Vous êtes sur la page 1sur 8

REVIEWS

The glomerular basement membrane


as a barrier to albumin
Jung Hee Suh and Jeffrey H. Miner
Abstract | The glomerular basement membrane (GBM) is the central, non-cellular layer of the glomerular
filtration barrier that is situated between the two cellular components—fenestrated endothelial cells and
interdigitated podocyte foot processes. The GBM is composed primarily of four types of extracellular matrix
macromolecule—laminin‑521, type IV collagen α3α4α5, the heparan sulphate proteoglycan agrin, and
nidogen—which produce an interwoven meshwork thought to impart both size-selective and charge-selective
properties. Although the composition and biochemical nature of the GBM have been known for a long time,
the functional importance of the GBM versus that of podocytes and endothelial cells for establishing the
glomerular filtration barrier to albumin is still debated. Together with findings from genetic studies in mice,
the discoveries of four human mutations affecting GBM components in two inherited kidney disorders, Alport
syndrome and Pierson syndrome, support essential roles for the GBM in glomerular permselectivity. Here,
we explain in detail the proposed mechanisms whereby the GBM can serve as the major albumin barrier and
discuss possible approaches to circumvent GBM defects associated with loss of permselectivity.

Suh, J. H. & Miner, J. H. Nat. Rev. Nephrol. 9, 470–477 (2013); published online 18 June 2013; doi:10.1038/nrneph.2013.109

Introduction
The glomerular basement membrane (GBM) is a thin diabetes mellitus. About 40% of individuals with diabetes
(250–400 nm) meshwork of extracellular matrix pro- develop diabetic nephropathy, which then leads to more
teins that is an integral part of the glomerular filtration patients with chronic kidney disease in need of dialysis.7
barrier. Most of the GBM is situated between two cellular Although it is clear that proteinuria and renal failure
layers—glomerular endothelial cells and podocytes—in originate from both genetic and environmental factors,
the peripheral capillary wall (Figure 1); the remaining in all but a few cases it is very difficult to clearly define
GBM segments lie between mesangial cells and podo- a genetic component. Much research has focused on the
cytes at the bases of the capillary loops.1 The GBM both cellular components of the glomerulus—the podocytes,
provides structural support for the glomerular capillar- endothelial cells, and mesangial cells—because they can
ies and harbours ligands for receptors on the surface of actively respond to genetic and environmental changes
the adjacent endothelial cells, podocytes, and mesangial by producing gene products and cell-signalling mol-
cells.2,3 Importantly, the GBM also contributes to glomer- ecules. However, changes in these cells can also give rise
ular permselectivity: as the second layer of the capillary to changes in the GBM, which can secondarily affect
wall that is encountered by filtrate, it is thought to restrict the properties and behaviour of the neighbouring cells
the passage of plasma proteins across the glomerular fil- through matrix-to-cell (outside-in) signalling events.
tration barrier. In support of this idea, of the nine major Similarly, primary changes in the GBM may exert func-
proteins found in the GBM, mutations in genes encod- tionally important effects on the neighbouring podo-
ing four of them are known to cause human kidney dis- cytes, endothelial cells, and mesangial cells, thereby
eases4,5 (Alport syndrome and Pierson syndrome) that affecting glomerular filtration.
involve proteinuria—the leakage of valuable plasma Whether and how the GBM contributes to the estab-
protein, mostly albumin, into the urine. lishment and function of the glomerular filtration barrier
Although mutations affecting GBM components to protein have been debated for several decades.8 More
are important causes of kidney disease, environmen- recent findings gleaned from genetic and physiological
Renal Division, tal changes that affect the glomerulus can also lead to studies have provided a better view of how the GBM could
Department of Internal alterations in the composition and structure of the GBM. function as a barrier. Further understanding the mecha-
Medicine, Washington
University School of
Diabetic nephropathy is one example in which the GBM is nisms in various disease models could help in the design
Medicine, 660 South adversely affected by the microenvironment.6 Diabetic of therapeutics that could prevent or reverse proteinuria
Euclid Avenue, St Louis, nephropathy is becoming more and more prevalent as by impacting GBM structure and function.
MO 63110, USA
(J. H. Suh, J. H. Miner). a result of the worldwide increases in obesity and type 2 This Review focuses mainly on the mechanisms by
which the GBM functions to establish and maintain the
Correspondence to:
J. H. Miner Competing interests glomerular filtration barrier. From results from genetic
minerj@wustl.edu The authors declare no competing interests. and biochemical studies in mice and humans, it is evident

470  |  AUGUST 2013  |  VOLUME 9  www.nature.com/nrneph


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

that the GBM is crucial to prevent the leakage of plasma Key points
proteins into the urine. We emphasize the critical role of
■■ The glomerular basement membrane (GBM) is the extracellular matrix
the GBM as a permselective barrier that can be altered in
component of the glomerular filtration barrier; it is flanked by the podocyte and
different ways by genetic defects that cause kidney disease. glomerular endothelial cell layers
■■ The major GBM components are laminin‑521, type IV collagen α3α4α5, nidogen,
GBM components’ role in permselectivity and the heparan sulphate proteoglycan agrin
In order to understand how the GBM might contrib- ■■ Mutations in COL4 genes that result in absence of the type IV collagen α3α4α5
ute to permselectivity, it is important to define its com- network cause Alport syndrome, a hereditary nephritis accompanied by hearing
position and to understand the properties of its major defects
■■ Mutations in laminin β2 (LAMB2) cause Pierson syndrome, a congenital
components. Like all basement membranes, the GBM is
nephrotic syndrome with associated eye and neurologic abnormalities
composed of laminin, type IV collagen, heparan sulphate ■■ Studies using mouse models of Pierson and Alport syndromes have shown that
proteoglycan, and nidogen.9 Components of the GBM are the defective GBM is more permeable to macromolecules than is the normal
synthesized by both podocytes and endothelial cells,10 and GBM, suggesting that it has a role in permselectivity
during glomerulogenesis the separate podocyte-derived
and endothelium-derived basement membranes fuse to
form the immature GBM. This feature is responsible at assemble to form at least fifteen distinct laminin trimers;16
least in part for the GBM being thicker than most other depending on the α–β–γ chain composition, these laminin
basement membranes, as both cell layers synthesize extra- trimers resemble cruciform, Y‑shaped, or rod-shaped
cellular matrix components and secrete them into the structures. Each chain has a laminin coiled-coil domain,17
extracellular space between them. Furthermore, one can and interactions between three coiled-coil domains and
infer that changes in either podocytes or endothelial cells limited interchain covalent bonding contribute to the for-
can result in altered GBM composition—and vice versa— mation of the long arm of all laminin trimers.18 The
possibly affecting the function of the glomerular filtra- remaining segment of each chain is called a short arm.
tion barrier. In this context, podocytes, endothelial cells, The short arms of the cruciform trimers contain a laminin
and the GBM can be viewed as being inter­connected; N‑terminal (LN) domain, which has an essential role in the
this idea is evident not only through the obvious direct polymerization of trimers to form a network.19,20 Laminin α
physical contacts within cell layers and between cells and chains are unique because they also contain a C‑terminal
the GBM, but also across the GBM through both cell– laminin globular (LG) domain, composed of five tandem
matrix–cell connections and cell–cell communication sub-domains, which is situated distal to the long arm.21
and/or signalling via diffusible factors. One excellent LG domains link laminin trimers in basement mem-
example of the latter is the vascular endothelial growth branes to neighbouring cells by serving as ligands for two
factor (VEGF) signalling axis, in which podocyte-derived major cellular receptors, integrins and dystroglycan.22,23
VEGF is crucial for endothelial cell homeostasis.11
Historically, the GBM was considered by some
researchers to be a ‘crude prefilter’, and slit diaphragms Urinary
space
between podocyte foot processes were thought to be Podocyte
responsible for the bulk of glomerular permselectivity.12
However, as proposed by Farquhar and Palade, functional
and physiological analyses of the glomerular filtration
barrier using various tracers revealed the importance of
the GBM as a size-selective and charge-selective filtration
FP
barrier.13–15 When neutral tracers of various sizes were
intravenously injected, molecules larger than albumin GBM
were found to be restricted to the inside of the glomeru-
lar capillary loops and were impaired from traversing
the GBM. Likewise, when neutral, anionic and cationic Endothelial
Albumin
tracers were infused, negatively charged molecules had cell
Capillary lumen
the most difficulty in crossing the GBM.15 From these
studies, the function and necessity of the components of GBM components
Laminin-521
the GBM were inferred based on their biochemical prop- Type IV collagen α3α4α5
Nidogen
erties. However, the most direct evidence as to whether HSPG
each component of the GBM is essential for establish-
ing the filtration barrier was provided by mouse genetic Figure 1 | The components of the GBM. The normal GBM is
studies and the discovery of human mutations that cause composed of laminin‑521 (α5β2γ1), type IV collagen α3α4α5,
defects in glomerular permselectivity. nidogen and HSPG (primarily agrin). As described in the main
text, podocytes and endothelial cells each contribute at least
a subset of these components to the GBM (arrows). Most of
Laminins the plasma albumin (turquoise circles) is restricted to the
Laminins are large heterotrimeric glycoproteins com- capillary lumen. Abbreviations: FP, foot processes; GBM,
posed of three different homologous chains: α, β, and γ. glomerular basement membrane; HSPG, heparan
In humans, five α chains, four β chains, and three γ chains sulphate proteoglycan.

NATURE REVIEWS | NEPHROLOGY VOLUME 9  |  AUGUST 2013  |  471


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Integrins are transmembrane αβ heterodimers that are of collagen IV displays multiple interruptions, imparting
crucial in many different contexts for cell-to-extracellular flexibility to the collagen IV protomer and to the network
matrix signalling and vice versa, as well as for direct cell– that it forms in basement membranes.
cell signalling.24 Integrin α3β1 is the predominant integ- Each collagen IV α chain has three domains: the
rin normally present on the basal surface of podocytes, N‑terminal 7S domain, the collagenous domain con-
and deletion of α3β1 results in severe kidney glomerular taining the interrupted Gly‑X-Y repeats (with X and
defects during development.25 Integrin α3β1 binds to Y usually being Lys or Pro), and the noncollagenous
laminin α5β1γ1 (LM‑511) and α5β2γ1 (LM‑521) through domain (NC1) at the C‑terminus. Collagen IV proto­
the α5 chain’s LG domain.26 The binding of α5LG to inte- mers are assembled inside the endoplasmic reticulum
grin α3β1 is essential for the formation of the typical and secreted into the extracellular space. There, they self-
glomerular capillary loop structure, probably because polymerize into a ‘chicken-wire-like’ network through
the mesangial cells that organize the capillaries also hexameric and dodecameric interactions involving the
express integrin α3β1.27 Dystroglycan also binds to the NC1 and 7S domains, respectively, and become heavily
LG domain of α chains,28 but the deletion of dystroglycan cross-linked through disulphide bonds, sulfilimine
from most kidney cells does not result in a kidney defect, bonds, and lysyl-oxidase-mediated crosslinks.34,35
suggesting that integrin α3β1 has a more important role In the GBM, a transition in the composition of colla-
than dystroglycan in extracellular matrix-to-cell adhesion gen IV chains occurs, from α1α1α2 in the immature GBM
and signalling in the kidney.29 to α3α4α5 in the mature GBM.36 This transition occurs
Secretory signal peptides are located at the N‑terminus coincidentally with the transition of laminin chains in
of each laminin chain, which targets them to the endoplas- the GBM. The molecular mechanisms controlling the
mic reticulum as they are synthesized. Laminin trimers switch of collagen IV and laminin chains in the GBM are
assemble and become glycosylated in the endoplasmic unknown. However, the collagen IV transition might be
reticulum and are processed further in the Golgi appara- required to accommodate the increased blood pressure
tus.30 After secretion into the extracellular space, laminin in adults, since α3α4α5 type IV collagen produces a more
trimers self-polymerize to form a laminin network by heavily cross-linked and more protease-resistant network
interactions between LN domains.20 This polymeriza- compared to the α1α1α2 type IV collagen network.37 The
tion step is a reversible and calcium-dependent process importance of the α3α4α5 type IV collagen network in
that requires a critical concentration of laminin trimers the glomerular capillary wall is illustrated by the fact
to form an initiating complex.19 This initial step is facili- that its absence causes Alport syndrome,35 a glomerular
tated by the binding of laminin trimers to laminin recep- disease that will be discussed in detail.
tors via their α‑chain LG domains,31 which increases the
local concentration of laminin trimers and helps trigger Nidogen
laminin polymerization. Interactions that involve the The nidogens, also known as entactins, are two homolo-
other basement membrane molecules (type IV collagen, gous glycoproteins containing three globular-like domains
nidogen, and sulphated proteoglycan) then enable the with two rod-like domains separating the globular-like
assembly of a basement membrane. Interestingly, mice domains.38 Nidogen‑1 and nidogen‑2, both found in the
that lack α1 and α2 type IV collagen do assemble base- GBM, are encoded by two different genes. Nidogen‑1
ment membranes during early embryonic development, binds to both laminin γ1 and type IV collagen, a finding
but these basement membranes are unstable, leading to that led to the hypothesis that nidogen acts as a bridge
embryonic death by embryonic day 11.32 However, the linking the separate laminin and type IV collagen net-
deletion of laminin γ1 (a component of all early laminins) works in basement membranes. 39 Deletion of either
in mice caused much earlier lethality (by embryonic nidogen gene in mice does not cause any significant
day 5.5) and the total absence of basement membranes.33 abnormalities, probably because of overlapping expression
These data indicate that laminin is indispensible for and redundant functions.40–43 Nidogen‑1 and nidogen‑2
the initial formation of basement membranes, whereas double-knockout mice, however, exhibit perinatal letha­
type IV collagen is not. In addition, the quantity of effi- lity due to lung and heart malformations, but many base-
ciently polymerizing laminin in the GBM is important for ment membranes form normally without nidogen, despite
restricting the passage of plasma macromolecules across the ability of nidogen to link laminin and type IV colla-
the glomerular filter, as will be discussed. gen networks.44 The data suggest that nidogens provide
extra stability to basement membranes under situations
Type IV collagen of unusual stress, but that they are not required for the
Type IV collagen is the most abundant protein found initial formation of basement membranes. As no defini-
in basement membranes, comprising about 50% of total tive evidence exists to indicate that either nidogen alone
protein mass. Six genetically distinct collagen IV α chains contributes to the properties of the GBM as a barrier, they
exist—α1 through α6—and these chains assemble to will not be discussed in that context.
form three different heterotrimers referred to as proto­
mers: α1α1α2, α3α4α5 and α5α5α6. Like all collagen Heparan sulphate proteoglycan
chains, the collagen IV chains contain Gly‑X-Y amino Heparan sulphate proteoglycans have sulphated glycos­
acid triplet repeats. However, unlike fibril-forming col- aminoglycan side chains linked to a protein core.
lagens of bone and cartilage, the Gly‑X-Y repeat region Although perlecan seems to be the prominent heparan

472  |  AUGUST 2013  |  VOLUME 9  www.nature.com/nrneph


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

sulphate proteoglycan in most basement membranes39 a 1 week b 2 weeks


and in the mesangial matrix, agrin is the major heparan
sulphate proteoglycan in the GBM.45 The sulphated
Podocyte
glycosaminoglycan side chains of agrin give it a highly
negative charge that is reflected by anionic sites within
the GBM that can be detected with cationic probes such
as polyethyleneimine and cationized ferritin.46,47 The
FP
N‑terminal domain of agrin binds avidly to the LM‑521
GBM
long arm, and its C‑terminus bears domains that can
bind to cell-­surface receptors such as dystroglycan and
integrins. 48 These properties of agrin suggest that it Albumin Endothelial
cell
could have importance in mediating charge selectivity
within the glomerular filtration barrier and in linking the GBM components
GBM to the adjacent cells. In addition, heparan sulphate Laminin-111, laminin-211, laminin-332, laminin-511
Type IV collagen α3α4α5
side chains are important for binding and sequestering Nidogen
growth factors in some contexts; one such growth factor HSPG
secreted by podocytes that must cross the GBM and
Figure 2 | Mutations in Lamb2 result in albuminuria in a mouse model of Pierson
could benefit from the presence of such side chains is syndrome. The GBM of Lamb2-knockout mice lacks laminin‑521 and instead contains
VEGF.11,49 However, podocyte-specific mutation of the ectopic laminins (shown in grey text), such as laminin‑111, laminin‑211, laminin‑332,
gene encoding agrin in mice did not lead to any struc- and laminin‑511. However, the laminin network made up of these ectopic laminins is
tural or functional defects in the glomerulus, although defective, leading to increased passage of plasma protein such as albumin (turquoise
it did greatly reduce the density of negatively charged circles) across the barrier. a | At 1 week of age, Lamb2-null mice have proteinuria due
sites within the GBM and cause a dramatic reduction in to the defective GBM, but without podocyte foot process effacement. b | At 2 weeks,
the level of full-length agrin protein in the GBM. These podocyte abnormalities can be detected, followed by increasing proteinuria and
widespread effacement. These results indicate that proteinuria precedes podocyte
results suggest that agrin and the negative charge of the
abnormalities in Lamb2-null mice, highlighting the importance of the GBM as a barrier
GBM do not play critical roles in the filtration barrier.46 to plasma protein. Abbreviations: FP, foot processes; GBM, glomerular basement
Furthermore, the deletion of both agrin and the heparan membrane; HSPG, heparan sulphate proteoglycan.
sulphate side chains on perlecan also had little if any
effect on permselectivity,50 despite the fact that a previ-
ous report had revealed a potential role for perlecan in the gene encoding laminin β2.53 Follow-up of Pierson
filtration in the context of protein overload.51 No critical et al.’s original cases via screening of healthy living family
role for agrin in the GBM, either as a matrix protein or members revealed two different mutations in LAMB2,
as an anionic macromolecule, has been proven, so agrin suggesting that the original cases were compound
will not be discussed further in relation to the glomerular hetero­zygotes.54 These findings highlight the critical
filtration barrier. role of laminin β2 for normal kidney function, especially
glomerular filtration barrier function.
Diseases associated with GBM malfunction The recognition of Pierson syndrome as a specific
As mentioned earlier, two relatively well understood disease entity by the discovery of LAMB2 mutations
genetic kidney diseases are known to target components spurred more reports of cases of Pierson syndrome.5,55 With
of the GBM: Pierson syndrome and Alport syndrome. increasing numbers of reports, it has become clear that
Although these two diseases both involve primary defects the onset and severity of Pierson syndrome varies greatly
in GBM components, they have very different clini- between patients in terms of progression of nephrotic syn-
cal presentations; these differences provide some clues drome and extra-renal manifestations, such as muscular
as to the specific roles of laminin and type IV collagen hypotonia and neurodevelopmental deficits, resulting in a
networks in influencing glomerular permselectivity. ‘spectrum’ of Pierson syndrome with genotype–phenotype
correlations.56,57 Truncating mutations, which are dispersed
Pierson syndrome throughout the LAMB2 gene, are generally associated with
In the early 1960s, Pierson et al. described two infants more severe phenotypes than are missense mutations, as
from one family with microcoria and congenital truncated proteins often lack the coiled-coil domain that
nephrotic syndrome.52 Both infants presented with severe is critical for the assembly of LM‑521 trimers.5 By contrast,
congenital nephrotic syndrome and early-onset end- pathogenic missense mutations in LAMB2 are found pri-
stage renal disease (ESRD), and died 2 weeks after birth. marily in the LN domain, probably resulting in defects in
The iris dilator muscles in their eyes showed aplasia and/ laminin polymerization and/or secretion.5,58,59 Thus, for
or atrophy and an abnormal lens. In 2004, Zenker and patients in the Pierson syndrome spectrum, the muta-
colleagues reported a case of congenital nephrotic syn- tions in LAMB2 result in a total absence or reduced level
drome with clinical features that included microcoria of LM‑521 in the GBM or in the formation and secretion of
and mesangial sclerosis, similar to the features described dysfunctional LM‑521 trimers.
by Pierson, and the condition was termed Pierson syn- The straightforward interpretation of the mechanism of
drome.53 Homozygosity mapping of five affected fami- proteinuria in Pierson syndrome is that the lack of LM‑521
lies identified autosomal recessive mutations in LAMB2, causes a defect in the ability of the GBM to attenuate the

NATURE REVIEWS | NEPHROLOGY VOLUME 9  |  AUGUST 2013  |  473


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

proper foot processes with slit diaphragms in functioning


glomeruli, at least in early stages of development. In mice,
Podocyte
and presumably also in humans, foot process effacement
follows later (shown at 2 weeks of age for mice in Figure 2)
and may even be caused by the increasing proteinuria that
eventually reaches nephrotic range.
In further support of a direct role for the GBM in
mediating permselectivity, the use of ferritin as an
electron-­dense tracer to monitor the permeability of the
GBM to plasma macromolecules showed that the lack
of LM‑521 in Lamb2-null mice was associated with an
increase in GBM permeability relative to the permeabil-
GBM
ity of the GBM in control littermates; this increase was
apparent before widespread foot process effacement and
loss of slit pores.62 Similar to previously reported results
Endothelial using albumin and ferritin tracing in situ,13,14,63 no accu-
Albumin cell mulation of ferritin occurred below the slit diaphragms,
suggesting that the slit diaphragms do not act as restric-
GBM components tive pores in the physiological state.62 These results are,
Laminin-521, laminin-111, laminin-211, laminin-221, laminin-511
Type IV collagen α1α1α2 however, in conflict with structural studies of the slit
Nidogen diaphragm that show the pore sizes to be of similar size
HSPG
to or smaller than the size of albumin.64
Figure 3 | Mutations in the genes encoding type IV collagen
α3, α4, or α5 result in albuminuria in a mouse model of Alport syndrome
Alport syndrome. The GBM of Col4a3-mutant mice lacks the Alport syndrome is a hereditary glomerular, auditory,
type IV collagen α3α4α5 network. Although there is and ocular disease caused by mutations in the COL4A3,
increased deposition of type IV collagen α1α1α2 (shown in
COL4A4, or COL4A5 genes that encode the type IV col-
grey text) as a compensatory mechanism, the resulting GBM
becomes split and thickened and accumulates multiple
lagen α3, α4, and α5 chains, respectively.4,65 Because the
ectopic laminins (shown in grey text). Eventually, loss of collagen IV network of the GBM consists primarily of
plasma protein such as albumin (turquoise circles) across cross-linked α3α4α5(IV) protomers made by podocytes,66
the filtration barrier increases and proteinuria occurs. the lack of any one of the three chains (for example, as
Abbreviations: GBM, glomerular basement membrane; a result of a null mutation) prevents the protomer from
HSPG, heparan sulphate proteoglycan. forming. Moreover, missense mutations—particularly
those that cause glycine substitutions in the collagen-
passage of albumin from the plasma to the Bowman space. ous domain—can lead to defects in protomer structure
Given the evidence for the involvement of both podo- and in the collagen IV network and therefore also cause
cytes and endothelial cells in regulating permselectivity, Alport syndrome.
however, an alternative hypothesis is that defects in the In the absence of the α3α4α5(IV) network, a com-
GBM prevent proper cell–matrix interactions and sig- pensatory increase occurs in the abundance of the
nalling to cells, resulting in cellular defects that lead to α1α1α2(IV) network, which is normally a minor com-
proteinuria. The rigorous investigation of these hypoth- ponent in the subendothelial aspect of the mature human
eses requires experiments that cannot be performed in GBM and is barely detectable in the normal mouse GBM.
humans. Fortunately, mice with a null mutation in Lamb2 This compensation enables the GBM to form and func-
have provided an excellent model in which to study tion properly for several years in humans, but eventu-
Pierson syndrome, as they recapitulate the congenital ally haematuria occurs that accompanies a characteristic
nephrotic syndrome and die at about 3 weeks of age with splitting and thickening of the GBM, as also occurs in
severe proteinuria and neuromuscular defects.60,61 They Alport mice (Figure 3). These changes are followed
also show accumulation of ectopic laminin chains in the by the onset of protein­uria that increases and signifies
GBM, including α1, α2, α3, β1, β3, and γ2 (Figure 2).62 declining glomerular filtration rate (GFR). The majority
This deposition of ectopic laminins into the GBM may of patients, most of whom are males carrying X‑linked
be a compensatory response to the loss of laminin β2 and COL4A5 mutations, eventually require renal replacement
thus LM‑521, but it is not sufficient to establish a fully therapy. By contrast, only a minority of female carriers of
functional glomerular filtration barrier.62 Interestingly, X‑linked Alport syndrome progress to ESRD. The rarer
the glomerular ultrastructure of proteinuric 1‑week-old autosomal forms of Alport syndrome affect males and
Lamb2-null mice reveals intact podocyte foot processes females equally.
(Figure 2) and slit diaphragms, suggesting a role for the The fact that proteinuria appears late in the disease
GBM as an independent and indispensible filtration process suggests that collagen IV is not as important as
barrier to plasma proteins.62 Importantly, these data show laminin for maintaining glomerular permselectivity.
that proper GBM laminin composition is not required However, studies of a mouse model of Alport syndrome
for what appears to be the elaboration by podocytes of show that the GBM in mice with Alport syndrome is

474  |  AUGUST 2013  |  VOLUME 9  www.nature.com/nrneph


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

more permeable to ferritin than is the GBM from normal into the GBM prevented the nephrotic syndrome that
mice, indicating that collagen IV is important for the fil- would have otherwise developed, and the mice lived
tration barrier.67 The areas of increased permeability also a long life.73 These data also suggest that in the GBM
showed ectopic deposition of laminin α1 plus increased the quantity of laminin is important, as LM‑511 could
levels of laminin α5,67 suggesting the possibility that sec- efficiently substitute for LM‑521 if expressed at a high
ondary changes in the laminin network, together with enough level. An exciting conclusion from this study
the defect in the collagen IV network, might be responsi- is that upregulation of the unaffected LAMB1 gene in
ble for the increased permeability of the GBM that occurs the podocytes of patients with Pierson syndrome may
in Alport syndrome. The fact that proteinuria appears be an effective therapy. Moreover, we have shown that
late in the course of the disease might be because any secretion of a secretion-defective mutant laminin β2
early increase in the level of filtered albumin is masked (C321R-LAMB2) could be improved in vitro by treat-
by increased albumin uptake by proximal tubular cells. ment with a chemical chaperone, 74 which promotes
Treatment of Alport syndrome in mice and humans proper protein folding. This result suggests that such
with angiotensin-converting-enzyme inhibitors has been a chemical chaperone could be a possible drug therapy
shown to slow the onset of proteinuria and the decline in for patients with Pierson syndrome who have missense
GFR.68,69 A reduction in blood pressure should put less LAMB2 mutations that impair protein folding and/or
stress on the defective, imperfectly cross-linked GBM, secretion. Additional studies in mice suggest that the
on the adjacent podocyte, and on the entire glomeru- three C‑terminal segments of the LG domain of laminin
lus, which exhibits increased deformability.70 Moreover, α5 are critical for a proper filtration barrier,75 for reasons
biomechanical strain has been proposed to change gene that are not yet understood. Future studies concentra­
expression in ways that exacerbate glomerular disease in ting on the possible mechanisms for this finding could
Alport syndrome.71 reveal novel approaches for tightening the glomerular
barrier to albumin and reducing albuminuria.
Conclusions Given the well-demonstrated importance of multiple
So what are the mechanisms whereby laminin and podocyte proteins for proper foot process architecture
collagen IV might be directly involved in glomeru- and for permselectivity (as is discussed in detail else-
lar perm­s electivity? According to concepts proffered where76), it is important to determine how this role can
by Smithies,72 the GBM behaves like a concentrated be reconciled with the concept that the GBM is a major
gel that has size-selective properties and into which contributor to the physical filtration barrier. Based on
macro­molecules such as albumin permeate primarily cell biological concepts put forth previously,77 we propose
by diffusion. As the major components of the GBM, that the reason that the podocyte cytoskeleton and slit
the laminin‑521 and collagen IV networks, in concert diaphragms are so critical for normal filtration is because
with the other GBM components, likely impart the they are connected (either directly or indirectly) to cell
GBM with its characteristic porosity. By analogy to surface receptors—primarily integrins—that link to the
the polyacrylamide gel, which also has size-selective GBM via LM‑521. In turn, these receptors are critical for
properties, it is easy to understand the following: firstly, organizing the arrangement of the GBM’s components
how reducing laminin or type IV collagen concentra- and can thereby regulate its architecture, its porosity, and
tion might be similar to reducing the percentage of thus its permselectivity.
acrylamide; and secondly, how changing laminin or
type IV collagen isoforms might be similar to changing
Review criteria
the ratio of acrylamide to bis-acrylamide. The result of
either of these scenarios could be increased permeabi­lity The articles cited in this Review were found by literature
searches of the PubMed database using search
via increased pore sizes. Consistent with this idea, our
terms including the following: “glomerular basement
hypothesis that Lamb2-null mice develop nephrotic syn- membrane”, “glomerular basement membrane disease”,
drome owing to a paucity of laminin in the GBM, and “laminin kidney”, “collagen IV kidney”, “proteoglycan
therefore a defective laminin network in the GBM, is kidney”, “Pierson syndrome”, and “Alport syndrome”.
supported by our data from transgenic mice with Papers published after 1990 were the major ones
podocyte-­specific overexpression of laminin β1 and thus consulted. Only full-text English-language papers were
LM‑511. On the Lamb2-null background, the forced considered; reference lists therein were not major
sources for the content presented.
secretion of high levels of LM‑511 from podocytes

1. Miner, J. H. Organogenesis of the kidney 4. Kruegel, J., Rubel, D. & Gross, O. Alport a mechanistic viewpoint. Kidney Int. 74, 22–36
glomerulus: focus on the glomerular basement syndrome—insights from basic and clinical (2008).
membrane. Organogenesis 7, 75–82 (2011). research. Nat. Rev. Nephrol. 9, 170–178 7. de Boer, I. H. et al. Temporal trends in the
2. Yurchenco, P. D. & Patton, B. L. Developmental (2013). prevalence of diabetic kidney disease in the
and pathogenic mechanisms of basement 5. Matejas, V. et al. Mutations in the human laminin United States. JAMA 305, 2532–2539 (2011).
membrane assembly. Curr. Pharm. Des. 15, beta2 (LAMB2) gene and the associated 8. Farquhar, M. G. The glomerular basement
1277–1294 (2009). phenotypic spectrum. Hum. Mutat. 31, 992–1002 membrane: not gone, just forgotten. J. Clin.
3. Miner, J. H. Building the glomerulus: a (2010). Invest. 116, 2090–2093 (2006).
matricentric view. J. Am. Soc. Nephrol. 16, 6. Jefferson, J. A., Shankland, S. J. & Pichler, R. H. 9. Miner, J. H. The glomerular basement
857–861 (2005). Proteinuria in diabetic kidney disease: membrane. Exp. Cell Res. 318, 973–978 (2012).

NATURE REVIEWS | NEPHROLOGY VOLUME 9  |  AUGUST 2013  |  475


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

10. St John, P. L. & Abrahamson, D. R. Glomerular 30. Chen, Y. M. & Miner, J. H. Glomerular basement 49. Park, J. E., Keller, G. A. & Ferrara, N. The vascular
endothelial cells and podocytes jointly membrane and related glomerular disease. endothelial growth factor (VEGF) isoforms:
synthesize laminin‑1 and ‑11 chains. Kidney Int. Transl. Res. 160, 291–297 (2012). differential deposition into the subepithelial
60, 1037–1046 (2001). 31. Colognato, H., Winkelmann, D. A. & extracellular matrix and bioactivity of
11. Eremina, V. et al. Glomerular-specific alterations Yurchenco, P. D. Laminin polymerization induces extracellular matrix-bound VEGF. Mol. Biol. Cell 4,
of VEGF‑A expression lead to distinct congenital a receptor-cytoskeleton network. J. Cell Biol. 1317–1326 (1993).
and acquired renal diseases. J. Clin. Invest. 111, 145, 619–631 (1999). 50. Goldberg, S., Harvey, S. J., Cunningham, J.,
707–716 (2003). 32. Poschl, E. et al. Collagen IV is essential for Tryggvason, K. & Miner, J. H. Glomerular filtration
12. Farquhar, M. G. Editorial: The primary basement membrane stability but dispensable is normal in the absence of both agrin and
glomerular filtration barrier—basement for initiation of its assembly during early perlecan-heparan sulfate from the glomerular
membrane or epithelial slits? Kidney Int. 8, development. Development 131, 1619–1628 basement membrane. Nephrol. Dial. Transplant.
197–211 (1975). (2004). 24, 2044–2051 (2009).
13. Farquhar, M. G. & Palade, G. E. Glomerular 33. Smyth, N. et al. Absence of basement 51. Morita, H. et al. Heparan sulfate of perlecan is
permeability. II. Ferritin transfer across the membranes after targeting the LAMC1 gene involved in glomerular filtration. J. Am. Soc.
glomerular capillary wall in nephrotic rats. J. Exp. results in embryonic lethality due to failure of Nephrol. 16, 1703–1710 (2005).
Med. 114, 699–716 (1961). endoderm differentiation. J. Cell Biol. 144, 52. Pierson, M., Cordier, J., Hervouuet, F. & Rauber, G.
14. Farquhar, M. G., Wissig, S. L. & Palade, G. E. 151–160 (1999). An unusual congenital and familial congenital
Glomerular permeability. I. Ferritin transfer 34. Vanacore, R. et al. A sulfilimine bond identified in malformative combination involving the eye and
across the normal glomerular capillary wall. collagen IV. Science 325, 1230–1234 (2009). kidney. J. Genet. Hum. 12, 184–213 (1963).
J. Exp. Med. 113, 47–66 (1961). 35. Hudson, B. G. The molecular basis of 53. Zenker, M. et al. Human laminin beta2 deficiency
15. Brenner, B. M., Hostetter, T. H. & Humes, H. D. Goodpasture and Alport syndromes: beacons for causes congenital nephrosis with mesangial
Molecular basis of proteinuria of glomerular the discovery of the collagen IV family. J. Am. sclerosis and distinct eye abnormalities. Hum.
origin. N. Engl. J. Med. 298, 826–833 (1978). Soc. Nephrol. 15, 2514–2527 (2004). Mol. Genet. 13, 2625–2632 (2004).
16. Miner, J. H. & Yurchenco, P. D. Laminin functions 36. Miner, J. H. Developmental biology of glomerular 54. Zenker, M., Pierson, M., Jonveaux, P. & Reis, A.
in tissue morphogenesis. Annu. Rev. Cell. Dev. basement membrane components. Curr. Opin. Demonstration of two novel LAMB2 mutations in
Biol. 20, 255–284 (2004). Nephrol. Hypertens. 7, 13–19 (1998). the original Pierson syndrome family reported
17. Paulsson, M. Basement membrane proteins: 37. Gunwar, S. et al. Glomerular basement 42 years ago. Am. J. Med. Genet. A 138, 73–74
structure, assembly, and cellular interactions. membrane. Identification of a novel (2005).
Crit. Rev. Biochem. Molec. Biol. 27, 93–127 disulfide‑cross‑linked network of alpha3, alpha4, 55. Lehnhardt, A. et al. Pierson syndrome in an
(1992). and alpha5 chains of type IV collagen and its adolescent girl with nephrotic range proteinuria
18. Ekblom, P. & Timpl, R. Cell‑to‑cell contact and implications for the pathogenesis of Alport but a normal GFR. Pediatr. Nephrol. 27, 865–868
extracellular matrix. A multifaceted approach syndrome. J. Biol. Chem. 273, 8767–8775 (2012).
emerging. Curr. Opin. Cell Biol. 8, 599–601 (1998). 56. Kagan, M., Cohen, A. H., Matejas, V., Vlangos, C.
(1996). 38. Kohfeldt, E., Sasaki, T., Gohring, W. & Timpl, R. & Zenker, M. A milder variant of Pierson
19. Yurchenco, P. D. & Cheng, Y. S. Self-assembly Nidogen‑2: a new basement membrane protein syndrome. Pediatr. Nephrol. 23, 323–327
and calcium-binding sites in laminin. A three-arm with diverse binding properties. J. Mol. Biol. 282, (2008).
interaction model. J. Biol. Chem. 268, 99–109 (1998). 57. Hasselbacher, K. et al. Recessive missense
17286–17299 (1993). 39. Timpl, R. Structure and biological activity of mutations in LAMB2 expand the clinical
20. Cheng, Y. S., Champliaud, M. F., Burgeson, R. E., basement membrane proteins. Eur. J. Biochem. spectrum of LAMB2-associated disorders.
Marinkovich, M. P. & Yurchenco, P. D. Self- 180, 487–502 (1989). Kidney Int. 70, 1008–1012 (2006).
assembly of laminin isoforms. J. Biol. Chem. 40. Miosge, N., Sasaki, T. & Timpl, R. Evidence of 58. Chen, Y. M., Kikkawa, Y. & Miner, J. H.
272, 31525–31532 (1997). nidogen‑2 compensation for nidogen‑1 A missense LAMB2 mutation causes congenital
21. Timpl, R. et al. Structure and function of laminin deficiency in transgenic mice. Matrix Biol. 21, nephrotic syndrome by impairing laminin
LG modules. Matrix Biol. 19, 309–317 (2000). 611–621 (2002). secretion. J. Am. Soc. Nephrol. 22, 849–858
22. Colognato, H. & Yurchenco, P. D. Form and 41. Miosge, N. et al. Ultrastructural colocalization of (2011).
function: the laminin family of heterotrimers. nidogen‑1 and nidogen‑2 with laminin‑1 in 59. Purvis, A. & Hohenester, E. Laminin network
Dev. Dyn. 218, 213–234 (2000). murine kidney basement membranes. formation studied by reconstitution of ternary
23. Henry, M. D. & Campbell, K. P. Dystroglycan Histochem. Cell Biol. 113, 115–124 (2000). nodes in solution. J. Biol. Chem. 287,
inside and out. Curr. Opin. Cell Biol. 11, 602–607 42. Schymeinsky, J. et al. Gene structure and 44270–44277 (2012).
(1999). functional analysis of the mouse nidogen‑2 60. Noakes, P. G. et al. The renal glomerulus of mice
24. Hynes, R. O. Integrins: bidirectional, allosteric gene: nidogen‑2 is not essential for basement lacking s‑laminin/laminin beta 2: nephrosis
signaling machines. Cell 110, 673–687 (2002). membrane formation in mice. Mol. Cell. Biol. 22, despite molecular compensation by laminin
25. Kreidberg, J. A. et al. Alpha 3 beta 1 integrin has 6820–6830 (2002). beta 1. Nat. Genet. 10, 400–406 (1995).
a crucial role in kidney and lung organogenesis. 43. Murshed, M. et al. The absence of nidogen 1 61. Noakes, P. G., Gautam, M., Mudd, J., Sanes, J. R.
Development 122, 3537–3547 (1996). does not affect murine basement membrane & Merlie, J. P. Aberrant differentiation of
26. Kikkawa, Y., Sanzen, N. & Sekiguchi, K. Isolation formation. Mol. Cell. Biol. 20, 7007–7012 (2000). neuromuscular junctions in mice lacking
and characterization of laminin‑10/11 secreted 44. Bader, B. L. et al. Compound genetic ablation of s‑laminin/laminin beta 2. Nature 374, 258–262
by human lung carcinoma cells. laminin‑10/11 nidogen 1 and 2 causes basement membrane (1995).
mediates cell adhesion through integrin alpha3 defects and perinatal lethality in mice. Mol. Cell. 62. Jarad, G., Cunningham, J., Shaw, A. S. &
beta1. J. Biol. Chem. 273, 15854–15859 Biol. 25, 6846–6856 (2005). Miner, J. H. Proteinuria precedes podocyte
(1998). 45. Groffen, A. J. et al. Agrin is a major heparan abnormalities in Lamb2‑/- mice, implicating the
27. Kikkawa, Y., Virtanen, I. & Miner, J. H. Mesangial sulfate proteoglycan in the human glomerular glomerular basement membrane as an albumin
cells organize the glomerular capillaries by basement membrane. J. Histochem. Cytochem. barrier. J. Clin. Invest. 116, 2272–2279 (2006).
adhering to the G domain of laminin alpha5 in 46, 19–27 (1998). 63. Ryan, G. B. & Karnovsky, M. J. Distribution of
the glomerular basement membrane. J. Cell Biol. 46. Harvey, S. J. et al. Disruption of glomerular endogenous albumin in the rat glomerulus: role
161, 187–196 (2003). basement membrane charge through podocyte- of hemodynamic factors in glomerular barrier
28. Wizemann, H. et al. Distinct requirements for specific mutation of agrin does not alter function. Kidney Int. 9, 36–45 (1976).
heparin and alpha-dystroglycan binding revealed glomerular permselectivity. Am. J. Pathol. 171, 64. Wartiovaara, J. et al. Nephrin strands contribute
by structure-based mutagenesis of the laminin 139–152 (2007). to a porous slit diaphragm scaffold as revealed
alpha2 LG4-LG5 domain pair. J. Mol. Biol. 332, 47. Rennke, H. G., Cotran, R. S. & by electron tomography. J. Clin. Invest. 114,
635–642 (2003). Venkatachalam, M. A. Role of molecular charge 1475–1483 (2004).
29. Jarad, G., Pippin, J. W., Shankland, S. J., in glomerular permeability. Tracer studies with 65. Noone, D. & Licht, C. An update on the
Kreidberg, J. A. & Miner, J. H. Dystroglycan does cationized ferritins. J. Cell Biol. 67, 638–646 pathomechanisms and future therapies of Alport
not contribute significantly to kidney (1975). syndrome. Pediatr. Nephrol. 28, 1025–1036
development or function, in health or after injury. 48. Bezakova, G. & Ruegg, M. A. New insights into (2013).
Am. J. Physiol. Renal Physiol. 300, F811–F820 the roles of agrin. Nat. Rev. Mol. Cell Biol. 4, 66. Abrahamson, D. R., Hudson, B. G.,
(2011). 295–308 (2003). Stroganova, L., Borza, D. B. & St John, P. L.

476  |  AUGUST 2013  |  VOLUME 9  www.nature.com/nrneph


© 2013 Macmillan Publishers Limited. All rights reserved
REVIEWS

Cellular origins of type IV collagen networks in Alport glomerular disease. Kidney Int. 76, 76. Brinkkoetter, P. T., Ising, C. & Benzing, T. The role
developing glomeruli. J. Am. Soc. Nephrol. 20, 968–976 (2009). of the podocyte in albumin filtration. Nat. Rev.
1471–1479 (2009). 72. Smithies, O. Why the kidney glomerulus does not Nephrol. http://dx.doi.org/10.1038/
67. Abrahamson, D. R. et al. Laminin compensation clog: a gel permeation/diffusion hypothesis of nrneph.2013.78.
in collagen alpha3(IV) knockout (Alport) renal function. Proc. Natl Acad. Sci. USA 100, 77. Faul, C., Asanuma, K., Yanagida-Asanuma, E.,
glomeruli contributes to permeability defects. 4108–4113 (2003). Kim, K. & Mundel, P. Actin up: regulation of
J. Am. Soc. Nephrol. 18, 2465–2472 (2007). 73. Suh, J. H., Jarad, G., Vandevoorde, R. G. & podocyte structure and function by components
68. Gross, O. et al. Preemptive ramipril therapy Miner, J. H. Forced expression of laminin beta1 of the actin cytoskeleton. Trends Cell Biol. 17,
delays renal failure and reduces renal fibrosis in in podocytes prevents nephrotic syndrome in 428–437 (2007).
COL4A3-knockout mice with Alport syndrome. mice lacking laminin beta2, a model for Pierson
Kidney Int. 63, 438–446 (2003). syndrome. Proc. Natl Acad. Sci. USA 108, Acknowledgements
69. Gross, O. et al. Early angiotensin-converting 15348–15353 (2011). The authors are supported by NIH grants
enzyme inhibition in Alport syndrome delays 74. Chen, Y. M. et al. Laminin β2 gene missense R01DK078314, R21DK095419, and P30DK079333
renal failure and improves life expectancy. mutation produces endoplasmic reticulum and by a grant from the Alport Syndrome Foundation.
Kidney Int. 81, 494–501 (2012). stress in podocytes. J. Am. Soc. Nephrol. http:// J. H. Suh is also supported by NIH training grant
70. Wyss, H. M. et al. Biophysical properties of dx.doi.org/10.1681/ASN.2012121149. T32DK007126.
normal and diseased renal glomeruli. Am. 75. Kikkawa, Y. & Miner, J. H. Molecular dissection of
J. Physiol. Cell Physiol. 300, C397–C405 (2011). laminin alpha 5 in vivo reveals separable domain- Author contributions
71. Meehan, D. T. et al. Biomechanical strain causes specific roles in embryonic development and The authors contributed equally to all aspects of this
maladaptive gene regulation, contributing to kidney function. Dev. Biol. 296, 265–277 (2006). manuscript.

NATURE REVIEWS | NEPHROLOGY VOLUME 9  |  AUGUST 2013  |  477


© 2013 Macmillan Publishers Limited. All rights reserved

Vous aimerez peut-être aussi