateral ureteral obstruction (32). The The proposed mechanisms involved increased tubular epithe-
preservation of renal structure was better when double TNF lial cell turnover and inhibition of plasminogen activator in-
receptor knockouts were used and was further increased when hibitor-1 (PAI-1) expression. Administration of an angiotensin

Downloaded from ajprenal.physiology.org on August 4, 2010


the action of angiotensin II was pharmacologically or geneti- enzyme inhibitor accelerated the obstruction release-induced
cally inhibited (33). It is thus interesting to investigate whether regression in the model of unilateral ureteral obstruction (47).
the different techniques of anti-TNF-␣ therapy that have been We have addressed the issue of renoprotection by investigating
lately developed for rheumatoid arthritis (24) can also extend the mechanisms by which AT1 receptor antagonists make
their field of application to treat renal fibrotic lesions. possible the regression of renal vascular and glomerular fibro-
The generation of free radicals (reactive oxygen and nitro- sis in an experimental model of hypertension-associated renal
gen species) is considered as initiator of inflammatory response failure [nitric oxide (NO) deficiency model] (9). Figure 1
in a variety of tissues, including the kidney. The link between summarizes the results obtained in this study. After 1 mo of
oxidative stress and renal infiltration of immune cells in the hypertension, renal function severely declined as evidenced by
kidney has been nicely reviewed recently (62), and we will increased levels of proteinuria and plasma creatinine, and the
avoid unnecessary repetition. We will briefly mention that mortality rate accounted for 20% of animals. The decline of
treatment with antioxidants gave impressive results concerning renal function was accompanied by an exaggerated gene and
vascular and renal protection in experimental animal models protein expression of transforming growth factor (TGF)-␤,
(regression of vascular remodeling, improvement of endothe- collagen I, and collagen IV within the renal vasculature and an
lial function, inhibition of inflammation, and decrease in blood abnormal accumulation of extracellular matrix in glomeruli. In
pressure). This experimental evidence produced considerable addition, activities of matrix metalloproteinases 2 and 9 were
interest because of the possibilities that therapies targeting increased several-fold in glomeruli. At this phase of renal
reactive oxygen species might be used against vascular injury failure, administration of an angiotensin II receptor antagonist
and end-organ damage. However, long-term treatment with for 1 wk decreased collagen I, collagen IV, and TGF-␤ gene
antioxidant vitamins gave disappointing results in large-scale and protein expression without affecting the increased level of
studies in patients with cardiovascular disease and/or diabetes metalloproteinases 2 and 9 activities in glomeruli. These cel-
(31, 75). It is possible that the current clinically available lular alterations were accompanied by a gradual regression of
antioxidants lack the efficacy necessary for use in long-term glomerulosclerosis, a partial restoration of renal function, and
treatments. It is also possible that oxidative stress and inflam- an arrest of mortality. When the antihypertensive treatment
mation are very important events early in the developing phase was extended to 1 mo, all cellular, structural, and functional
of vascular and renal disease, whereas they participate little parameters of the kidney were normalized, indicating that the
during the established or advanced phases of organ failure. In progression of renal vascular fibrosis is a reversible process.
this case, the efficiency of an anti-inflammatory or antioxidant Based on these data, we proposed that the mechanism of the
strategy could largely depend on the degree of the evolution of regression was dual: inhibition of the exaggerated synthesis of
the disease. extracellular matrix (due to blockade of the angiotensin II-
TGF-␤ pathway) and increased rate of matrix degradation (due
BLOCKADE OF VASOCONSTRICTOR PEPTIDE ACTION
to metalloproteinase activity, probably associated with the
degree of fibrosis and independent of angiotensin blockade).
The appearance of renal sclerotic injury and the develop- Subsequently, other investigators confirmed the reversibility of
ment of renal failure are frequent complications of hyperten- fibrotic process after angiotensin II blockade in the model of
sion (72). Until recently, the classic role attributed to vasocon- acute nephronic reduction (1, 2). Eight weeks after surgery,
strictor peptides was that of the “increased pressure effector.” animals were treated for 1 mo with an angiotensin-converting
In this view, the abnormal extracellular matrix formation in enzyme (ACE) inhibitor. Morphological evaluation indicated
mesangial and vascular smooth muscle cells and the develop- regression of preexisting glomerular, tubular, and vascular
ment of renal vascular and glomerular sclerosis were pheno- lesions and reversal of glomerular hypertrophy. The decreased
typic adaptations (remodeling) to the increased tension within number of podocytes, following renal ablation, was not re-
the renal vasculature. However, recent advances in the cellular stored by the pharmacological treatment, suggesting that glo-
AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org
Invited Review
REGRESSION OF RENAL FIBROSIS F229

Downloaded from ajprenal.physiology.org on August 4, 2010


Fig. 1. Prolonged deficiency of endogenous nitric oxide [induced by the nitric oxide synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME)] is associated
with the development of renal failure. Four weeks of L-NAME treatment produced a dramatic increase in proteinuria and creatininemia that was accompanied
by increased levels of collagen I, transforming growth factor (TGF)-␤, and matrix metalloproteinases (MMPs) in renal vessels and glomeruli. Treatment of the
animals with an angiotensin receptor blocker (ARB; losartan) normalized renal function and reversed renal fibrosis by a dual mechanism: inhibition of collagen
synthesis (ANG II, TGF-␤ dependent) and degradation of extracellular matrix (MMP activity dependent). Adapted from Ref. 9.

merular regeneration largely depends on the degree of damage onstrated that endothelin gene and/or protein expression is
of glomerular podocytes. In agreement to this notion, mesan- increased during nephropathy and colocalizes with fibrotic
gial proliferation was reduced and interstitial changes were lesions. Inversely, pharmacological antagonism of endothelin
reversed after favorable treatment, whereas the number of receptors delayed the evolution and/or prevented renal failure.
sclerotic glomeruli remained unchanged in biopsies of IgA We investigated the efficiency of endothelin receptor antago-
nephropathy patients (39). nism to treat renal failure in the NO deficiency model. This
model was applied in a transgenic strain of mice harboring the
Endothelin
firefly luciferase reporter gene under the control of the pro-
Endothelin is also an important mediator of vascular and moter of the ␣2 chain, the collagen type I gene (12). Due to the
renal fibrosis (4, 15, 37, 67, 70) (Fig. 2). The aforementioned sensitivity of reporter gene assays, changes in the expression of
studies, performed in a variety of experimental models, dem- the collagen I gene were detected in a highly sensitive manner

Fig. 2. ANG II-dependent signaling pathways producing phe-


notypic changes in vascular, endothelial, mesangial, and epi-
thelial cells and leading to glomerular remodeling and to
alterations of renal structure and function. ET-1, endothelin-1;
PAI-1, plasminogen activator inhibitor-1; DAG, diacylglyc-
erol; IP3, inositol 1,4,5-trisphosphate.

AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org


Invited Review
F230 REGRESSION OF RENAL FIBROSIS

(8, 15). Because mice displayed slower kinetics compared with agent. BMP-7 is highly expressed in the kidney, and its genetic
rats for the development of hypertension and renal fibrosis, the deletion in mice leads to severe impairment of eye, skeletal,
induction of the pathology and the therapeutic treatment lasted and kidney development (38). Preventive or curative treatment
longer (20 and 10 wk, respectively) (7). Endothelin receptor with BMP-7 preserved or restored renal histology and renal
antagonism, introduced after the appearance of fibrotic lesions, function in a rat model of unilateral ureteral obstruction; these
reduced mortality, partially decreased the abnormal collagen effects of BMP-7 were slightly better compared with the
formation within the renal vasculature by inhibiting collagen I protection obtained with an ACE inhibitor (40, 55). In addition,
gene activation, and did not alter the increased levels of blood systemic administration of recombinant human BMP-7 led to
pressure. The treated animals displayed a less severe degree of repair severely damaged renal tubular epithelial cells and to
glomerular lesions even compared with those at the beginning improve renal function and survival in mice with nephrotoxic
of the treatment, thus suggesting that renal vascular fibrosis serum nephritis and in two genetic models for chronic renal
could regress independently of the systemic hemodynamics injury and fibrosis (mice deficient in the ␣3-chain of type IV
(7). In subsequent studies, we investigated whether repair of collagen and MRL/MpJlpr/lpr lupus mice) (76, 77). These
endothelial dysfunction could restore renal failure in this improvements were attributed to the decreased synthesis of
model. Reactivation of endogenous NO synthesis substantially profibrotic molecules and to the increased activity of matrix
improved blood pressure and renal structure, suggesting that metalloproteinase (MMP) 2. These results indicate the poten-
correction of the initiating cause (NO inhibition) could also be tial of BMP-7 to reverse the TGF-␤-induced injury and to
an efficient therapy (Placier S, Boffa JJ, Chatziantoniou C, and repair renal tissue in a variety of experimental models.
Dussaule JC, unpublished observations). An additional endogenous peptide antagonizing the effects

Downloaded from ajprenal.physiology.org on August 4, 2010


of TGF-␤ is the HGF. The therapeutic potential of HGF has
INHIBITION OF TGF-␤ ACTION been reviewed recently (52). We will briefly mention that
TGF-␤ is an agent promoting extracellular matrix synthesis delayed administration of recombinant HGF retarded the pro-
and is considered to play a major role as mediator of the gression of renal lesions by blunting the myofibroblast accu-
fibrogenic action of several vasoconstrictor peptides, especially mulation and collagen deposition within the kidney. This
that of angiotensin II (10) (Fig. 2). Initially, the strategy aimed action of HGF is likely related to a mitogen-activated protein
to block the action of TGF-␤ at the systemic (decorin, blocking kinase-dependent blockade of TGF-␤-induced nuclear translo-
anti-body) or receptor (soluble receptors) level. To this end, cation of SMADs (73, 74). Continuous infusion of HGF in the
preventive protocols gave encouraging results: exogenous ad- rat remnant kidney model decreased tubulointerstitial collagen
ministration of decorin or prolonged infusion of anti-TGF-␤ deposition and renal fibrosis; in contrast, blocking endogenous
antibody protected rat kidneys from the development of glo- HGF by an anti-HGF neutralizing antibody increased intersti-
merulonephritis (11), and genetically engineered overexpres- tial collagen and the severity of renal fibrosis. Interestingly,
sion of decorin in the skeletal muscle of rats inhibited the HGF infusion increased, and conversely HGF antibody sup-
fibrogenic action of TGF-␤ and protected kidneys against pressed, the in situ gelatinolytic activity in remnant kidneys,
glomerulosclerosis (41). Alternatively, use of soluble receptors thus supporting the hypothesis that activation of MMP2 and 9
of TGF-␤ prevented the development of renal and cardiac plays a protective role against renal fibrosis (29).
fibrosis (45, 71). However, these approaches presented impor- PROMISING STRATEGIES WAITING TO BE TESTED
tant limitations (increased concentrations of decorin lack the
anti-TGF-␤ specificity and can produce secondary effects, The above-mentioned strategies (summarized on Table 1)
whereas the cost and method of delivery, intravenous infusion, are variations targeting a common axis: vasoconstrictor-TGF-
of soluble receptors make long-term treatment unrealistic) that ␤-induced fibrosis. To date, they are the only ones that have
tempered the initial enthusiasm and did not allow their exten- demonstrated their efficiency in inducing a reversal of fibrosis
sion to curative protocols. in experimental models. However, alternative approaches have
For these reasons, strategies have been proposed using been proposed over the last few years that appear to act
agents that target the signaling pathway of TGF-␤ and block its independently of the vasoconstrictor-TGF-␤ fibrotic pathway.
fibrogenic action. Bone morphogenic protein-7 (BMP-7) and These strategies gave impressive results in preventing progres-
HGF are two among these systems that have been tested sion and renal structural and functional alterations. Neverthe-
experimentally in vivo. BMP-7, a 35-kDa homodimeric protein less, it remains to be tested whether they will be efficient in
and member of the TGF-␤ superfamily that antagonizes the promoting regression. In the following sections, we will present
action of TGF-␤, has been used with success as antifibrotic some of these promising (but not tested yet) hypotheses.

Table 1. Therapeutic strategies that regressed renal fibrosis and improved renal function
Model Treatment Species Reference(s)

5/6 Nephrectomy Angiotensin II inhibition Rat 1, 2


L-NAME Angiotensin II antagonism Rat 9
Ureteral obstruction Angiotensin II inhibition Rat 47
Aging Angiotensin II antagonism Rat 53
L-NAME Endothelin antagonism Transgenic mouse 7
Nephrotoxic serum nephritis BMP-7 Mouse 77
L-NAME, NG-L-arginine methyl ester; BMP-7, bone morphometric protein-7.

AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org


Invited Review
REGRESSION OF RENAL FIBROSIS F231
Tyrosine Kinase Receptors of Growth Factors tion, clinical follow-up revealed a correlation between gene
expression data and the progression of renal disease and
A relatively novel concept regarding the signaling pathways allowed segregation of the biopsies into progressive or stable
of potent vasoconstrictors is that they can phosphorylate the disease course based on gene expression profiles (35). Binding
receptors of growth factors (5) (Fig. 2). Increased expression or of the extracellular matrix is, however, a complex process
activation of these receptors has been observed during the requiring participation of several families of proteins that are
development of several forms of nephropathies (46), thus also essential for many physiological properties of cells. In-
making attractive the hypothesis that this activation could hibiting collagen I, collagen III, or fibronectin anchorage, for
participate in the progression of renal fibrotic disease. To this instance, would have a therapeutic sense if a specific receptor
end, nuclease-resistant, high-affinity aptamers that neutralized (or receptor-like protein) could be found and if the inhibition of
the effects of PDGF-B inhibited glomerular and interstitial the binding with this inhibitor does not alter normal cell
fibrosis in a rat model of mesangioproliferative glomeruloscle- functions.
rosis (59). We tested the interaction among vasoconstrictors,
epidermal growth factor (EGF) receptor transactivation, and Degradation of Extracellular Matrix Components
the collagen I gene, and we found that endothelin induced a
rapid phosphorylation of the EGF receptor that mediated col- Extracellular matrix degradation is another complex process
lagen I gene activation in the renal cortex. Moreover, we in which the family of MMPs plays a major role. Several recent
observed that EGF receptor was activated within glomeruli studies correlated the development of renal fibrosis with acti-
concomitantly with the development of glomerulosclerosis in vation of metalloproteinases (34, 63). Studies testing the effi-

Downloaded from ajprenal.physiology.org on August 4, 2010


the NO deficiency model. Use of an EGF receptor-tyrosine ciency of MMP inhibitor treatment during renal fibrosis gave
kinase inhibitor in a preventive way normalized MAPK acti- contradictory results. The initially proposed antibiotics such as
vation, inhibited the abnormal increase in collagen I gene tetracyclin and doxycyclin as inhibitors of MMPs lack the
expression, decreased proteinuria and creatininemia, and pre- necessary specificity. Administration of a more specific met-
vented the development of renal vascular and glomerular alloproteinase inhibitor (batimastat) significantly reduced cyst
fibrosis (26, 28). From these studies it appears that blockade or number and kidney weight in the (cy/⫹) rat model of autoso-
antagonism of growth factors offers an interesting perspective mal-dominant polycystic kidney disease (57). In contrast, ba-
on a therapeutic strategy against fibrosis. However, an eventual timastat had no protective effect in the renal ische-
problem for transfer to humans is the side effect(s) of such mia-reperfusion injury model in rats (78). As noted above, our
treatments. The well-known toxicity of long-term blockade of data suggested an antifibrotic-protective role of MMPs in the
growth factor action probably excludes life-long treatment. NG-nitro-L-arginine methyl ester model (9). The recent obser-
Thus it is more conceivable to combine them for a short period vations that MMPs can cleave other than collagen proteins (50)
with “traditional” chronic renoprotective therapy. and that participate in vascular remodeling (49) make the issue
even more complex. Thus the debate continues as to whether
Stabilization of the Extracellular Matrix MMPs are or are not beneficial for regression of fibrosis, and
additional studies are necessary to define the role of MMP in
Anchorage and stabilization of the extracellular matrix are
renal fibrosis.
other steps that can eventually serve as the targets for thera-
peutic intervention. Tissue transglutaminase 2 belongs to the Agents Associated with Angiotensin II Action: Plasminogen
enzymes contributing to the stabilization of the extracellular Activator Inhibitor-1 and Aldosterone
matrix by forming ␥-glutamyl-lysine cross-linking. Its activa-
tion was observed within the renal tissue in two experimental PAI-1, an enzyme inhibiting the plasminogen proteolysis to
models of renal fibrosis (subtotal nephrectomy and diabetic plasmin, also reduces MMP activation; increased PAI-1 levels
nephropathy) (43, 65). Subsequent studies observed a several- have been observed in experimental nephropathies or during
fold increase in tissue transglutaminase expression in human aging (22, 53). Inversely, genetic invalidation of PAI-1 expres-
renal biopsy samples from a range of chronic renal diseases sion protected animals from the development of vascular or
(42). It is possible that inhibition of transglutaminase activity renal interstitial fibrosis (44, 58). The activation of PAI-1
will make the matrix more fragile and thus easier to degrade during fibrotic mechanisms appears to be due to the action of
the abnormal extracellular matrix. Supporting this notion, a angiotensin II, and treatment with angiotensin inhibitors or
specific inhibitor of tissue transglutaminase normalized glu- receptor antagonists was accompanied by decreased levels of
cose-induced deposition of extracellular matrix proteins in PAI-1 within the kidney (27). Based on these results, several
renal proximal tubular epithelial cells (66). It would be inter- investigators have proposed to inhibit PAI-1 as a means of
esting to test whether a similar effect can be produced in vivo. treating renal fibrosis. However, PAI-1 null mice displayed a
Integrins are considered as extracellular matrix receptors. worse degree of renal failure in the anti-glomerular basement
Expansion of mesangial matrix and podocyte foot process membrane model, indicating that PAI-1 plays a more complex
effacement were attenuated in Alport mice that were also null role (36).
for integrin-␣1 expression (17). In addition, fibronectin accu- Until recently, aldosterone has been considered as a rela-
mulation and binding to integrin ␣5␤1 were reduced and tively minor component of angiotensin-induced fibrogenic ac-
glomerular sclerotic lesions were attenuated by heparin treat- tion. Recent studies, however, suggested that inhibition of
ment in a murine model of lupus nephritis (69). Interestingly, aldosterone could have a direct nephroprotective effect inde-
integrin-␤4, one of the genes involved in cell-cell contact, pendently of angiotensin II. Thus early administration of an-
showed differential regulation in inflammation and fibrosis in tagonists of aldosterone receptors prevented glomerular dam-
tubulointerstitial samples from routine renal biopsies. In addi- age and proteinuria in the stroke-prone spontaneously hyper-
AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org
Invited Review
F232 REGRESSION OF RENAL FIBROSIS

tensive rat (61), 5⁄6 nephrectomy (30), radiation injury (14), CLINICAL PERSPECTIVES
aldosterone/salt-induced hypertension (6), cyclosporine A-in-
Contrary to the initial belief in nonrepaired damage, the
duced nephrotoxicity (25), and unilateral ureteral obstruction
reversibility of renal fibrosis and the associated improvement
(68) models in rodents. It would be interesting to test the
of renal function have been clearly demonstrated in several
efficiency of aldosterone blockade in curative protocols and to
experimental models. Figure 3 summarizes the factors that
compare this action (if any) to ACE inhibition or angiotensin
have been implied in the development of renal fibrosis and its
receptor blockade.
regression. In human nephropathies, however, the actual treat-
ments target mainly blood pressure control and do not seem
SALIENT QUESTIONS AND AREAS FOR FUTURE RESEARCH capable of offering more relief than a delay in the rate of
The above-presented studies indicate that impressive progress progression. For this reason, it is very important to combine
has been made in our understanding of the mechanisms control- novel treatments targeting specifically fibrogenesis with classic
ling progression and regression of renal fibrosis. From these antihypertensive agents. Among the strategies that have been
studies, however, emerged several new and important questions explored over the last few years, those antagonizing the fibrotic
that need to be addressed in future research. One of the most action of TGF-␤ appear to give the most promising results.
challenging is the link between reversal of fibrosis and refunction Additional studies elucidating some puzzling issues of progres-
of the kidney; in other words, is it enough to eliminate the sion-regression are still necessary to advance our knowledge.
excessive accumulation of extracellular matrix protein to make A key issue is the existence and the definition of a no-return
point. The current belief is consistent with the notion that the

Downloaded from ajprenal.physiology.org on August 4, 2010


nephrons work well again? Or, is there a “no-return point” and, if
so, how can we define this point? Although most investigators no-return point is associated with phenotypic alterations of
agree that the no-return point is directly associated to podocyte podocytes. If so, it is important to identify its molecular,
loss, there is no clear definition of the irreversible stage in terms cellular, and mechanistic characteristics and to try to find
of quantity (number of podocytes), quality (kind of functional or markers detecting or, even better, predicting these alterations
phenotype alteration), and exclusivity (interactions with other as early as possible.
renal cells). Each of these axes of definition raises numerous other The following lines, extracted from the poem “Stopping By
questions: can the irreversible loss of function be attributed to the Woods on a Snowy Evening,” by Robert Frost, could apply to
disappearance or (inversely) to overexpression of a cytoskeletal or the issue of treating chronic renal failure and regressing fibrosis
structural protein? Is the loss of other renal structures (for in- on the eve of the 21st century:
stance, glomerular or interstitial capillaries) reparable? How can The woods are lovely, dark, and deep,
injured tubular epithelial cells recover? Do they require the re- But I have promises to keep,
cruitment of differentiating or stem cells? We don’t believe that And miles to go before I sleep,
the above-raised questions will remain unanswered for a long And miles to go before I sleep
period, given the state-of-the-art techniques that are available to Although it would be difficult to predict the length of the road
investigators today (genetically engineered animals, genomics, remaining, the recently emerging data leave no doubt that
proteomic analysis). promises will be kept.

Fig. 3. Progression and regression of renal fibrosis are dynamic processes under the control of profibrotic (right) and antifibrotic (left) systems. Strategies that
gave promising results as preventive treatments but have yet to be tested for their curative efficiency are the final 5 entries at the bottom right.

AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org


Invited Review
REGRESSION OF RENAL FIBROSIS F233
GRANTS 18. Crenshaw G, Bigler S, Salem M, and Crook ED. Focal segmental
glomerulosclerosis in African Americans: effects of steroids and angio-
This work was financially supported by the Institut National de la Santé et
tensin converting enzyme inhibitors. Am J Med Sci 319: 320 –325, 2000.
de la Recherche Médicale and the Faculté de Médecine St. Antoine.
19. Delanian S, Porcher R, Balla-Mekias S, and Lefaix JL. Randomized,
placebo-controlled trial of combined pentoxifylline, and tocopherol for
REFERENCES regression of superficial radiation-induced fibrosis. J Clin Oncol 21:
2545–2550, 2003.
1. Adamczak M, Gross ML, Amann K, and Ritz E. Reversal of glomerular 20. Diez J, Querejeta R, Lopez B, Gonzalez A, Larman M, and Martinez
lesions involves coordinated restructuring of glomerular microvasculature. Ubago JL. Losartan-dependent regression of myocardial fibrosis is asso-
J Am Soc Nephrol 15: 3063–3072, 2004. ciated with reduction of left ventricular chamber stiffness in hypertensive
2. Adamczak M, Gross ML, Krtil J, Koch A, Tyralla K, Amann K, and patients. Circulation 105: 2512–2517, 2002.
Ritz E. Reversal of glomerulosclerosis after high-dose enalapril treatment 21. Dragun D, Lukitsch I, Tullius SG, Qun Y, Park JK, Schneider W,
in subtotally nephrectomized rats. J Am Soc Nephrol 14: 2833–2842, Luft FC, and Haller H. Inhibition of intercellular adhesion molecule-1
2003. with antisense deoxynucleotides prolongs renal isograft survival in the rat.
3. Ando Y, Moriyama T, Oka K, Takatsuji K, Miyazaki M, Akagi Y, Kidney Int 54: 2113–2122, 1998.
Kawada N, Isaka Y, Izumi M, Yokoyama K, Yamauchi A, Horio M, 22. Eddy A and Giachelli CM. Renal expression of genes that promote
Ando A, Ueda N, Sobue K, Imai E, and Hori M. Enhanced interstitial interstitial inflammation and fibrosis in rats with protein-overload protein-
expression of caldesmon in IgA nephropathy and its suppression by uria. Kidney Int 47: 1546 –1557, 1995.
glucocorticoid-heparin therapy. Nephrol Dial Transplant 14: 1408 –1417, 23. Farci P, Roskams T, Chessa L, Peddis G, Mazzoleni AP, Scioscia R,
1999. Serra G, Lai ME, Loy M, Caruso L, Desmet V, Purcell RH, and
4. Benigni A, Zoja C, Corna D, Orisio S, Facchinetti D, Benati L, and Balestrieri A. Long-term benefit of interferon ␣ therapy of chronic
Remuzzi G. Blocking both type A and B endothelin receptors in the hepatitis D: regression of advanced hepatic fibrosis. Gastroenterology
kidney attenuates renal injury and prolongs survival in rats with remnant

Downloaded from ajprenal.physiology.org on August 4, 2010


126: 1740 –1749, 2004.
kidney. Am J Kidney Dis 27: 416 – 423, 1996.
24. Feldmann M. Development of anti-TNF therapy for rheumatoid arthritis.
5. Berk BC. Angiotensin II signal transduction in vascular smooth muscle:
Nature Rev Immunol 2: 364 –371, 2002.
pathways activated by specific tyrosine kinases. J Am Soc Nephrol 10,
25. Feria I, Pichardo I, Juarez P, Ramirez V, Gonzalez MA, Uribe N,
Suppl 11: S62–S68, 1999.
Garcia-Torres R, Lopez-Casillas F, Gamba G, and Bobadilla NA.
6. Blasi ER, Rocha R, Rudolph AE, Blomme EA, Polly ML, and McMa-
Therapeutic benefit of spironolactone in experimental chronic cyclospor-
hon E. Aldosterone/salt induces renal inflammation and fibrosis in hyper-
ine A nephrotoxicity. Kidney Int 63: 43–52, 2003.
tensive rats. Kidney Int 63: 1791–1800, 2003.
26. Flamant M, Tharaux PL, Placier S, Henrion D, Coffman T, Chatzi-
7. Boffa JJ, Tharaux PL, Dussaule JC, and Chatziantoniou C. Regression
antoniou C, and Dussaule JC. Epidermal growth factor transactivation
of renal vascular fibrosis by endothelin receptor antagonism. Hypertension
mediates the tonic and fibrogenic effects of endothelin in the aortic wall of
37: 490 – 496, 2001.
transgenic mice. FASEB J 17: 327–329, 2003.
8. Boffa JJ, Tharaux PL, Placier S, Ardaillou R, Dussaule JC, and
Chatziantoniou C. Angiotensin II activates collagen type I gene in the 27. Fogo AB. Renal fibrosis and the renin-angiotensin system. Adv Nephrol
renal vasculature of transgenic mice during inhibition of nitric oxide Necker Hosp 31: 69 – 87, 2001.
synthesis: evidence for an endothelin-mediated mechanism. Circulation 28. François H, Placier S, Flamant M, Tharaux PL, Chansel D, Dussaule
100: 1901–1908, 1999. JC, and Chatziantoniou C. Prevention of renal vascular and glomerular
9. Boffa JJ, Ying L, Placier S, Stefanski A, Dussaule JC, and Chatzi- fibrosis by epidermal growth factor receptor inhibition. FASEB J 18:
antoniou C. Regression of renal vascular and glomerular fibrosis: role of 926 –928, 2004.
angiotensin II receptor antagonism and metalloproteinases. J Am Soc 29. Gong R, Rifai A, Tolbert EM, Centracchio JN, and Dworkin LD.
Nephrol 14: 1132–1144, 2003. Hepatocyte growth factor modulates matrix metalloproteinases and plas-
10. Border WA and Noble NA. Interactions of transforming growth factor-␤ minogen activator/plasmin proteolytic pathways in progressive renal in-
and angiotensin II in renal fibrosis. Hypertension 31: 181–188, 1998. terstitial fibrosis. J Am Soc Nephrol 14: 3047–3060, 2003.
11. Border WA, Noble NA, Yamamoto T, Harper JR, Yamaguchi Y, 30. Greene EL, Kren S, and Hostetter TH. Role of aldosterone in the
Pierschbacher MD, and Ruoslahti E. Natural inhibitor of transforming remnant kidney model in the rat. J Clin Invest 98: 1063–1068, 1996.
growth factor-␤ protects against scarring in experimental kidney disease. 31. Gruppo Italiano per lo Studio della Sopravvivenza nell’Infarto Mio-
Nature 360: 361–364, 1992. cardico. Dietary supplementation with n-3 polyunsaturated fatty acids and
12. Bou-Gharios G, Garrett LA, Rossert J, Niederreither K, Eberspae- vitamin E after myocardial infarction: results of the GISSI-Prevenzione
cher H, Smith C, Black C, and de Crombrugghe B. A potent far- trial. Lancet 354: 447– 455, 1999.
upstream enhancer in the mouse pro␣2(I) collagen gene results expression 32. Guo G, Morrissey J, McCracken R, Tolley T, and Klahr S. Role of
of reporter genes in transgenic mice. J Cell Biol 134: 1333–1344, 1996. TNFR1 and TNFR2 receptors in tubulointerstitial fibrosis of obstructive
13. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, nephropathy. Am J Physiol Renal Physiol 277: F766 –F772, 1999.
Parving HH, Remuzzi G, Snapinn SM, Zhang Z, and Shahinfar S. 33. Guo G, Morrissey J, McCracken R, Tolley T, Liapis H, and Klahr S.
Effects of losartan on renal and cardiovascular outcomes in patients with Contributions of angiotensin II and tumor necrosis factor-␣ to the devel-
type 2 diabetes and nephropathy. N Engl J Med 345: 861– 869, 2001. opment of renal fibrosis. Am J Physiol Renal Physiol 280: F777–F785,
14. Brown NJ, Nakamura S, Ma L, Nakamura I, Donnert E, Freeman M, 2001.
Vaughan DE, and Fogo AB. Aldosterone modulates plasminogen acti- 34. Hayashi K, Horikoshi S, Osada S, Shofuda K, Shirato I, and Tomino
vator inhibitor-1 and glomerulosclerosis in vivo. Kidney Int 58: 1219 – Y. Macrophage-derived MT-1 MMp and increased MMP-2 activity are
1227, 2000. associated with glomerular damage in crescentic glomerulonephritis.
15. Chatziantoniou C, Boffa JJ, Ardaillou R, and Dussaule JC. Nitric J Pathol 191: 299 –305, 2000.
oxide inhibition induces early activation of type I collagen gene in renal 35. Henger A, Kretzler M, Doran P, Bonrouhi M, Schmid H, Kiss E,
resistance vessels and glomeruli in transgenic mice: role of endothelin. Cohen CD, Madden S, Porubsky S, Grone EF, Schlondorff D, Nelson
J Clin Invest 101: 2780 –2789, 1998. PJ, and Grone HJ. Gene expression fingerprints in human tubulointer-
16. Chen W, Bennett CF, Wang ME, Dragun D, Tian L, Stecker K, Clark stitial inflammation and fibrosis as prognostic markers of disease progres-
JH, Kahan BD, and Stepkowski SM. Perfusion of kidneys with unfor- sion. Kidney Int 65: 904 –917, 2004.
mulated “naked” intercellular adhesion molecule-1 antisense oligode- 36. Hertig A, Berrou J, Allory Y, Breton L, Commo F, Costa De Beau-
oxynucleotides prevents ischemic/reperfusion injury. Transplantation 68: regard MA, Carmeliet P, and Rondeau E. Type 1 plasminogen activator
880 – 887, 1999. inhibitor deficiency aggravates the course of experimental glomerulone-
17. Cosgrove D, Rodgers K, Meehan D, Miller C, Bovard K, Gilroy A, phritis through overactivation of transforming growth factor-␤. FASEB J
Gardner H, Kotelianski V, Gotwals P, Amatucci A, and Kalluri R. 17: 1904 –1906, 2003.
Integrin ␣1␤1 and transforming growth factor-␤1 play distinct roles in 37. Hocher B, Thöne-Reineke C, Rohmeiss P, Schmager F, Slowinski T,
Alport glomerular pathogenesis and serve as dual targets for metabolic Burst V, Siegmund F, Quertermous T, Bauer C, Neumayer HH,
therapy. Am J Pathol 157: 1649 –1659, 2000. Schleuning WD, and Theuring F. Endothelin-1 transgenic mice devel-

AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org


Invited Review
F234 REGRESSION OF RENAL FIBROSIS

ops glomerulosclerosis, interstitial fibrosis, and renal cysts but not hyper- prevents renal scarring in experimental glomerulonephritis. J Am Soc
tension. J Clin Invest 99: 1380 –1389, 1997. Nephrol 12: 909 –918, 2001.
38. Hogan BL. Bone morphogenic proteins in development. Curr Opin Genet 60. Parving HH, Lehnert H, Brochner-Mortensen J, Gomis R, Andersen
Dev 6: 432– 438, 1996. S, and Arner P. The effect of irbesartan on the development of diabetic
39. Hotta O, Furuta T, Chiba S, Tomioka S, and Taguma Y. Regression of nephropathy in patients with type 2 diabetes. N Engl J Med 345: 870 – 878,
IgA nephropathy: a repeat biopsy study. Am J Kidney Dis 39: 493–502, 2001.
2002. 61. Rocha R, Chander PN, Khanna K, Zuckerman A, and Stier CT.
40. Hruska KA, Guo G, Wozniak M, Martin D, Miller S, Liapis H, Mineralocorticoid blockade reduces vascular injury in stroke-prone hyper-
Loveday K, Klahr S, Sampath TK, and Morrissey J. Osteogenic tensive rats. Hypertension 31: 451– 458, 1998.
protein-1 prevents renal fibrogenesis associated with ureteral obstruction. 62. Rodriguez-Iturbe B, Varzir ND, Herrera-Acosta J, and Johnson RJ.
Am J Physiol Renal Physiol 279: F130 –F143, 2000. Oxidative stress, renal infiltration of immune cells, and salt-sensitive
41. Isaka Y, Brees D, Ikegaya K, Kaneda Y, Imai E, Noble NA, and hypertension: all for one and one for all. Am J Physiol Renal Physiol 286:
Border WA. Gene therapy by skeletal muscle expression of decorin F606 –F616, 2004.
prevents fibrotic disease in rat kidney. Nat Med 2: 418 – 423, 1996. 63. Shiozawa S. Participation of macrophage in glomerular sclerosis through
42. Johnson TS, El-Koraie AF, Skill NJ, Baddour NM, El Nahas AM, the expression and activation of matrix metalloproteinases. Pathol Int 50:
Njloma M, Adam AG, and Griffin M. Tissue transglutaminase and the 441– 457, 2000.
progression of human renal scarring. J Am Soc Nephrol 14: 2052–2062, 2003.
64. Shoji T, Nakanishi I, Suzuki A, Hayashi T, Togawa M, Okada N, Imai
43. Johnson TS, Skill NJ, El Nahas AM, Oldroyd SD, Thomas GL,
E, Hori M, and Tsubakihara Y. Early treatment with corticosteroids
Douthwaite JA, Haylor JL, and Griffin M. Transglutaminase transcrip-
ameliorates proteinuria, proliferative lesions, and mesangial phenotypic
tion and antigen translocation in experimental renal scarring. J Am Soc
modulation in adult diffuse proliferative IgA nephropathy. Am J Kidney
Nephrol 10: 2146 –2157, 1999.
44. Kaikita K, Fogo AB, Ma L, Schoenhard JA, Brown NJ, and Vaughan Dis 35: 194 –201, 2000.
DE. Plasminogen activator inhibitor-1 deficiency prevents hypertension 65. Skill NJ, Griffin M, El Nahas AM, Sanai T, Haylor JL, Fisher M,

Downloaded from ajprenal.physiology.org on August 4, 2010


and vascular fibrosis in response to long-term nitric oxide synthase Jamie MF, Mould NN, and Johnson TS. Increases in renal ⑀-(␥-
inhibition. Circulation 104: 839 – 844, 2001. glutamyl)-lysine crosslinks result from compartment-specific changes in
45. Kasuga H, Ito Y, Sakamoto S, Kawachi H, Shimizu F, Yuzawa Y, and tissue transglutaminase in early experimental diabetic nephropathy: patho-
Matsuo S. Effects of anti-TGF-␤ type II receptor antibody on experimen- logic implications. Lab Invest 81: 705–716, 2001.
tal glomerulonephritis. Kidney Int 60: 1745–1755, 2001. 66. Skill NJ, Johnson TS, Coutts IG, Saint RE, Fisher M, Huang L, El
46. Klahr S and Morrissey JJ. The role of vasoactive compounds, growth Nahas AM, Collighan RJ, and Griffin M. Inhibition of transglutaminase
factors and cytokines in the progression of renal disease. Kidney Int Suppl activity reduces extracellular matrix accumulation induced by high glu-
75: S7–S14, 2000. cose levels in proximal tubular epithelial cells. J Biol Chem 279: 47754 –
47. Koo JW, Kim Y, Rozen S, and Mauer M. Enalapril accelerates remod- 47762, 2004.
eling of the renal interstitium after release of unilateral ureteral obstruction 67. Tharaux PL, Chatziantoniou C, Casellas D, Fouassier L, Ardaillou R,
in rats. J Nephrol 16: 203–209, 2003. and Dussaule JC. Vascular endothelin-1 gene expression, synthesis and
48. Lan HY, Nikolic-Paterson DJ, Zarama M, Vannice JL, and Atkins effect on renal type I collagen synthesis and nephroangiosclerosis during
RC. Suppression of experimental crescentic glomerulonephritis by the nitric oxide synthase inhibition in rats. Circulation 99: 2185–2191, 1999.
interleukin-1 receptor antagonist. Kidney Int 43: 479 – 485, 1993. 68. Trachtman H, Weiser AC, Valderrama E, Morgado M, and Palmer
49. Lehoux S, Lemarie CA, Esposito B, Lijnen HR, and Tedgui A. LS. Prevention of renal fibrosis by spironolactone in mice with complete
Pressure-induced matrix metalloproteinase-9 contributes to early hyper- unilateral ureteral obstruction. J Urol 172: 1590 –1594, 2004.
tensive remodeling. Circulation 109: 1041–1047, 2004. 69. Van Vliet A, van Alderwegen I, Baelde H, de Heer E, and Bruijn JA.
50. Lelongt B, Bengatta S, Delauche M, Lund LR, Werb Z, and Ronco Fibronectin accumulation in glomerulosclerotic lesions: self-assembly
PM. Matrix metalloproteinase 9 protects mice from anti-glomerular base- sites and the heparin II binding domain. Kidney Int 61: 481– 489, 2002.
ment membrane nephritis through its fibrinolytic activity. J Exp Med 193: 70. Verhagen AM, Rabelink TJ, Braam B, Opgenorth TJ, Grone HJ,
793– 802, 2001. Koomans HA, and Joles JA. Endothelin A receptor blockade alleviates
51. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, hypertension and renal lesions associated with chronic nitric oxide syn-
Ritz E, Atkins RC, Rohde R, and Raz I, Collaborative Study Group. thase inhibition. J Am Soc Nephrol 9: 755–762, 1998.
Renoprotective effect of the angiotensin-receptor antagonist irbesartan in 71. Wang B, Hao J, Jones SC, Yee MS, Roth JC, and Dixon IM. Decreased
patients with nephropathy due to type 2 diabetes. N Engl J Med 345: Smad 7 expression contributes to cardiac fibrosis in the infarcted rat heart.
851– 860, 2001. Am J Physiol Heart Circ Physiol 282: H1685–H1696, 2002.
52. Liu Y. Hepatocyte growth factor in kidney fibrosis: therapeutic potential and 72. Weistuch JM and Dworkin LD. Does essential hypertension cause end-
mechanisms of action. Am J Physiol Renal Physiol 287: F7–F16, 2004. stage renal disease? Kidney Int 41: S33–S37, 1992.
53. Ma LJ, Nakamura S, Whitsitt JS, Marcantoni C, Davidson JM, and 73. Yang J, Dai C, and Liu Y. Hepatocyte growth factor suppresses renal
Fogo AB. Regression of sclerosis in aging by an angiotensin inhibition-
interstitial myofibroblast activation and intercepts Smad signal transduc-
induced decrease in PAI-1. Kidney Int 58: 2425–2436, 2000.
tion. Am J Pathol 163: 621– 632, 2003.
54. Maschio G, Alberti D, Janin G, Locatelli F, Mann JF, Motolese M,
74. Yang J and Liu Y. Delayed administration of hepatocyte growth factor
Ponticelli C, Ritz E, and Zucchelli P. Effect of the angiotensin-convert-
reduces renal fibrosis in obstructive nephropathy. Am J Physiol Renal
ing-enzyme inhibitor benazepril on the progression of chronic renal
insufficiency. The Angiotensin-Converting-Enzyme Inhibition in Progres- Physiol 284: F349 –F357, 2003.
sive Renal Insufficiency Study Group. N Engl J Med 334: 939 –945, 1996. 75. Yusuf S, Dagenais G, Pogue J, Bosch J, and Sleight P. Vitamin E
55. Morrissey J, Hruska K, Guo G, Wang S, Chen Q, and Klahr S. Bone supplementation and cardiovascular events in high-risk patients: the Heart
morphogenetic protein-7 improves renal fibrosis and accelerates the return Outcomes Prevention Evaluation Study Investigators. N Engl J Med 342:
of renal function. J Am Soc Nephrol 13, Suppl 1: S14 –S21, 2002. 154 –160, 2000.
56. Nishikawa K, Guo YJ, Miyasaka M, Tamatani T, Collins AB, Sy MS, 76. Zeisberg M, Bottiglio C, Kumar N, Maeshima Y, Strutz F, Muller GA,
McCluskey RT, and Andres G. Antibodies to intercellular adhesion mole- and Kalluri R. Bone morphogenic protein-7 inhibits progression of
cule 1/lymphocyte function-associated antigen 1 prevent crescent formation in chronic renal fibrosis associated with two genetic mouse models. Am J
rat autoimmune glomerulonephritis. J Exp Med 177: 667– 677, 1993. Physiol Renal Physiol 285: F1060 –F1067, 2003.
57. Obermuller N, Morente N, Kranzlin B, Gretz N, and Witzgall R. A 77. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz
possible role for metalloproteinases in renal cyst development. Am J F, and Kalluri R. BMP-7 counteracts TGF-␤1-induced epithelial-to-
Physiol Renal Physiol 280: F540 –F550, 2001. mesenchymal transition and reverses chronic renal injury. Nat Med 9:
58. Oda T, Jung YO, Kim HS, Cai X, Lopez-Guisa JM, Ikeda Y, and 964 –968, 2003.
Eddy AA. PAI-1 deficiency attenuates the fibrogenic response to ureteral 78. Ziswiler R, Daniel C, Franz E, and Marti HP. Renal matrix metallo-
obstruction. Kidney Int 60: 587–596, 2001. proteinase activity is unaffected by experimental ischemia-reperfusion
59. Ostendorf T, Kunter U, Grone HJ, Bahlmann F, Kawachi H, Shimizu injury and matrix metalloproteinase inhibition does not alter outcome of
F, Koch KM, Janjic N, and Floege J. Specific antagonism of PDGF renal function. Exp Nephrol 9: 118 –124, 2001.

AJP-Renal Physiol • VOL 289 • AUGUST 2005 • www.ajprenal.org

You might also like