ArticlePDF Available

Alport syndrome from bench to bedside: the potential of current treatment beyond RAAS blockade and the horizon of future therapies

Authors:

Abstract and Figures

The hereditary type IV collagen disease Alport syndrome (AS) always leads to end-stage renal failure. Yesterday, for the past 90 years, this course was described as 'inevitable'. Today, RAAS blockade has changed the 'inevitable' course to a treatable disease. Tomorrow, researchers hope to erase the 'always' from 'always leads to renal failure' in the textbooks. This review elucidates therapeutic targets that evolve from research: (i) kidney embryogenesis and pathogenesis; (ii) phenotype-genotype correlation and the role of collagen receptors and podocytes; (iii) the malfunctioning Alport-GBM; (iv) tubulointerstitial fibrosis; (v) the role of proteinuria in pathogenesis and prognosis; and (vi) secondary events such as infections, hyperparathyroidism and hypercholesterolaemia. Therefore, moderate lifestyle, therapy of bacterial infections, Paricalcitol in adult patients with hyperparathyroidism and HMG-CoA-reductase inhibitors in adult patients with dyslipoproteinemia might contribute to a slower progression of AS and less cardiovascular events. In the future, upcoming treatments including stem cells, chaperon therapy, collagen receptor blockade and anti-microRNA therapy will expand our perspective in protecting the kidneys of Alport patients from further damage. This perspective on current and future therapies is naturally limited by our personal focus in research, but aims to motivate young scientists and clinicians to find a multimodal cure for AS.
No caption available
… 
Content may be subject to copyright.
Nephrol Dial Transplant (2014) 29: iv124iv130
doi: 10.1093/ndt/gfu028
Full Review
Alport syndrome from bench to bedside: the potential of
current treatment beyond RAAS blockade and the
horizon of future therapies
Oliver Gross1,*, Laura Perin2,*and Constantinos Deltas3,*
1
Clinic of Nephrology and Rheumatology, University Medicine Goettingen, Goettingen, Germany,
2
Saban Research Institute, Childrens
Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA and
3
Molecular Medicine Research Center and Laboratory
of Molecular and Medical Genetics, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
Correspondence and offprint requests to: Oliver Gross; E-mail: gross.oliver@med.uni-goettingen.de
*
All authors contributed equally to this work.
ABSTRACT
The hereditary type IV collagen disease Alport syndrome (AS)
always leads to end-stage renal failure. Yesterday, for the past
90 years, this course was described as inevitable. Today,
RAAS blockade has changed the inevitablecourse to a treata-
ble disease. Tomorrow, researchers hope to erase the always
from always leads to renal failurein the textbooks. This
review elucidates therapeutic targets that evolve from research:
(i) kidney embryogenesis and pathogenesis; (ii) phenotype-
genotype correlation and the role of collagen receptors and
podocytes; (iii) the malfunctioning Alport-GBM; (iv) tubu-
lointerstitial brosis; (v) the role of proteinuria in pathogenesis
and prognosis; and (vi) secondary events such as infections,
hyperparathyroidism and hypercholesterolaemia. Therefore,
moderate lifestyle, therapy of bacterial infections, Paricalcitol
in adult patients with hyperparathyroidism and HMG-CoA-
reductase inhibitors in adult patients with dyslipoproteinemia
might contribute to a slower progression of AS and less cardio-
vascular events. In the future, upcoming treatments including
stem cells, chaperon therapy, collagen receptor blockade and
anti-microRNA therapy will expand our perspective in pro-
tecting the kidneys of Alport patients from further damage.
This perspective on current and future therapies is naturally
limited by our personal focus in research, but aims to motivate
young scientists and clinicians to nd a multimodal cure
for AS.
Keywords: Alport syndrome, chaperon therapy, discoidin
domain receptor 1, kidney brosis, microRNA-21
ESSENTIALS FOR FUTURE THERAPIES:
PATHOGENESIS OF ALPORT SYNDROME
AND KIDNEY EMBRYOGENESIS
Alport syndrome (AS) is a hereditary type IV collagen disease,
which always leads to progressive renal brosis and end-stage
renal failure [1]. Three different type IV collagen trimers are
deposited in basement membranes: α1/α1/α2, α3/α4/α5 and
α5/α5/α6 (IV) [2]. The mature glomerular basement mem-
brane (GBM) predominantly contains α3/α4/α5 type IV col-
lagen chains. Mutations in the type IV collagen genes
COL4A3/4/5, which encode the α3/α4/α5 chains, cause AS.
These mutations interfere with the correct assembly of the α3/
α4/α5 (IV) collagen network in the GBM and hinder the de-
velopmental switch from the embryonic α1/α1/α2(IV)
network to the mature α3/α4/α5 (IV) network, causing the
persistence of an immature GBM [3,4]. Consequently, a
thickening and splitting of the GBM in AS causes progressive
renal brosis leading to end-stage renal failure [5]. Maturation
of the GBM develops in the neonatal age; therefore, a child
with AS is not born with an abnormal GBM (but develops
AS during maturation of the GBM)leaving a therapeutic
© The Author 2014. Published by Oxford University Press
on behalf of ERA-EDTA. All rights reserved.
iv124
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
window for very early therapy. In fact, the defective Alport-
GBM can be restored in mice [6].
GENOTYPE-PHENOTYPE-CORRELATION
POINTS TO THE ACHILLESHEEL IN
ALPORT PATHOGENESIS: PODOCYTES AND
THEIR COLLAGEN RECEPTORS
The α3/α4/α5 type IV collagen chains are exclusively pro-
duced by podocytes [7]. The genotype-phenotype correlation
in AS [8] points to a role of the α3/α4/α5 (IV) producing and
sensing cells in AS pathogenesis: the podocytes sense their
GBM via collagen receptors such as α1β1 and α2β1 integrins
and discoidin domain receptor 1 (DDR1). All of these collagen
receptors have been shown to inuence Alport pathogenesis
[5,9,10], rendering them as possible therapeutic targets [11].
THEROLEOFASPONGYALPORT-GBM
FOR THERAPY
The GBM in most AS patients consists of α1/α2 (IV) chains
only, making this altered GBM more porous [12] (resulting in
acanthocyte formation) and more susceptible to endopro-
teolysis [3]. The GBM in AS patients is thought to be more
vulnerable by increased (or even normal) ltration pressure.
Therefore, thickening and splitting of the GBM in AS in part
is a stress response of the podocytes. Any medication reducing
the mechanical stress on the podocyte, such as RAAS blockade,
reduces the risk of GBM ruptures and focal segmental glomer-
ulosclerosis, which is a common light microscopical glomeru-
lar feature in AS.
THEROLEOFTUBULOINTERSTITIAL
FIBROSIS IN THE COURSE AND
PROGNOSIS OF AS
AS is a glomerular disease; however, tubulointerstitial brosis is
the key feature in progressive renal damage leading to end-stage
renal failure. For example, RAAS blockade is not able to hinder
thickening and splitting of the GBM in AS, but markedly delays
tubulointerstitial brosis [13]. The glomerular disease and the
podocyte stress response lead to distribution and secretion of
probrotic chemokines and cytokines in the primary urine that
are re-absorbed by the tubular cells. The re-absorbed probrotic
chemokines lead to tubular scar tissue formation that nally de-
molishes the kidney. Therefore, until now, the amount of tubu-
lointerstitial brosis is the most accurate histological prognostic
factor regarding the evaluation of kidney function.
THEROLEOFPROTEINURIAIN
PATHOGENESIS AND PROGNOSIS OF AS
Proteinuria reects ongoing glomerular inammatory damage
in all autoimmune diseases such as lupus nephritis and most
types of glomerulonephritis. Therefore, the amount of
proteinuria serves as an important prognostic factor in order
to evaluate response to therapy. In contrast, the amount of
proteinuria is a poor prognostic marker in AS, because it does
not automatically correlate with inammation and scar tissue
formation. For example, loss of 5 g protein per day (without
TGFβand CTGF) can have a better prognosis than only1g
per day with relatively high levels of probrotic chemokines.
We think that progression from haematuria to microalbumi-
nuria and from microalbuminuria to overt proteinuria rep-
resent very important steps in the course of Alport disease.
However, once the patient has reached the level of overt pro-
teinuria, increasing amounts of proteinuria do not necessarily
result in a worse prognosis or disease progression (unpub-
lished data from the European Alport registry [13]). In con-
trast, there is increasing evidence that the quality and quantity
of proinammatory and probrotic proteins in the urine de-
termines the course of AS [14].
THE UNDERESTIMATED COLLATERAL
DAMAGES: RECURRENT INFECTIONS,
HYPERPARATHYROIDISM,
CARDIOVASCULAR DISEASE AND
HYPERCHOLESTEROLAEMIA
In mice with AS, bacterial CpG-DNA accelerates glomerulo-
sclerosis by inducing a M1 proinammatory macrophage
phenotype and podocyte loss [15]. Vice versa, patients with
ongoing renal failure are immuno-compromised and are more
likely to get recurrent bacterial infections. Therefore, these
bacterial infections should be treated rigorously and good
dental health might contribute to a slower progression of AS.
Most patients with progressive renal failure develop secondary
hyperparathyroidism due to their impaired calcium-phosphate-
vitamin D balance. In mice with AS, the vitamin D receptor
activator Paricalcitol (but not Calcitriol) showed a synergistic
nephroprotective effect on top of early ACE inhibition [16].
Paricalcitol is board-approved for therapy of secondary hy-
perthyroidism. Therefore, in adult patients with AS and incipi-
ent hyperparathyroidism, Paricalcitol might be an additional
treatment option to delay renal failure.
Young patients with chronic renal disease have a 1000-fold
higher risk of cardiovascular effects compared with healthy sub-
jects. Proteinuria in a nephrotic range causes hypercholestero-
laemia. In mice with AS, the HMG-CoA-inhibitor cerivastatin
(statin) prolonged the lifespan until renal failure and delayed
uraemia [17]. These effects were associated with decreased
renal brosis and a reduction of inammatory cell inltration.
Statins are board-approved for therapy of dyslipoproteinemia.
Therefore, in adult patients with AS and incipient hypercholes-
terolaemia, statins might be an additional treatment option to
delay renal failure and prevent cardiovascular events.
Figure 1summarizes the key features of possible therapies
on top of RAAS blockade. All medications in Figure 1are
board-approved for other medical conditions, but all possible
therapies are off-label in AS and are very likely to stay off-label
in the future. Medications in Figure 1do not represent an
expert recommendation for therapy of AS, but summarizes
FULL REVIEW
Future therapy of Alport syndrome iv125
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
medical therapies currently used (or could be used) in adult
patients with AS.
FUTURE THERAPIES: COLLAGEN IV
PATHOLOGY AND IDEAS FOR NEW
THERAPEUTIC APPROACHES
Collagens are large triple-helical proteins participating in a
variety of processes in connective tissues, including tissue scaf-
folding, cell adhesion and tissue repair, among others. Struc-
turally, there are two major categories, the brillar and the
non-brillar collagens. Prime examples of the rst are the type
I, II, III and V collagens that exert a fundamental gluing role
in extracellular matrix, holding cells together; a prime example
of the latter is type IV collagen, the most abundant component
of all basement membranes (BM), with a crucial role in the
kidney glomerular ltration barrier [18]. A critical character-
istic relating to this review is the participation of brillar col-
lagens in higher order structures, through multiple nucleation
events, a process that in case of mutations is susceptible to
strong dominant negative effects. This is exemplied in dis-
eases such as osteogenesis imperfecta where heterozygous
mutations in type I collagen [(α1)
2
,α2)] interfere with triple-
helix formation, affecting 75% of molecules and delaying
protein secretion. These defective helices interfere next with
proper nucleation and bril formation in bones and other con-
nective tissues [19,20]. One can anticipate that the more de-
fective molecules are secreted, the worse the phenotype is
going to be. Actually, it was shown that the phenotype was
milder if a premature termination of translation prevented
chain association and triple-helix formation, which in turn
prevented secretion of defective molecules.
To the contrary, type IV collagen of BM participates in
network formation which is not the result of molecular nu-
cleation events. Certainly there are interactions with other BM
macromolecules such as laminin, nidogen and proteoglycans
but there is no extensive nucleation and therefore less drastic
dominant-negative effects are expected. For some type IV col-
lagen mutations we hypothesize that should it be possible to
have more molecules secreted in the extracellular matrix and
participate in the meshwork, the phenotype might be milder
compared with no secretion at all. About 50% of AS patients
inherit missense or small in-frame mutations in the X-linked
COL4A5 gene, where mature protomers are either secreted
normally or abnormally, based on immunostaining. Absent or
weak staining is due to poor secretion, most probably because
quality control in the ER recognizes and degrades the mis-
folded chains [21]. This is accomplished through the unfolded
protein response (UPR) pathway which is activated by the ER
stress. UPR activation aims at restoring homeostasis by pro-
moting proper protein folding using chaperones. When this
fails for various reasons, the unfolded or misfolded molecules
are removed by degradation. Prolonged ER stress may lead to
protein translation pause or even to podocyte apoptosis or
death through other mechanisms and foot processes efface-
ment. Mechanisms aimed at enhancing the intracellular cha-
perone machinery or externally administered small chemicals
that mimic chaperones could promote triple-helix formation
and consequently allow hypomorphic mutants to exert their
function, even though not perfectly, once found in the BM.
This has been shown in numerous other occasions, such as
cystic brosis and nephrogenic diabetes insipidus [22,23].
A most recent example relating to BM pathology was
the transgenic expression of the mutant rat C321R-LAMB2
gene in Lamb2
-/
mice, a model that recapitulates Pierson
FIGURE 1: Medical therapies currently used (or could be used) in adult patients with AS and risk factors that might negatively contribute to
progression of disease. Medications do not necessarily represent an expert recommendation for therapy of AS.
FULL REVIEW
iv126 O. Gross et al.
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
syndrome. This arrangement attenuated the severe proteinur-
ia, while it caused ER stress, noting that mutant protein was
better than no protein. Also, the same authors showed that the
use of chemical chaperone taurodeoxycholic acid in cells facili-
tated protein folding and trafcking and greatly increased
secretion of the mutant LAMB2 [24]. Previously, Ohashi et al.
had shown that mutant podocin due to NPHS2 mutation p.
R138Q could not reach its destination in plasma membrane of
cultured cells due to retention in the ER. Incubation with
chemical chaperones caused folding of the mutant podocin
and redistribution to the plasma membrane [25]. With regard
to collagens, Murray et al. showed recently that treatment with
the chemical chaperone 4-phenyl butyric acid reduced intra-
cellular accumulation of mutant collagen IV in cultured
patient primary cells. This ameliorated the cellular phenotype
of a COL4A2 mutation that caused haemorrhagic stroke [26].
It is tempting to hypothesize that once found in its correct des-
tination the mutant protein will function properly or near
properly, depending on the exact nature of the mutation. Hy-
pomorphic and milder mutations that interfere with molecule
folding may be selected against during a very strict quality
control early on in the ER; however, it is reasonable to hypoth-
esize that once folded with the accessory contribution of exter-
nal chaperones, these molecules may reach their destination
and function sub-optimally but adequately and prevent severe
disease progression.
Recently we created a new AS knockin mouse model, carry-
ing missense mutation COL4A3-G1332E that demonstrated
UPR activation in glomeruli. Hopefully, this will serve as a tool
for testing a variety of available chemical chaperones and
other alternative therapeutic approaches for alleviating or
halting disease progression [27].
FUTURE THERAPIES: STEM CELL-BASED
THERAPIES FOR AS
Stem cells and renal progenitors might offer a possible novel
treatment of AS. Being able to deliver to the affected glomeruli
a cell that could potentially become a mature podocyte, and
produce new functional GBM, could be considered the Holy
Grailin the treatment of AS. Even if different groups [2830]
have claimed podocyte differentiation from stem cells in
Alport mice, these publications still need validation and do
not convincingly demonstrate that podocyte differentiation and
consequent restoration of the GBM really occurs. In fact, the
concept that direct differentiation of stem cells into organ-
specic mature cell types occurs and can rescue the progression
of disease has almost been abandoned; stem cell integration
and differentiation is a very rare event that cannot sustain the
complete regeneration of kidney [31] or other organs.
In particular, our group has demonstrated [32] that differ-
entiation of injected stem cells (amniotic uid stem cells) into
podocytes does not occur in vivo in an animal model of AS,
despite signicant protection of glomerular structure and
function.The main mechanism of action of stem cells appears
to be via paracrine activity ( possibly affecting the TGFβaxis)
leading to attenuation of brosis and chronic inammation
[3234], in addition to stimulating ingress of healingtype II
macrophages (M2) [34]. In addition, stem cells seem to induce
blockade of the angiotensin II pathway that prevents further
damage to the glomeruli and favours preservation of podocyte
number. Thus, based on studies published so far, cell-based
therapies have the potential to slow but not prevent renal injury
in AS. Despite these encouraging results and increases in the
lifespan of treated mice, the restoration of a functional GBM is
still the elusive but requisite goal to actually cure AS [6].
In continuing efforts to nd a cell source that produces the
proper GBM, we and others have been working to obtain new
podocytesincluding in the form of nephron progenitors that
can be induced to become podocytes. Several studies con-
ducted on the differentiation of embryonic stem cells (ESC)
and adult stem cells into intermediate mesoderm or cap me-
senchyme (embryological tissues from which the kidney
originates) have demonstrated the possibility of inducing com-
mitted cell differentiation in renal lineages [6,3538]. Other
groups [39] have demonstrated the possibility of expanding
nephron progenitors using induced pluripotent stem cell (IPS)
technology, while Song et al. [40] have a newly developed IPS
line of cells that resemblein vivo podocytes. However, gener-
ation of differentiated renal structures from IPS or ESC has
very low efciency and is probably insufcient for strategies to
directly translate these cells into potential therapeutic agents
[41]. We have recently reported the isolation of a subpopu-
lation within the human amniotic uid that possess character-
istics of nephron progenitors and that can be differentiated
into podocytes expressing the collagen IV trimer [42].
With the challenges of brosis, chronic inammation and
podocytes incapable of laying down the proper type IV col-
lagen network, a cell-based approach would ideally be able to
act simultaneously on all these aspects in order to offer a
robust treatment for AS. In this regard, different cell-based
strategies might be combined together. Stem cells, being more
immune-privileged than adult renal cells, can be systemically
infused and act in reducing brosis and inammation, while
direct replenishment of new non-defective podocytes derived
from nephron progenitors might be the solution to replace the
failing membrane (Figure 2). Nevertheless, one of the biggest
challenges will be getting the cells across the GBM. An impor-
tant feature of the novel podocyte progenitor cells mentioned
above is their potential use in the development of small mol-
ecule-based therapies [6,43] aimed at salvaging disturbed type
IV collagen production. Thus, although we are still far from
being able to harness novel cell sources like stem or progenitor
cells to support new therapeutic approaches, several promising
avenues seem possible.
FUTURE THERAPIES: COLLAGEN IV
RECEPTOR BLOCKADE IN AS
In general, the GBM structure is maintained by an equilibrium
of synthesis and degradation. In Alport pathogenesis, in-
creased synthesis of defective α3/4/5 type IV collagen, imma-
ture α1/α1/α2 type IV collagen and other basement
membrane components in the GBM results in excessive
FULL REVIEW
Future therapy of Alport syndrome iv127
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
accumulation of matrix proteins [5]. Ablating the function of
collagen receptors such as Discoidin domain receptor 1
(DDR1), integrin α1β1orα2β1 might hinder or slow down
podocytesmatrix accumulation in AS [9,10]. For example,
loss of DDR1 reduced proinammatory, probrotic cells via
signalling of TGFβ, CTGF, NFκB and IL-6 and decreased
deposition of extracellular matrix. Immunogold staining and
in situ hybridization identied podocytes as key players in
DDR1-mediated brosis and inammation [10].
This supports our hypothesis that podocytematrix inter-
action via collagen receptors plays an important part in pro-
gression of brosis. We postulate that specic receptor
blockers such as DDR1 blockers might become a new thera-
peutic option in patients with AS in the near future.
FUTURE THERAPIES: ANTI-MICRORNA
THERAPY IN AS
MicroRNA-21 (miR-21) has been shown to play a distinct role
in the progression of kidney scarring in different animal
models and humans [44]. Mice treated with anti-miR-21 oli-
gonucleotides suffered far less interstitial brosis in response
to kidney injury. Analysis of gene expression proles identied
groups of genes involved in metabolic pathways, including the
lipid metabolism pathway regulated by peroxisome prolifera-
tor-activated receptor-α(Pparα), a direct miR-21 target [44].
Further, miR-21 Sponge inhibited TGFβ-stimulated phos-
phorylation of Akt kinase, resulting in attenuation of phos-
phorylation of different Akt substrates that regulate mesangial
cell hypertrophy [45]. Additionally, inhibition of miR-21
reduced TGFβ-stimulated bronectin and collagen expression
[45]. TGFβis a very well-known key player in Alport patho-
genesis and progressive kidney brosis in AS [13].
These studies demonstrated that miR-21 contributes to -
brogenesis and is a promising candidate target for antibrotic
therapy in AS. We postulate that anti-miR-21 compounds
might become a new therapeutic option in patients with AS in
the near future.
Additional potential future targets of nephroprotective
therapy in AS such as the complement system, chemokine re-
ceptor blockade, TNFα-blockade and inhibition of matrix-
metallo-proteinases are summarized in a recent review [11].
PERSPECTIVE CURRENT AND FUTURE
THERAPY
Yesterday, in the textbooks the course of AS was described as
inevitablefor the past 90 years after the rst description by
Alport [46].
Today, RAAS blockade has changed the inevitablecourse
of AS to a treatable disease [47,48] and led to treatment rec-
ommendations [49,50]. Our review summarized additional
therapies that are currently used in humans and that might
inuence the course of AS (Figure 1). There is preliminary
scientic evidence for effectiveness in animal models.
However, the long-term effects of these therapies still need to
be evaluated in humans with AS. These therapies might well
be additive to RAAS blockade and might further delay renal
failure by years. Some medications are already board-approved
for other indications; therefore, international Alport registries
hopefully will generate the evidence for patients with AS.
Tomorrow, our review draws a bright horizon of several
promising new therapies, all with different targets additive to
existing therapies. This revives hope that AS has not only
become a treatable disease, but end-stage renal failure can be
prevented in most patients by multimodal therapy.
FIGURE 2: Schematic representation of possible mechanisms of injury repair in AS by stem cells and progenitors.
FULL REVIEW
iv128 O. Gross et al.
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
ACKNOWLEDGEMENTS
This article summarizes the presentations on future therapies
of Alport syndrome held at the 2014 International workshop
on Alport Syndrome, 35 January 2014, Said Business School,
Oxford, UK. The research activities of CD are supported by a
grant co-funded by the European Regional Development Fund
and the Republic of Cyprus through the Research Promotion
Foundation (Strategic Infrastructure Project NEW INFRA-
STRUCTURE/STRATEGIC/ 0308/24). L.P. was supported by
a grant of the Alport Syndrome Foundation and the Baxter
foundation. The presented research of O.G. is supported by
the Association pour l`Information et la Recherche sur les ma-
ladies rénale Génétiques (AIRG) France, the KfH-Foundation
Preventive Medicine, AbbVie GmbH & Co. KG Germany, the
German Kidney Foundation, the Deutsche Forschungsge-
meinschaft GR 1852/41 and 42, the European Renal Associ-
ation ERA/EDTA, the Kidney Foundation of Canada and the
Alport Syndrome Foundation.
CONFLICT OF INTEREST STATEMENT
None declared.
REFERENCES
1. Hudson BG, Tryggvason K, Sundaramoorthy M et al. Alports syndrome,
Goodpastures syndrome, and type IV collagen. N Engl J Med 2003; 348:
25432556
2. Boutaud A, Borza DB, Bondar O et al. Type IV collagen of the glomerular
basement membrane. Evidence that the chain specicity of network as-
sembly is encoded by the noncollagenous NC1 domains. J Biol Chem
2000; 275: 3071630724
3. Kalluri R, Shield CF, Todd P et al. Isoform switching of type IV collagen is
developmentally arrested in X-linked Alport syndrome leading to in-
creased susceptibility of renal basement membranes to endoproteolysis.
J Clin Invest 1997; 99: 24702478
4. Abrahamson DR, Prettyman AC, Robert B et al. Laminin-1 reexpression
in Alport mouse glomerular basement membranes. Kidney Int 2003; 63:
826834
5. Kruegel J, Rubel D, Gross O. Alport syndromeinsights from basic and
clinical research. Nat Rev Nephrol 2013; 9: 170178
6. Lin X, Suh JH, Go G et al. Feasibility of repairing glomerular basement
membrane defects in Alport syndrome. J Am Soc Nephrol 2013; Novem-
ber 21. [Epub ahead of print]
7. Abrahamson DR, Hudson BG, Stroganova L et al. Cellular origins of type
IV collagen networks in developing glomeruli. J Am Soc Nephrol 2009;
20: 14711479
8. Gross O, Netzer KO, Lambrecht R et al. Meta-analysis of genotype-
phenotype correlation in X-linked Alport syndrome: impact on clinical
counselling. Nephrol Dial Transplant 2002; 17: 12181227
9. Cosgrove D, Rodgers K, Meehan D et al. Integrin alpha1beta1 and trans-
forming growth factor-beta1 play distinct roles in Alport glomerular
pathogenesis and serve as dual targets for metabolic therapy. Am J Pathol
2000; 157: 16491659
10. Gross O, Girgert R, Beirowski B et al. Loss of collagen-receptor DDR1
delays renal brosis in hereditary type IV collagen disease. Matrix Biol
2010; 29: 346356
11. Noone D, Licht C. An update on the pathomechanisms and future thera-
pies of Alport syndrome. Pediatr Nephrol 2013; 28: 10251036
12. Ryu M, Migliorini A, Miosge N et al. Plasma leakage through glomerular
basement membrane ruptures triggers the proliferation of parietal epi-
thelial cells and crescent formation in non-inammatory glomerular
injury. J Pathol 2012; May 3. doi:10.1002/path.4046. [Epub ahead of print]
13. Gross O, Beirowski B, Koepke ML et al. Preemptive ramipril therapy
delays renal failure and reduces renal brosis in COL4A3-knockout mice
with Alport syndrome. Kidney Int 2003; 63: 438446
14. Pohl M, Danz K, Gross O et al. Diagnosis of Alport syndromesearch for
proteomic biomarkers in body uids. Pediatr Nephrol 2013; 28:
21172123
15. Ryu M, Kulkarni OP, Radomska E et al. Bacterial CpG-DNA accelerates
Alport glomerulosclerosis by inducing an M1 macrophage phenotype and
tumor necrosis factor-α-mediated podocyte loss. Kidney Int 2011; 79:
189198
16. Rubel D, Stock J, Ciner A et al. Antibrotic, nephroprotective effects of
Paricalcitol versus Calcitriol on top of ACE-inhibitor therapy in the
COL4A3 knockout mouse model for progressive renal brosis. Nephrol
Dial Transplant 2014; 29: 10121019
17. Koepke ML, Weber M, Schulze-Lohoff E et al. Nephroprotective effect of
the HMG-CoA-reductase inhibitor cerivastatin in a mouse model of pro-
gressive renal brosis in Alport syndrome. Nephrol Dial Transplant 2007;
22: 10621069
18. Kadler KE, Baldock C, Bella J et al. Collagens at a glance. J Cell Sci 2007;
120(Pt 12): 19551958
19. Constantinou CD, Nielsen KB, Prockop DJ. A lethal variant of osteo-
genesis imperfecta has a single base mutation that substitutes cys-
teine for glycine 904 of the alpha 1(I) chain of type I procollagen. The
asymptomatic mother has an unidentied mutation producing an
overmodied and unstable type I procollagen. J Clin Invest 1989; 83:
574584
20. Vogel BE, Doelz R, Kadler KE et al. A substitution of cysteine for glycine
748 of the alpha 1 chain produces a kink at this site in the procollagen
I molecule and an altered N-proteinase cleavage site over 225 nm away.
J Biol Chem 1988; 263: 1924919255
21. Hashimura Y, Nozu K, Kaito H et al. Milder clinical aspects of X-linked
Alport syndrome in men positive for the collagen IV alpha5 chain. Kidney
Int 2013; December 4. doi:10.1038/ki.2013.479. [Eepub ahead of print]
22. Powell K, Zeitlin PL. Therapeutic approaches to repair defects in
deltaF508 CFTR folding and cellular targeting. Adv Drug Deliv Rev 2002;
54: 13951408
23. Morello JP, Salahpour A, Laperriere A et al. Pharmacological chaperones
rescue cell-surface expression and function of misfolded V2 vasopressin
receptor mutants. J Clin Invest 2000; 105: 887895
24. Chen YM, Zhou Y, Go G et al. Laminin beta2 gene missense mutation
produces endoplasmic reticulum stress in podocytes. J Am Soc Nephrol
2013; 24: 12231233
25. Ohashi T, Uchida K, Uchida S et al. Intracellular mislocalization of
mutant podocin and correction by chemical chaperones. Histochem Cell
Biol 2003; 119: 257264
26. Murray LS, Lu Y, Taggart A et al. Chemical chaperone treatment reduces
intracellular accumulation of mutant collagen IV and ameliorates the cel-
lular phenotype of a COL4A2 mutation that causes haemorrhagic stroke.
Hum Mol Genet 2014; 23: 283292
27. Pieri M, Stefanou C, Zaravinos A et al. Evidence for activation of the un-
folded protein response in collagen IV nephropathies. J Am Soc Nephrol
2014; 25: 260275
28. Sugimoto H, Mundel TM, Sund M et al. Bone-marrow-derived stem cells
repair basement membrane collagen defects and reverse genetic kidney
disease. Proc Natl Acad Sci USA 2006; 103: 73217326
29. LeBleu V, Sugimoto H, Mundel TM et al. Stem cell therapies benet
Alport syndrome. J Am Soc Nephrol 2009; 20: 23592370
30. Prodromidi EI, Poulsom R, Jeffery R et al. Bone marrow-derived cells con-
tribute to podocyte regeneration and amelioration of renal disease in a
mouse model of Alport syndrome. Stem Cells 2006; 24: 24482455
31. Perin L, Da Sacco S, De Filippo RE. Regenerative medicine of the kidney.
Adv Drug Deliv Rev 2011; 63: 379387
32. Sedrakyan S, Da Sacco S, Milanesi A et al. Injection of amniotic uid stem
cells delays progression of renal brosis. J Am Soc Nephrol 2012; 23:
661673
FULL REVIEW
Future therapy of Alport syndrome iv129
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
33. Wise AF, Ricardo SD. Mesenchymal stem cells in kidney inammation
and repair. Nephrology (Carlton) 2012; 17: 110
34. Williams TM, Little MH, Ricardo SD. Macrophages in renal development,
injury, and repair. Semin Nephrol 2010; 30: 255267
35. Kobayashi T, Tanaka H, Kuwana H et al. Wnt4-transformed mouse em-
bryonic stem cells differentiate into renal tubular cells. Biochem Biophys
Res Commun 2005; 336: 585595
36. Kramer J, Steinhoff J, Klinger M et al. Cells differentiated from mouse em-
bryonic stem cells via embryoid bodies express renal marker molecules.
Differentiation 2006; 74: 91104
37. Bruce SJ, Rea RW, Steptoe AL et al.In vitro differentiation of murine em-
bryonic stem cells toward a renal lineage. Differentiation 2007; 75:
337349
38. Hendry CE, Vanslambrouck JM, Ineson J et al. Direct transcriptional re-
programming of adult cells to embryonic nephron progenitors. J Am Soc
Nephrol 2013; 24: 14241434
39. Mae S, Shono A, Shiota F et al. Monitoring and robust induction of ne-
phrogenic intermediate mesoderm from human pluripotent stem cells.
Nat Commun 2013; 4: 1367
40. Song B, Smink AM, Jones CV et al. The directed differentiation of human
iPS cells into kidney podocytes. PLoS One 2012; 7: e46453
41. Takasato M, Maier B, Little MH. Recreating kidney progenitors from plur-
ipotent cells. Pediatr Nephrol 2013; September 13. [Epub ahead of print]
42. Da Sacco S, Lemley KV, Sedrakyan S et al. A novel source of cultured po-
docytes. PLoS One 2013; 8: e81812
43. Drozdova T, Papillon J, Cybulsky AV. Nephrin missense mutations: induc-
tion of endoplasmic reticulum stress and cell surface rescue by reduction
in chaperone interactions. Physiol Rep 2013; 1: e00086
44. Chau BN, Xin C, Hartner J et al. MicroRNA-21 promotes brosis of the
kidney by silencing metabolic pathways. Sci Transl Med 2012; 4: 121ra18
45. Dey N, Ghosh-Choudhury N, Kasinath BS et al. TGFβ-stimulated micro-
RNA-21 utilizes PTEN to orchestrate AKT/mTORC1 signaling for mesan-
gial cell hypertrophy and matrix expansion. PLoS One 2012; 7: e42316
46. Alport AC. Hereditary familial congenital haemorrhagic nephritis. Br
Med J 1927; 1: 504506
47. Gross O, Licht C, Anders HJ et al. Early angiotensin converting enzyme
inhibition in Alport syndrome delays renal failure and improves life
expectancy. Kidney Int 2012; 81: 494501
48. Temme J, Kramer A, Jager KJ et al. Outcomes of male patients with
Alport syndrome on renal replacement therapy. Clin J Am Soc Nephrol
2012; 7: 19691976
49. Kashtan CE, Ding J, Gregory M et al. Clinical practice recommendations
for the treatment of Alport syndrome: a statement of the Alport Syndrome
Research Collaborative. Pediatr Nephrol 2013; 28: 511
50. Savige J, Gregory M, Gross O et al. Expert guidelines for the management
of Alport syndrome and thin basement membrane nephropathy. J Am Soc
Nephrol 2013; 24: 364375
Received for publication: 26.12.2013; Accepted in revised form: 17.1.2014
FULL REVIEW
iv130 O. Gross et al.
at Niedersaechsische Staats- u. Universitaetsbibliothek Goettingen on September 1, 2014http://ndt.oxfordjournals.org/Downloaded from
... AS recently has become a treatable disease, as renal failure can be delayed by years and life expectancy improved by angiotensinconverting enzyme inhibitors (ACEi) [10][11][12]. However, new therapies beyond RAAS blockade are strongly needed to further delay renal fibrosis in AS [13]. ...
... Therefore, the albumin-binding of anti-miR-21 compounds might be an advantage in chronic kidney diseases. Regardless of the ultimate mechanism of uptake, the tubule epithelia have the highest levels of uptake and are in the greatest need of protection from damage in AS [2,13]. Interestingly, our RNA-seq data showed a superior capability of anti-miR-21 at the doses tested during the course of these studies (when compared to ACEi) to normalize profibrotic and inflammatory pathways towards the wildtype level. ...
Article
Full-text available
Col4a3−/− Alport mice serve as an animal model for renal fibrosis. MicroRNA-21 (miR-21) expression has been shown to be increased in the kidneys of Alport syndrome patients. Here, we investigated the nephroprotective effects of Lademirsen anti-miR-21 therapy. We used a fast-progressing Col4a3−/− mouse model with a 129/SvJ background and an intermediate-progressing F1 hybrid mouse model with a mixed genetic background, with angiotensin-converting enzyme inhibitor (ACEi) monotherapy in combination with anti-miR-21 therapy. In the fast-progressing model, the anti miR-21 and ACEi therapies showed an additive effect in the reduction in fibrosis, the decline of proteinuria, the preservation of kidney function and increased survival. In the intermediate-progressing F1 model, the anti-miR-21 and ACEi therapies individually improved kidney pathology. Both also improved kidney function and survival; however, the combination showed a significant additive effect, particularly for survival. RNA sequencing (RNA-seq) gene expression profiling revealed that the anti-miR-21 and ACEi therapies modulate several common pathways. However, anti-miR-21 was particularly effective at normalizing the expression profiles of the genes involved in renal tubulointerstitial injury pathways. In conclusion, significant additive effects were detected for the combination of anti-miR-21 and ACEi therapies on kidney function, pathology and survival in Alport mouse models, as well as a strong differential effect of anti-miR-21 on the renal expression of fibrotic factors. These results support the addition of anti-miR-21 to the current standard of care (ACEi) in ongoing clinical trials in patients with Alport syndrome.
... [15]. Считается, что блокада РААС способна снизить высокое внутриклубочковое артериальное давление и, как следствие, риск разрыва базальной мембраны и прогрессирования связанных с этим протеинурии и гломерулярного склероза [31]. Кроме того, блокируя действие альдостерона, можно воспрепятствовать увеличению продукции фибронектина и трансформирующего фактора роста бета, ассоциированных с развитием гломерулосклероза и тубулоинтерстициального фиброза [32]. ...
Article
Background . Alport syndrome is a systemic, hereditary, progressive disease characterized by ultrastructural changes in the glomerular basement membrane caused by pathogenic variants of type IV collagen genes. The use of angiotensin-converting enzyme inhibitors (ACEI) for nephroprotection is effective at the microhematuria and/or albuminuria stage. Treatment tactics in case of nephrotic syndrome development in such patients remains the subject of discussion. Clinical case description . The patient was diagnosed with proteinuria at the neonatal period and hematuria at the age of one month. The hereditary nephritis was diagnosed at the age of 6 years; the ACEI was administered, however, the proteinuria continued to increase. The diagnosis was confirmed at the age of 8.5 years via the puncture nephrobiopsy: collagenopathy, type IV, focal segmental glomerular sclerosis. Moreover, chronic bilateral sensorineural hearing loss and bilateral myopic astigmatism were diagnosed. Ciclosporin A (125 mg/day) was additionally prescribed. The increase in the cystatin C, urea, uric acid, cholesterol levels in blood was mentioned after 14 months of treatment. These parameters decreased after reducing cyclosporine A dose to 100 mg/day, however, proteinuria has increased. Angiotensin II receptor blocker (candesartan 8 mg/day) was prescribed to enhance nephroprotective therapy at the age of 10 years 2 months. Another increase of the immunodepressant dose was performed at the age of 11, it led to decrease in the estimated glomerular filtration rate and increase of creatinine, cystatin C, urea, cholesterol, uric acid, and potassium levels in the blood. These changes were considered as cyclosporine-dependent. The dose of cyclosporine A was reduced to 125 mg/day, and to 100 mg/day from the age of 14. There was no progression of chronic kidney disease at the follow-up at the age of 15.5 years. Conclusion . Nephroprotective treatment of a child with Alport syndrome initiated after the development of nephrotic syndrome did not stop the chronic kidney disease progression. Whereas relatively high doses of ciclosporin A have reduced proteinuria but led to nephrotoxicity and cyclosporin dependence.
... The necessity for identifying patients at high risk of fast kidney function decline remains unfulfilled [100]. Patients with Alport syndrome have been found to have tubulointerstitial fibrosis, a sign of renal disease development [101,102]. ...
Article
Background: The epidermal growth factor (EGF) is a globular protein that is generated in the kidney, especially in the loop of Henle and the distal convoluted tubule. While EGF is nonexistent or hardly detectable in plasma, it is present in normal people's urine. Until now, risk stratification and chronic kidney disease (CKD) diagnosis have relied on estimated glomerular filtration rate (eGFR) and urine albumin/creatinine ratio (uACR), both of which reflect glomerular function or impairment. Tubular dysfunction, on the other hand, may also be associated with renal failure. Summary: Because decreased urine EGF (uEGF) indicates tubular atrophy and interstitial fibrosis, this biomarker, together with eGFR and uACR, may be employed in the general population for risk assessment and diagnosis of CKD. uEGF levels have been shown to correlate with intrarenal EGF mRNA expression and have been found to decrease in a variety of glomerular and non-glomerular kidney disorders. Key message: uEGF, uEGF/creatinine, or uEGF/monocyte chemotactic peptide-1 are possible "new generation" biomarkers linked to a variety of kidney diseases that deserve further investigation as a single biomarker or as part of a multi-biomarker panel.
... In detail, Paricalcitol and HMG-CoA-reductase-inhibitors (Statins) both showed pleiotropic nephroprotective effects in COL4A3 −/− mice as animal models for AS [42,43]. Both compounds are part of the treatment recommendations, if indicated because of secondary hyperparathyroidism (Paricalcitol) or elevated cholesterol levels (Statin) [44,45]. Green circles indicate compounds, which positively influence both, the chronic kidney disease and the high cardiovascular risk in AS. ...
Article
Full-text available
In 1927 Arthur Cecil Alport, a South African physician, described a British family with an inherited form of kidney disease that affected males more severely than females and was sometimes associated with hearing loss. In 1961, the eponymous name Alport syndrome was adopted. In the late twentieth century three genes responsible for the disease were discovered: COL4A3, COL4A4, and COL4A5 encoding for the α3, α4, α5 polypeptide chains of type IV collagen, respectively. These chains assemble to form heterotrimers of type IV collagen in the glomerular basement membrane. Scientists, clinicians, patient representatives and their families, and pharma companies attended the 2019 International Workshop on Alport Syndrome, held in Siena, Italy, from October 22 to 26, and the 2021 online Workshop from November 30 to December 4. The main topics included: disease re-naming, acknowledging the need to identify an appropriate term able to reflect considerable clinical variability; a strategy for increasing the molecular diagnostic rate; genotype-phenotype correlation from monogenic to digenic forms; new therapeutics and new therapeutic approaches; and gene therapy using gene editing. The exceptional collaborative climate that was established in the magical medieval setting of Siena continued in the online workshop of 2021. Conditions were established for collaborations between leading experts in the sector, including patients and drug companies, with the aim of identifying a cure for Alport syndrome.
... Based on the pathophysiology of AS, any medication that can reduce the intraglomerular blood pressure, such as Renin-Angiotensin-Aldosterone System (RAAS) blockers, is considered to be able to prevent the mechanical stress on the podocyte and the risk of GBM ruptures that could potentially lead to the development of proteinuria and glomerular sclerosis [5]. RAAS blockers include Angiotensin-Converting Enzyme inhibitors (ACEi), Angiotensin receptor blockers [6], and Aldosterone receptor antagonists such as Spironolactone (SP). ...
Article
Full-text available
Background: The goal of the treatment of Alport syndrome (AS) is to delay the progression of kidney damage. The current standard of care is the use of Renin Angiotensin Aldosterone System (RAAS) blockers: angiotensin-converting enzyme inhibition (ACEi), angiotensin receptor blockade, and, recently, spironolactone (SP). Aim of the study: the purpose of this retrospective study is to evaluate the efficacy (reduction of proteinuria and changes of glomerular function) and safety of a sequential introduction of RAAS blockers up to a triple RAAS blockade in pediatric proteinuric patients with AS. Methods: in this retrospective study (1995 to 2019), we evaluated proteinuria values in AS patients, during the 12 months following the beginning of a new RAAS blocker, up to a triple blockade. ACEi was always the first line of treatment; then ARB and SP were sequentially added if uPCR increased by 50% from the basal level in 2 consecutive samples during a 3-months observation period, or when uPCR ratio was >2 mg/mg. Results: 26 patients (mean age at treatment onset was 10.55 ± 5.02 years) were enrolled. All patients were on ACEi, 14/26 were started on a second drug (6/14 ARB, 8/14 SP) after a mean time of 2.2 ± 1.7 years, 7/26 were on triple RAAS blockade after a further period of 5.5 ± 2.3 years from the introduction of a second drug. Repeated Measure Anova analysis of log-transformed data shows that the reduction of uPCR values after Time 0 from the introduction of the first, second and third drug is highly significant in all three cases (p values = 0.0016, 0.003, and 0.014, respectively). No significant changes in eGFR were recorded in any group, apart from a 15-year-old boy with X-linked AS, who developed kidney failure. One patient developed mild hyperkaliemia, and one gynecomastia and symptomatic hypotension. No life-threatening events were recorded. Conclusions: double and triple RAAS blockade is an effective and safe strategy to reduce proteinuria in children with AS. Nevertheless, we suggest monitoring eGFR and Kaliemia during follow-up.
Article
BACKGROUND. Alport syndrome is a rare hereditary kidney disease that causes progressive renal failure. There are significant differences in the progression of the disease between patients with Alport syndrome. Identifying patients with a high risk of rapid progression in order to optimally balance benefits and risks for prescribing therapy has become particularly important at this time. In this study, we wanted to assess whether the factors of proteolysis in blood and urine are associated with the nature of the course and to assess their prognostic value for children with Alport syndrome. THE AIM: To determine the level in blood serum and urinary excretion of MMP-2, MMP-3 and MMP-9 and their inhibitors TIMP-1 and 2, PAI-I, to show the relationship of their changes with the character of the course of Alport syndrome in children as an additional criterion for progression. PATIENTS AND METHODS. The study included 32 children with Alport syndrome. The level of MMP-2, MMP-3 and MMP-9 and their inhibitors TIMP-1 and 2, PAI-I, in blood serum and urine was determined by ELISA. A decrease in eGFR of ≥ 30 % at 2 years from baseline was chosen to indicate a progressive course of Alport syndrome. RESULTS. 28.1 % of children with Alport syndrome had a progressive course of the dis ease, 71.9 % had a slowly progressive course. The frequency of a decrease in MMP-9 and an increase in TIMP-1 both in blood (88.9 versus 43.5 % and 77.8 versus 21.7 %; p = 0.044 and 0.006, respectively) and in urine (100 versus 47, 8 % and 88.9 versus 30.4 %; 0.012 and 0.005, respectively) were statistically significantly more often detected in children with Alport syndrome with a progressive course of the disease than in a slowly progressive course. CONCLUSION. Type 9 matrix metalloproteinase and type 1 tissue matrix metalloproteinase inhibitor can be considered as risk factors for the progression of Alport syndrome in children.
Article
Full-text available
Alport syndrome (AS) is the most frequent monogenic inherited glomerulopathy and is also genetically and clinically heterogeneous. It is caused by semi-dominant pathogenic variants in the X-linked COL4A5 (NM_000495.5) gene or recessive variants in the COL4A3/COL4A4 (NM_000091.4/NM_000092.4) genes. The disease manifests in early childhood with persistent microhematuria and can progress to proteinuria and kidney failure in adolescence or early adulthood if left untreated. On biopsy, pathognomonic features include alternate thinning, thickening and lamellation of the glomerular basement membrane (GBM), in the presence of podocyte foot process effacement. Although previous studies indicate a prevalence of AS of about 1/50,000, a recent publication reported a predicted rate of pathogenic COL4A5 variants of 1/2320. We herewith present 98 patients (40 M/58 F) from 26 Greek families. We are selectively presenting the families segregating the X-linked form of AS with pathogenic variants in the COL4A5 gene. We found 21 different pathogenic variants, 12 novel: eight glycine and one proline substitutions in the collagenous domain, one cysteine substitution in the NC1 domain, two premature termination of translation codons, three splicing variants, one 5-bp insertion/frameshift variant, one indel-frameshift variant and four gross deletions. Notably, patients in six families we describe here and three families we reported previously, carried the COL4A5-p.G624D substitution, a founder defect encountered all over Europe which is hypomorphic with mostly milder symptomatology. Importantly, on several occasions, the correct genetic diagnosis reclassified patients as patients with AS, leading to termination of previous immunosuppressive/cyclosporine A therapy and a switch to angiotensin converting enzyme inhibitors (ACEi). With the understanding that all 98 patients span a wide range of ages from infancy to late adulthood, 15 patients (11 M/4 F) reached kidney failure and 11 (10 M/1 F) received a transplant. The prospects of avoiding lengthy diagnostic investigations and erroneous medications, and the advantage of delaying kidney failure with very early administration of renin-angiotensin-aldosterone system (RAAS) blockade, highlights the importance of timely documentation of AS by genetic diagnosis.
Chapter
While traditionally the function of the extracellular matrix and the basement membrane was considered to be providing structural support, it is now clear that this only covers one aspect of its multiple functions. This is also illustrated by our growing knowledge of the role of collagen IV, a major component of basement membranes, in development, health, and disease. With the extracellular matrix and collagen IV increasingly being recognised as key players in a growing number of diseases from stroke and vascular defects to kidney disease, deafness, and eye abnormalities, it is paramount that we increase our fundamental understanding of these complex molecules ranging from their biosynthesis to their role in human disease. Recently, exciting progress has been made in delineating the mechanisms by which mutations in collagen IV cause disease, and these are being exploited to develop mechanism-based treatments. Yet many important questions remain that need addressing to develop treatments for diseases associated with collagen IV.
Article
Full-text available
Background. Alport syndrome (AS) is a hereditary nephropathy characterized by progressive renal failure, hearing loss and ocular lesions. Numerous mutations of the COL4A5 gene encoding the α5‐chain of type IV collagen have been described, establishing the molecular cause of AS. The goal of the present study was to identify the genotype–phenotype correlations that are helpful in clinical counseling. COL4A5‐mutations (n=267) in males were analysed including 23 German Alport families. Methods. Exons of the COL4A5 gene were PCR‐amplified and screened by Southern blot, direct sequencing or denaturing gradient gel electrophoresis. Phenotypes were obtained by questionnaires or extracted from 44 publications in the literature. Data were analysed by Kaplan–Meier statistics, χ² and Kruskal–Wallis tests. Results. Genotype–phenotype data for 23 German Alport families are reported. Analysis of these data and of mutations published in the literature showed the type of mutation being a significant predictor of end‐stage renal failure (ESRF) age. The patients' renal phenotypes could be grouped into three cohorts: (1) large rearrangements, frame shift, nonsense, and splice donor mutations had a mean ESRF age of 19.8±5.7 years; (2) non‐glycine‐ or 3′ glycine‐missense mutations, in‐frame deletions/insertions and splice acceptor mutations had a mean ESRF age of 25.7±7.2 years and fewer extrarenal symptoms; (3) 5′ glycine substitutions had an even later onset of ESRF at 30.1±7.2 years. Glycine‐substitutions occurred less commonly de novo than all other mutations (5.5% vs 13.9%). However, due to the evolutionary advantage of their moderate phenotype, they were the most common mutations. The intrafamilial phenotype of an individual mutation was found to be very consistent with regards to the manifestation of deafness, lenticonus and the time point of onset of ESRF. Conclusions. Knowledge of the mutation adds significant information about the progress of renal and extrarenal disease in males with X‐linked AS. We suggest that the considerable prognostic relevance of a patient's genotype should be included in the classification of the Alport phenotype.
Article
Full-text available
Amniotic fluid is in continuity with multiple developing organ systems, including the kidney. Committed, but still stem-like cells from these organs may thus appear in amniotic fluid. We report having established for the first time a stem-like cell population derived from human amniotic fluid and possessing characteristics of podocyte precursors. Using a method of triple positive selection we obtained a population of cells (hAKPC-P) that can be propagated in vitro for many passages without immortalization or genetic manipulation. Under specific culture conditions, these cells can be differentiated to mature podocytes. In this work we compared these cells with conditionally immortalized podocytes, the current gold standard for in vitro studies. After in vitro differentiation, both cell lines have similar expression of the major podocyte proteins, such as nephrin and type IV collagen, that are characteristic of mature functional podocytes. In addition, differentiated hAKPC-P respond to angiotensin II and the podocyte toxin, puromycin aminonucleoside, in a way typical of podocytes. In contrast to immortalized cells, hAKPC-P have a more nearly normal cell cycle regulation and a pronounced developmental pattern of specific protein expression, suggesting their suitability for studies of podocyte development for the first time in vitro. These novel progenitor cells appear to have several distinct advantages for studies of podocyte cell biology and potentially for translational therapies.
Article
Full-text available
X-linked Alport syndrome is caused by mutations in the COL4A5 gene encoding the type IV collagen α5 chain (α5(IV)). Complete absence of α5(IV) in the renal basal membrane is considered a pathological characteristic in male patients; however, positive α5(IV) staining has been found in over 20% of patients. We retrospectively studied 52 genetically diagnosed male X-linked Alport syndrome patients to evaluate differences in clinical characteristics and renal outcomes between 15 α5(IV)-positive and 37 α5(IV)-negative patients. Thirteen patients in the α5(IV)-positive group had non-truncating mutations consisting of nine missense mutations, three in-frame deletions, and one splice-site mutation resulting in small in-frame deletions of transcripts. The remaining two showed somatic mutations with mosaicism. Missense mutations in the α5(IV)-positive group were more likely to be located before exon 25 compared with missense mutations in the α5(IV)-negative group. Furthermore, urinary protein levels were significantly lower and the age at onset of end-stage renal disease was significantly higher in the positive group than in the negative group. These results help to clarify the milder clinical manifestations and molecular characteristics of male X-linked Alport syndrome patients expressing the α5(IV) chain.Kidney International advance online publication, 4 December 2013; doi:10.1038/ki.2013.479.
Article
Full-text available
Nephrin, an important component of the podocyte filtration slit diaphragm, plays a key role in the maintenance of glomerular permselectivity. Mutations in nephrin lead to proteinuria and congenital nephrotic syndrome. Nephrin undergoes posttranslational modifications in the endoplasmic reticulum (ER) prior to export to the plasma membrane. We examined the effects of human nephrin disease-associated missense mutations on nephrin folding in the ER and on cellular trafficking in cultured cells. Compared with wild-type (WT) nephrin, the mutants showed impaired glycosylation and enhanced association with the ER chaperone, calnexin, as well as accumulation in the ER. Nephrin mutants demonstrated enhanced ubiquitination, and they underwent ER-associated degradation. Certain nephrin mutants did not traffic to the plasma membrane. Expression of nephrin mutants resulted in the stimulation of the activating transcription factor-6 pathway of the unfolded protein response, and an increase in the ER chaperone, Grp94. We treated cells with castanospermine (an inhibitor of glucosidase I) in order to decrease the association of nephrin mutants with calnexin. Castanospermine increased plasma membrane expression of nephrin mutants; however, full glycosylation and signaling activity of the mutants were not restored. Modulation of ER quality control mechanisms represents a potential new approach to development of therapies for proteinuric kidney disease, including congenital nephrotic syndrome.
Article
Full-text available
Thin-basement-membrane nephropathy (TBMN) and Alport syndrome (AS) are progressive collagen IV nephropathies caused by mutations in COL4A3/A4/A5 genes. These nephropathies invariably present with microscopic hematuria and frequently progress to proteinuria and CKD or ESRD during long-term follow-up. Nonetheless, the exact molecular mechanisms by which these mutations exert their deleterious effects on the glomerulus remain elusive. We hypothesized that defective trafficking of the COL4A3 chain causes a strong intracellular effect on the cell responsible for COL4A3 expression, the podocyte. To this end, we overexpressed normal and mutant COL4A3 chains (G1334E mutation) in human undifferentiated podocytes and tested their effects in various intracellular pathways using a microarray approach. COL4A3 overexpression in the podocyte caused chain retention in the endoplasmic reticulum (ER) that was associated with activation of unfolded protein response (UPR)-related markers of ER stress. Notably, the overexpression of normal or mutant COL4A3 chains differentially activated the UPR pathway. Similar results were observed in a novel knockin mouse carrying the Col4a3-G1332E mutation, which produced a phenotype consistent with AS, and in biopsy specimens from patients with TBMN carrying a heterozygous COL4A3-G1334E mutation. These results suggest that ER stress arising from defective localization of collagen IV chains in human podocytes contributes to the pathogenesis of TBMN and AS through activation of the UPR, a finding that may pave the way for novel therapeutic interventions for a variety of collagenopathies.
Article
Full-text available
The COL4A3-/- mouse serves as animal model for progressive renal fibrosis. Using this animal model, the present study investigates the nephroprotective effects of Paricalcitol versus Calcitriol alone and on top of ACE-inhibitor therapy. Eighty six mice were divided into six groups: (PC) with Paricalcitol 0.1 mcg/kg, (CA) Calcitriol 0.03 mcg/kg (dose equipotent), (PLAC) vehicle 0.1 mL i.p. five times per week, (ACE + PC) Paricalcitol plus Ramipril, (ACE + CA) Calcitriol plus Ramipril and (ACE + PLAC) vehicle plus Ramipril 10 mg/kg/day p.o. ACE therapy started pre-emptively in Week 4, PC/CA therapy was initiated in 6-week-old animals with ongoing renal fibrosis and lasted for 8 weeks. Four to six animals were sacrificed after 9.5 weeks and kidneys were further investigated using histological, immunohistological and Western-blot techniques. Survival until end-stage renal failure was determined in the remaining animals. PC, but not CA, prolonged lifespan until renal failure by 13% compared with untreated controls (P = 0.069). ACE-inhibition prolonged lifespan by >50%. Added on top of ACE inhibition, ACE + PC (but not ACE + CA) even further prolonged lifespan by additional 18.0% (P < 0.01 versus ACE + PLAC) and improved renal function (blood urea nitrogen; P < 0.05 versus ACE + CA). Accumulation of extracellular matrix and renal scarring was decreased in PC and ACE + PC-treated mice. The present study demonstrated a substantial nephroprotective and antifibrotic effect of the vitamin D-receptor activator Paricalcitol on top of early ACE inhibition in the COL4A3-/- model of progressive kidney fibrosis. The synergistic effect of Paricalcitol on top of RAAS-blockade might as well be valuable in other chronic kidney diseases.
Article
Full-text available
Haemorrhagic stroke accounts for approximately 20% of stroke cases and porencephaly is a clinical consequence of perinatal cerebral haemorrhaging. Here we report the identification of a novel dominant G702D mutation in the collagen domain of COL4A2 (collagen IV alpha chain 2) in a family displaying porencephaly with reduced penetrance. COL4A2 is the obligatory protein partner of COL4A1 but in contrast to most COL4A1 mutations, the COL4A2 mutation does not lead to eye or kidney disease. Analysis of dermal biopsies from patient and his unaffected father, who also carries the mutation, revealed that both display basement membrane (BM) defects. Intriguingly, defective collagen IV incorporation into the dermal BM was only observed in the patient and was associated with endoplasmic reticulum (ER) retention of COL4A2 in primary dermal fibroblasts. This intracellular accumulation led to ER-stress, unfolded protein response activation, reduced cell proliferation and increased apoptosis. Interestingly, absence of ER retention of COL4A2 and ER-stress in cells from the unaffected father indicate that accumulation and/or clearance of mutant COL4A2 from the ER may be a critical modifier for disease development. Our analysis also revealed that mutant collagen IV is degraded via the proteasome. Importantly, treatment of patient cells with a chemical chaperone decreased intracellular COL4A2, ER-stress and apoptosis, demonstrating that reducing intracellular collagen accumulation can ameliorate the cellular phenotype of COL4A2 mutations. Importantly, these data highlight that manipulation of chaperone levels, intracellular collagen accumulation and ER-stress are potential therapeutic options for collagen IV diseases including haemorrhagic stroke.
Article
Embryonic stem (ES) cells have the potential to differentiate into various progenitor cells. Here we investigated the capacity of mouse ES cells to differentiate into renal tubular cells both in vitro and in vivo. After stably transfecting Wnt4 cDNA to mouse ES cells (Wnt4-ES cells), undifferentiated ES cells were incubated by the hanging drop culture method to induce differentiation to embryoid bodies (EBs). During culturing of the EBs derived from the Wnt4-ES cells, aquaporin-2 (AQP2) mRNA and protein were expressed within 15-20 days. The expression of AQP2 in Wnt4-EBs was enhanced in the presence of hepatocyte growth factor (HGF) and activin A. We next performed in vivo experiments by transplanting the Wnt4-EBs into the mouse renal cortex. Four weeks after transplantation, some portions of the EB-derived cells expressing AQP2 in the kidney assembled into tubular-like formations. In conclusion, our in vitro and in vivo experiments revealed two new findings: first, that cultured Wnt4-EBs have an ability to differentiate into renal tubular cells; and second, that Wnt4, HGF, and activin A may promote the differentiation of ES cells to renal tubular cells. (c) 2005 Elsevier Inc. All rights reserved.
Article
Alport syndrome is a hereditary glomerular disease that leads to kidney failure. It is caused by mutations affecting one of three chains of the collagen α3α4α5(IV) heterotrimer, which forms the major collagen IV network of the glomerular basement membrane (GBM). In the absence of the α3α4α5(IV) network, the α1α1α2(IV) network substitutes, but it is insufficient to maintain normal kidney function. Inhibition of angiotensin-converting enzyme slows progression to kidney failure in patients with Alport syndrome but is not a cure. Restoration of the normal collagen α3α4α5(IV) network in the GBM, by either cell- or gene-based therapy, is an attractive and logical approach toward a cure, but whether or not the abnormal GBM can be repaired once it has formed and is functioning is unknown. Using a mouse model of Alport syndrome and an inducible transgene system, we found that secretion of α3α4α5(IV) heterotrimers by podocytes into a preformed, abnormal, filtering Alport GBM is effective at restoring the missing collagen IV network, slowing kidney disease progression, and extending life span. This proof-of-principle study demonstrates the plasticity of the mature GBM and validates the pursuit of therapeutic approaches aimed at normalizing the GBM to prolong kidney function.
Article
Access to human pluripotent cells theoretically provides a renewable source of cells that can give rise to any required cell type for use in cellular therapy or bioengineering. However, successfully directing this differentiation remains challenging for most desired endpoints cell type, including renal cells. This challenge is compounded by the difficulty in identifying the required cell type in vitro and the multitude of renal cell types required to build a kidney. Here we review our understanding of how the embryo goes about specifying the cells of the kidney and the progress to date in adapting this knowledge for the recreation of nephron progenitors and their mature derivatives from pluripotent cells.