You are on page 1of 11

Review

The nature and biology of basement


membranes

Ambra Pozzi a, d , Peter D. Yurchenco b and Renato V. Iozzo c


a - Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University, Nashville, TN, United States
b - Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, United
States
c - Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Sidney Kimmel Cancer
Center, Thomas Jefferson University, Philadelphia, PA, United States
d - Veterans Affairs Hospitals, Nashville, TN, United States

Correspondence to Ambra Pozzi: at: Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt
University, Nashville, TN, United States ambra.pozzi@vanderbilt.edu
http://dx.doi.org/10.1016/j.matbio.2016.12.009

Abstract
Basement membranes are delicate, nanoscale and pliable sheets of extracellular matrices that often act as
linings or partitions in organisms. Previously considered as passive scaffolds segregating polarized cells,
such as epithelial or endothelial cells, from the underlying mesenchyme, basement membranes have now
reached the center stage of biology. They play a multitude of roles from blood filtration to muscle homeostasis,
from storing growth factors and cytokines to controlling angiogenesis and tumor growth, from maintaining skin
integrity and neuromuscular structure to affecting adipogenesis and fibrosis. Here, we will address
developmental, structural and biochemical aspects of basement membranes and discuss some of the
pathogenetic mechanisms causing diseases linked to abnormal basement membranes.
© 2017 Elsevier B.V. All rights reserved.

Introduction where the physiological and pathological functions


of BM components are critically assessed.
Basement membranes (BMs) are cell-adherent
extracellular matrices widely distributed in metazoan
tissues. First identified in skeletal muscle 176 years How many proteins are incorporated
ago [1], elucidation of BM constituents, structure, into a typical basement membrane?
functions and genetics has required advances in
multiple fields stretched over many years. The The core structural components of BMs are
beginning of a molecular understanding of BMs laminins, collagen IV, nidogens, and the heparan
dates to the 1970s and 1980s when Kefalides sulfate proteoglycans (HSPGs) perlecan and agrin
discovered collagen IV and Kuhn, Timpl, Martin, (Fig. 1). We envision core components as those
Bruckner, Robey, Rhode and others exploited the macromolecules for which there is an embryonic
Engelbreth-Holm-Swarm tumor as a source for phenotype of failed or structurally-defective BM
obtaining soluble BM components for analysis. assembly upon knockout with the provision
Since then, a combination of biochemical, biophys- that compensation can sometimes mask a
ical, cell biological, genetic, bioengineering and structure-forming role. These glycoproteins and
other approaches led to our current understanding proteoglycans, initially secreted in a soluble state,
of BMs. We are pleased to present a special edition become organized into insoluble cell scaffoldings
of Matrix Biology entitled “Basement Membranes in and constitute a complex meshwork of proteins
Health and Disease” containing a collection of present in multicellular organisms [2]. In addition,
twenty-six articles from specialists in the field BMs contain “matricellular proteins” that provide

0022-2836/© 2017 Elsevier B.V. All rights reserved. Matrix Biol. (2017) 57–58, 1–11
2 Basement membrane biology

Fig. 1. Core basement membrane components and binding interactions. Laminins (Lms) are central organizers of BMs.
Lm111, a prototypical laminin expressed in embryogenesis, binds to cell surface sulfated glycolipids (SGL), several
integrins, α-dystroglycan (αDG), nidogens (Nd), agrin, and polymerizes via its LN domains. Collagen-IV (three isoforms)
and perlecan bind to nidogen, completing the core basement membrane scaffold. Agrin and perlecan HS chains attach to
growth factors, promoting their interactions with receptor tyrosine kinases (RTK). Integrin and αDG attach through adaptor
proteins to the cytoskeleton. Lm411, an isoform that does not polymerize, exhibits weak integrin and αDG binding but
strong binding to SGLs (gal sulfatide). Lm511/521, polymerizing laminins, binds to multiple integrins both in the LG
domains and α5 short arm, to the Lutheran receptor (Lu), and moderately well to αDG. Lm3A32, a non-polymerizing
laminin found in epithelia, binds strongly to α6β4 integrin of hemidesmosomes and links to collagen VII of the anchoring
fibrils.

additional, often tissue-specific, functions but are not Evolution and embryogenesis of base-
essential for BM assembly or architecture [3].
Examples are SPARC and nephronectin. Various ment membranes
growth factors/morphogens, many belonging to the Laminin domains integrated within proteins such
TGFβ superfamily, are found tethered to BMs (in as cadherins exist in single-cell motile choanofla-
particular to proteoglycan HS chains) and act to gellates representing primitive species that pre-date
provide specific signals to BM-adherent cells. the evolutionary emergence of BMs [4]. These
Additionally, collagen XV and XVIII are found at the unicellular organisms can aggregate to form clus-
stromal interface. Finally, proteinases and their ters, suggesting BM/cadherin precursors serve
inhibitors, regulatory macromolecules and serum cell-cell adhesive functions. BMs are thought to
factors are found associated with BMs. Collectively, have first emerged in metazoans as a requirement
these components, organized into or associated with for organizing epithelial tissues. Laminin α-like, β-like
BMs, provide cell and tissue support and act as and γ-like subunits, each with a laminin N-terminal
complex signaling platforms. (LN) and coiled-coil domains, and collagen IV-like
Basement membrane biology 3

subunits have been identified in the sponges A given BM contains at least one laminin, one type
Amphimedon and Chondrosia [5]. of collagen IV heterotrimer, nidogen, and HSPGs.
It is said that ontogeny recapitulates phylogeny. Proteomic analyses of the internal limiting membrane,
BMs first appear in mouse embryos at embryonic (E) a thick BM adjacent to the retina that could be cleanly
day 4–4.5 as the primitive endoderm differentiates isolated, reported considerable complexity with sixty
into an epithelial layer from the inner cell mass and one components identified [11]. These included
secretes two laminins, collagen IV, nidogen-1 and different laminins, collagens, nidogens, proteogly-
perlecan [6,7] that assemble into a BM. The BM, in cans, growth factors, proteases, and other secreted
turn, induces conversion of the inner cell mass into factors. Using a proteomic approach to compare
the epiblast, a polarized epithelium. Inactivation of different organs, considerable variability of tissue
either the common LAMC1 or LAMB1 genes in mice distribution of known BM proteins has been found [11].
results in a failure of all BM assembly with
peri-implantation lethality by E5.5 [8]. By this stage
and thereafter, laminin expression and BM assembly Basement membrane assembly
are critical for morphogenesis extending through Basement membrane formation is largely one of
gastrulation and organogenesis. Although collagen self-assembly involving cell surface adhesion, inter-
IV is present in the earliest BMs in mammals, an component binding and polymerization. Laminins, by
absolute requirement for collagen IV does not occur binding to cells, self, and other BM components, are
until later during vasculogenesis (E10.5–11.5), likely essential for initiating this process. Laminin assembly is
when BMs capable of resisting mechanical stresses receptor-facilitated, occurs on “competent” cell sur-
are needed. Nidogens and perlecan are both faces that express laminin-binding molecules and is
essential components before or around birth, but accompanied by polymerization mediated by the LN
not during the earliest stages of differentiation. Loss domains [12]. The BM, in good part through laminins, is
of these components affects morphogenesis at anchored to integrin and dystroglycan receptors,
various stages [7,9]. connecting the nascent BM to the underlying cytoskel-
Laminins are a fairly large family of heterotrimers eton. This anchorage is crucial for activation of
each consisting of an α-, a β- and a γ-subunit joined signaling cascades in the case of integrins in all tissues
together through a long coiled-coil domain. There and likely for dystroglycan as well in select tissues.
are five α, four β and three γ chains that have been Other cell surface components (e.g. sulfatides) can
identified in mammals. Not all αβγ combinations are promote laminin adhesion without cytoskeletal anchor-
allowed and only 16 different mammalian laminin age and have been found to play an important role in
isoforms have been shown to assemble from the Schwann cell BM assembly. Specialized receptors
possible combinations. Common to most laminins is such as the Lutheran protein uniquely interact with
the γ1-subunit, first expressed in the peri-implantation Lm511, while receptor tyrosine kinases bind to growth
period and essential for both early and late morpho- factors/morphogens tethered to the HS chains of
genesis [8]. Laminins are either a cross-shaped HSPGs to affect cell functions. The laminin scaffold
(three short arms and one long coiled-coil arm; e.g. enables recruitment and assembly of the remaining
Lm111, Lm211, Lm511), Y-shaped (due to absence components. Nidogen-1 and the less common nido-
of the α-short arm; e.g., Lm411), or rod-shaped gen-2 bind to the laminin through the γ-subunit short
(with truncated short arms; Lm3A32). The major arm [12], acting as bridge to the collagen IV network. Of
receptor-binding domains are the C-terminal note, deletion of epidermal γ-subunit of laminin leads to
laminin-type globular (LG) domains located at the defects in hair morphogenesis [13], whereas laminin α5
C-terminal moiety of the α-subunit. These can interact subunit in keratinocyte BM is required epidermal/
with laminin-class integrins (α3β1, α6β1, α7β1, α6β4), dermal communication [14]. Perlecan binds to nidogen,
α-dystroglycan, sulfated glycolipids, heparin, and, in αDG and collagen IV, and the last through the HS
the case of Lm511, the Lutheran receptor. The chains. Collagen IV self-assembles into a network-like
polymerization loci are located in the N-terminal LN polymer through N-terminal (7S), lateral, and C-termi-
domains. Nidogens and agrin bind to internal nal (NC1) domain interactions that become covalently
domains within the Lmγ subunit while perlecan and stabilized through the 7S and NC1 domains. In
collagen IV bind to nidogen [9]. epithelia, Lm3A32 plays a key role in forming strong
Collagen IV is composed of 6 α chains named α1-α6 attachments between integrin α6β4 and collagen VII of
that assemble in order to form heterotrimers. Three anchoring fibrils at the stromal-BM interface [12].
major type IV collagens, each a heterotrimer, are
present in different BMs [10]. The most ubiquitous
trimer consists of two α1 and one α2 chains and is Mechano/chemical signaling mechanisms
present in nearly all BMs. The α3α4α5 and α5α5α6
heterotrimers, on the other hand, have restricted Both mechanical and chemical interactions of
distributions; notably in the renal glomeruli, neuromus- BM and integrins and other receptors are required
cular junctions, eyes, inner ears and other locations. for cell signaling [15]. A breakthrough in our
4 Basement membrane biology

understanding of the mechanical role came from a through BM to receptor to cytoskeleton has been
study showing that matrix stiffness is capable of broken.
driving cell differentiation through different pathways Mutations of the LAMB2 gene cause Pierson
[16]. This contribution has largely been explored with syndrome. Pierson syndrome is a rare form of focal
synthetic gels whose viscoelastic properties can segmental glomerulosclerosis in which patients
be manipulated and that are attached to integrin develop proteinuria, eye abnormalities that include
ligands at known density. An emerging concept microcoria, and sometimes defects of the neuro-
is that integrin-ligated ECMs such as BMs exert muscular junction [20] (Table 1). Milder forms of the
their mechanical effect by “pulling” integrins disease are seen with in-frame point mutations
against resistance created by the viscoelastic affecting the LN domain. The proteinuria occurs
nature of the ECM polymer and by the cell before onset of foot-process effacement, evidence
glycocalyx that is compressed between the ECM that the glomerular BM contributes to perm-selective
ligands and short integrin ectodomains [51]. An filtration in the kidney.
intracellular talin-based molecular clutch mediates A major genetic defect in collagen IV is the cause
mechano-transmission of the signal to the actin of Alport syndrome. Most commonly, the disease is
cytoskeleton that is modulated by the motor activity X-linked resulting from mutations of the COL4A5
of non-muscle myosin-II. gene. However, autosomal transmitted forms are
seen as well resulting from mutations of COL4A4
and COL4A3 (Table 1). The disease is characterized
Human genetic diseases linked to base- by the nephritic syndrome (hematuria + proteinuria)
ment membrane constituents: can we and associated hearing deficits. The renal glomer-
survive without basement membranes? ular BM has a basket-weave appearance with focal
attenuations at the ultrastructural level. At the light
Defects of BM components such as the Lmγ1 histological level, glomerulonephritis with crescent
chain, essential for embryogenesis, are presumed to formation is present.
result in embryonic death well before diseases might Glycine substitutions in COLA41, resulting in
become manifest. Non-embryonic-lethal mutations reduced secretion, can cause porencephaly (degen-
in BM component genes are found for other laminin erative brain cavities in newborn infants) and
subunits, collagen IV, and perlecan. Several of these cerebral hemorrhage [22]. Humans with COLA41
diseases are briefly discussed below and summa- mutations were more recently found to develop small
rized in Table 1. vessel disease and hemorrhagic stroke [23]. It was
One set of laminin disorders are the Lmα2/ proposed that mutation of COL4A1 might cause a
merosin-deficient congenital muscular dystrophies spectrum of cerebrovascular phenotypes in which
(MDC1A and limb-girdle type) arising from mutations COL4A1 mutations may be predisposed to
in the LAMA2 gene. The diseases primarily manifest hemorrhage.
in skeletal muscle but also presents in peripheral Notably, Gly missense mutations in COL6A1 gene
nerves and brain [17]. The dystrophies resulting from causes two forms of congenital myopathy (Table 1):
a total or near-total absence of α2-laminins are very Ulrich congenital muscular dystrophy with severe
severe and associated with failure to ambulate and hypotonia starting in early childhood, and Bethlehem
early, often respiratory death. A milder limb-girdle myopathy, a milder form with joint laxity and
form can be seen with a number of single amino acid hypotonia [27]. In Table 1, we have listed also
substitutions or short deletions affecting the LN or genetic diseases of COL17A1, although not a strict
other short arm domains [18]. Muscular dystrophies constituent of BMs. Unlike most collagens, collagen
also arise from mutations affecting laminin receptors XVII is a transmembrane protein and a structural
(DG, integrin α7β1), the DG-binding protein dystro- component of hemidesmosomes. Missense and
phin, and the stromal interface protein collagen VI. nonsense mutations in this gene are associated
These components are connected through linkage. with epithelial recurrent erosion syndrome [30,31]
Another set of laminin disorders is seen in Herlitz and junctional epidermolysis bullosa [32], both
junctional epidermolysis bullosa, a lethal blistering congenital diseases that affect BM integrity.
disease resulting from splitting of the epidermal BM HSPGs include collagen XVIII, perlecan and agrin.
and arising from mutations of the LAMA3, LAMB3 or Genetic deficiencies linked to COL18A1 gene are
LAMC2 [19]. Skin blistering diseases also include generally due to reduced production or changes in
those resulting from ruptures at the level or keratin the C-terminal region of the chain (Table 1).
(EB simplex, keratin gene mutations) and anchoring Knobloch syndrome is characterized by ocular
fibrils (EB dystrophica, collagen VII) [28,29]. Notably, defects associated with high myopia, vitreoretinal
collagen VII is elevated in patients with systemic degeneration and occipital encephalocele [33–35].
sclerosis [43]. Both MDC1A and Herlitz disease can Changes in the C-terminal portion of the chain result
be viewed as primarily resulting from a loss of BM in duplication of the renal collecting system [36].
integrity in which a transverse link from stroma Thus, fine alterations in collagen XVIII structure and
Basement membrane biology
Table 1. Human genetic diseases associated with mutations in basement membrane constituents.
Gene Mutation type Mutation effects Disease References
LAMA2 Substitutions, deletions, duplications, a) Absent/trace Lmα2 expression – functionally a) Absent/trace Lmα2 (most cases): severe [17]
insertions, and inversions resulting in null.b) Normal/reduced Lmα2 expression seen non-ambulatory congenital muscular dystrophy
protein substitutions, deletions, frame in single amino acid substitutions and short with early demise.b) Normal/reduced Lmα2
shifts, nonsense, and complex variant in-frame deletions in LN and other short arm expression: ambulatory (similar to limb-girdle
Lmα2 chains. domains type) muscular dystrophy
LAMA3LAMB3LAMC2 Frame shift, nonsense and missense a) Premature chain termination.b) Missense Junctional epidermolysis bullosa (JEB):a) Herlitz [19]
mutations mutation or putative splicing mutation in trans JEB – severeb) Non-Herlitz JEB – milder
phenotype
LAMB2 Nonsense, missense, spice site mutations, a) Truncated and functionally null Lmβ2.b) Pierson syndrome. Congenital nephrotic [20]
multiple sites. Missense mutations Substitutions: normal/reduced expression of syndrome with eye abnormalities, microcoria.
clustered in LN domain. Lmβ2 Muscular weakness/myasthenia may be present
COL4A1 Frameshift Disruption of the C-terminal NC1 domain Hematuria, chronic kidney disease, renal cysts [21]
COL4A1 G1236RG749S Amino acid change in the triple-helical domain Porencephaly, hemorrhagic strokes [22]
COL4A1 G1769A Amino acid change in the triple-helical domain Retinal hemorrhage, retinopathy [23,24]
COLA4A3COL4A4COL4A5 Splicing variants, missense mutations Altered chain expression Alport syndrome, deafness, lenticonus, cataract, [25,26]
maculopathy
COL6A1 Gly missense mutations Amino acid change in the triple helix Ulrich congenital muscular dystrophy: severe [27]
hypotonia, early childhoodBethlehem myopathy:
joint laxity and hypotonia
COL7A1 Amino acid mutations Loss of α1 chain expression Dystrophic epidermolysis bullosa [28,29]
COL17A1COL17A1 Missense mutationsNonsense mutation Exon truncationPremature stop codons Epithelial recurrent erosion dystrophyJunctional [30,31,32]
epidermolysis bullosa
COL18A1COL18A1 F r a m e - s h i f t m u t a t i o n s N o n s e n s e Al ter ed cha i n r edu cti on Lo ss o f cha i n Knobloch syndrome: high myopia, vitreoretinal [33–35,36]
mutationsMissense mutationsInsertion, productionChange in the C-terminal chain degeneration, occipital encephalocele, fasting
frame deletions Splice site mutation triglyceridemiaDuplication of kidney collecting
system
HSPG2 Duplicated exon 34 and frameshifts Functionally null Dyssegmental dysplasia, Silver-Handmaker type [37–39]
due to altered FGF2 signaling leading to aberrant
cartilage architecture
HSPG2 Missense and errors in alternative splicing Truncated or ablated Domain V (endorepellin) Schwartz-Jampel syndrome originating from [38,40]
disrupted neuromuscular junction arrangement
and function
AGRN Missense and nonsense mutations at the Failure of neuromuscular transmission, pre- and Congenital myasthenic syndrome, easy [41,42]
N-terminus and LG2 domain post-synaptic fatigability and muscle weakness

5
6 Basement membrane biology

protein levels can lead to aberrant and diverse Neural and non-neural (muscle) agrins, derived
phenotypes. from a single alternatively spliced gene, are BM
Perlecan is one of the few gene products HSPGs with an N-terminal laminin-binding NtA
expressed by both vascular and non-vascular tissue domain, intervening follistatin, S/T and SEA do-
[44], and can act as a strong elastic tether [45] as mains, and a C-terminal complex of 3 LG domains
well as a modulator of cell adhesion, proliferation spaced by EGF-like modules [78]. The neural splice
and growth factor signaling [46]. For example, variant (A8B8), secreted by peripheral nerve termini,
perlecan is found as an HSPG in BMs of most if is critical for recruitment of post-synaptic acetylcho-
not all the vascularized organs, but is also expressed line receptors at the neuromuscular junction, where-
as a hybrid HS/chondroitin sulfate proteoglycan in as non-neural (“muscle”) agrin that lacks the A/B
cartilage [47,48]. Perlecan has a multi-domain sequence inserts is widely expressed. The normal
protein core, including critical C-terminal LG do- functions of non-neural agrin are less well under-
mains with α-dystroglycan and integrin-binding stood. Agrin LG domains bind strongly to αDystro-
activities, important for developmental and tumor glycan and weakly to integrin α3β1 [79,80]. Notably,
angiogenesis [49–53]. Indeed, the C-terminal do- an internally shortened non-neural agrin (“minia-
main of perlecan, termed endorepellin, is processed grin”), that consists of the laminin-binding NtA
by various proteases involved in multiple biological domain and the C-terminal three LG domains,
functions, and is anti-angiogenic [54–60] and can be partially rescues the dyW dystrophic phenotype
pro-autophagic [61,62]. In contrast, the parent [81]. Mechanistically, this rescue could be due to
molecule perlecan can exert anti-autophagic func- enabling the compensating Lm411 to now bind αDG.
tions [63,64]. This miniagrin enables Lm111 lacking any LG
Mutations in gene encoding perlecan (HSPG2) domains to assemble a BM on Schwann cells [82].
cause dyssegmental dysplasia Silver-Handmaker This has led to the proposal that non-neural agrin
(DDSH) type. The pathogenetic mechanism of acts as a “collateral linker” by binding to the
DDSH lies on genetic duplication of exon 34 and coiled-coil of laminins and to αLG [12].
frameshifts. This leads to a functionally null protein Agrin LG3 domain has been found, together with the
causing altered FGF2 signaling and subsequent LG3 domain of perlecan, in the urine [83], and is also
aberrant cartilage architecture [37–39]. Mutations in released by neurotrypsin at the synapse [84]. Perlecan
HSPG2 also cause Schwartz-Jampel syndrome LG3 domain has been found in many biological fluids
(SJS), also known as chondrodystrophic myotonia, including blood, urine and cerebrovascular fluids
a disorder characterized by myotonia and skeletal [83,85–87]. Interestingly, LG3 could represent a
chondrodysplasia [40,65]. The myotonia in SJS biomarker for breast cancer detection as low levels of
patients results from loss of the role of perlecan LG3 correlate with tumor progression and invasion [88].
AChE-binding HS chains at the neuromuscular
junction while the skeletal dysplasia results from
loss of the CS-E chains that regulate collagen II Can we answer some of the key ques-
assembly in cartilage [65]. tions in basement membrane biology?
Mutations in the gene encoding agrin (AGRN)
cause congenital myasthenic syndrome [41,42]. So far we have dwelled on demonstrating how
Pathogenetically, missense and nonsense mutations BMs are important for the maintenance of tissue
at the N-terminus and LG2 domain lead to pre- and homeostasis and how they function in various
post-synaptic failure of neuromuscular transmission. diseases processes. In this special issue a number
Consequently, these infants exhibit muscle weakness of leading investigators of various aspects of BM
(myasthenia) that worsens during physical exertion. biology will attempt to answer some biologically-r-
There is central and recurrent theme in BM elevant questions. Compositionally, BMs are tissue
biology, that is, the ubiquitous processing of BMs and organ specific, as clearly shown by proteomic
constituents by various proteases, especially those analysis of BMs isolated from various tissues
belonging to matrix metalloproteinases and bone (Randles MJ et al.). The demonstration that BMs
morphogenetic proteins [66–72]. This limited proteo- are important for the maintenance of tissue homeo-
lytic process generates fragments full of biological stasis comes from the finding that mutations in BM
activity. Examples are endorepellin [73] and endo- genes lead to complex and severe phenotypes. To
statin, derived from partial processing of the C-ter- this end, mutations in the COL4A1 and COL4A2
mini of perlecan [74] and collagen XVIII [75], chains have been associated to multisystem disor-
respectively. Also fragments of perlecan domain IV ders with abnormalities in the vasculature, brain,
have been recently described in advanced forms of eyes, kidneys, and muscles. Genotype–phenotype
prostate cancer [76]. Intriguingly, peptides from correlations from more than 100 COL4A1 and
perlecan domain IV support cell adhesion and COL4A2 mutations in patients and more than 15
spreading [77], potentially acting as pro-invasive Col4a1 and Col4a2 mutations in mice show that
factors. triple-helical glycine substitutions are the most
Basement membrane biology 7

frequent classes of mutations. Interestingly, the on collagens and laminins are targeted by auto-an-
position of the mutation rather than the type of the tibodies and lymphocytes in Goodpasture's disease,
substituting amino acid seems to be important in rheumatoid arthritis, post-lung transplant bronchiol-
determining the severity of the disease (Jeanne M itis obliterans syndrome, and multiple autoimmune
and Gould DB). Mutations in any of three collagen IV dermatoses (Foster MH). In the context of tumori-
genes, COL4A3, COL4A4, or COL4A5 lead to Alport genesis, collagen XIX, a minor collagen that belongs
syndrome, a disease characterized by kidney to the FACIT family, plays an important role in
disease, hearing loss, and eye abnormalities (Cos- preventing tumor cell invasion by inhibiting the
grove D and Liu S). In addition to collagen IV, formation of new blood vessels (Oudart JB et al.).
collagen XVIII is another ubiquitously expressed BM In order to invade and metastasize, tumor cells need
component and mutations in COL18A1 gene result to cross the BM that separates them from the
in Knobloch syndrome (Table 1). Studies with vascular bed. BM invasion by malignant cells is
gene-modified mice have elucidated some aspects influenced by crosstalk between three inter-related
of this rare disease and have highlighted the factors: extracellular matrix stiffening, epithelial
importance of collagen XVIII in the development of cytoskeletal contractility, and growth factor/cytokine
the eye and its function in normal situations and signaling (Chang TT et al.). Epithelial contractility
tissue dysregulation (Heliasvaara R et al.). Finally, induces greater stromal stiffening as well as forma-
mutations in collagen VII, a matrix component tion of invadosomes, specialized regions of cells rich
primarily expressed in the BMs of human skin, in proteases. These cell structures sense and
corneal epithelium, and gastrointestinal mucous transduce forces that promote extracellular matrix
membranes, have been associated to Dystrophic degradation by targeting protease expression and
Epidermolysis Bullosa (Uitto et al.). activity at invadosomes (Mrkonjic S et al.).
In addition to collagen, mutations in laminins are The most common defect of BMs is seen in
also associated with disease. Laminin α4, α5, and β2 longstanding diabetes mellitus, causing almost
chains specifically localize to the neuromuscular pathognomonic thickening of microvascular BMs in
junction where they regulate the proper alignment of the retina, peripheral nerve and kidney glomerulus
presynaptic and post-synaptic structures of motor with associated blindness, neuropathy and protein-
neurons and muscle fibers. Mutations of these uria/chronic renal failure. Diabetic nephropathy is the
laminin genes lead to neuromuscular diseases leading cause of chronic kidney disease in the U.S.
including Pierson syndrome and Lambert-Eaton and is often seen in combination with cardiovascular
myasthenic syndrome as summarized in Table 1 disease. The basis for the BM thickening, which
and reviewed by (Rogers RS and Nishimune H). In precedes proteinuria, is not well understood or
vitro studies have revealed that laminin substrata studied, but may involve changes in synthesis,
“instruct” myotubes on where and how to form degradation/remodeling, and/or glycation and that
synapses. Interestingly, culturing myotubes on may be a response to podocyte injury [89].
substrate bound laminin targets the formation of BM components interact with cells via specific
complex acetylcholine receptor aggregates to the receptors, including integrins, discoidin domain
lower cell surface via engagement of the receptor receptors (DDRs), and dystroglycan. These interac-
tyrosine kinase MuSK (Vezina-Audette et al.). tions play a key role in initiating various cell functions
Although for most of these genetic BM linked including extracellular matrix assembly. To this end,
diseases there are limited treatment options, assembly of laminin polymers has been proposed to
cell-based therapeutic approaches based on deliv- be dependent on the three short arms of the
ering pluripotent and/or differentiated cells are cross-shaped laminin heterotrimer [18,82], but is
emerging as a plausible and promising treatment facilitated by the concentrating effect of binding to
for these devastating pathologies (Nyström et al.). cell surface receptors.
In addition to mutations in genes encoding BM Each short arm contains an N-terminal LN
components, changes in the composition of the BMs polymerization domain followed by laminin-type
can also lead to pathological conditions, indicating epidermal growth factor-like (LE) domains inter-
that the type of BM component is a key regulator of spersed with globular domains. In the β1/β2 sub-
tissue function. In this context, while normal large units, an LF domain of unknown function resides in
arteries usually express the laminin β1 chain, in the middle flanked on either side by LE domains. A
atherosclerotic plaques that form in the media of recent high-resolution crystal structure of specific
large arteries, smooth muscle cells switch from β1 to laminin β2 region LE domains interrupted by an LF
β2 chain and produce laminin α5 chain. This results domain has allowed a better understanding on how
in a switch of behavior with smooth muscle cells these domains interact in the short arm thus advising
acquiring a more pronounced proliferative and our understanding of laminin structure (Pulido D et
migratory phenotype (Di Russo J et al.). al.).
BM components are also involved in auto-immune In addition to matrix assembly, cell-matrix interac-
diseases and tumorigenesis. To this end, epitopes tions are important for the regulation of cell
8 Basement membrane biology

proliferation, migration, adhesion, and extracellular dation (Cruz-Acuña R and García AJ). Another
matrix production. Among the matrix receptors, promising source to study BM functions is the use
integrin α3β1 is a major laminin receptor. The of decellularized organs from discarded human
interaction of this receptor with laminins (mainly organs or mice. One advantage of this technique is
Lm332) is essential for the initiation and the that it preserves the three-dimensional structure of
maintenance of tumor cell proliferation, while in ECM matrix scaffolds. In addition, organs from
endothelial cells laminin-integrin α3β1 interaction is healthy and diseased donors can be used thus
associated to decreased endothelial cell proliferation allowing the analysis of cell function, differentiation,
and migration (Ramovs V et al.). Another laminin immune responses in health and disease (Petrosyan
receptor is the integrin α6β4. This receptor is A et al.). Finally, these organs can offer an excellent
localized to hemi-desmosomes and is critical for source for the analysis of their physical and
the establishment of tight adhesions to laminin mechanical properties. These properties are influ-
substrata. Loss of either the α6 or β4 subunit in the enced by many factors, including cell type, develop-
collecting duct of the kidney results in increased mental stage of the organ, degree of disease, type of
susceptibility to injury (Viquez O et al.). Besides BM components, as well as the temporal expression
integrins, DDRs, a class of special receptor tyrosine of ‘BM toolkits’ such as BM binding proteins and/or
kinases that interact with and are activated by enzymes able to modify and crosslink BM compo-
collagens, play a significant role. DDR1, for exam- nents (Jones-Paris CR et al.). The analysis of the
ple, binds both fibrillar and non-fibrillar collagens physical and mechanical properties of BMs in
(e.g. collagen IV) and this interaction triggers isolated organs might enable a better understanding
receptor activation and promotes collagen IV pro- of the physical environment experienced by specific
duction (Borza CM et al.). A third class of BM cell types and to selectively control cell behavior in
receptors are dystroglycans that together with vivo and for tissue engineering (Miller RT).
integrins play a key role in mediating laminin
assembly and polarization of epiblasts in early
embryogenesis (Li S et al.). This study suggests Future research questions and direc-
that integrins and dystroglycans can compensate tions
with each other in promoting laminin assembly.
Finally, key components of BM are HSPGs. Although we are beginning to gain a systematic
Perlecan is a large HSPG expressed by many understanding of BM in biology, many challenges
different cell types, including endothelial cells. The face us going forward. For example, we have a poor
C-terminal domain of perlecan, named endorepellin, understanding of BM turnover, particularly during
plays an anti-angiogenic role by interacting with embryogenesis and following injury. We need to
VEGFR2 and is pro-autophagic by promoting the better understand the reason for having different
formation of LC3-positive autophagosomes inde- isoforms of laminins, nidogens, and collagen IV
pendently of nutrition deprivation (Gubbiotti MA et during and after embryonic development. BM pro-
al.). Another beneficial effect of HSPGs is observed teins all have multiple domains with functions
in the renal glomeruli where they modulate local assigned to a minority of loci. Are the other domains
complement activation by recruiting factor H, which simply spacer domains, or are there many new
in turn selectively inactivates C3b, thereby limiting interactions that still need to be discovered? We are
complement activation. Thus, HS/factor H interac- only beginning to comprehend mechano-chemical
tion might be beneficial in preventing and/or reducing signaling as it applies to BM-cell interactions. How,
immuno-mediated kidney diseases such as mem- for example, is viscoelasticity of the BM controlled?
branous nephropathy and lupus nephritis (Borza Is the principle function of the different integrin
DB). HSPGs also serve as a reservoir for HS-binding specificities one of controlling the ligand affinities
growth factors, including members of the FGF family and collective avidities that in turn affect signaling
and facilitate the interaction between these growth strength? We need to learn if our comprehension of
factors and their receptors. Thus, HSPGs orches- BM assembly mechanisms and resulting structure
trate growth factor availability and activity, two key can be used to intervene in those diseases that
important factors in organogenesis and branching result from BM defects. Biomaterials can be altered
morphogenesis (Patel VN et al.). with BM-derived constituents to alter their cell
The identification of physiological and pathological interactions. How should this be accomplished?
roles of BM components has been made possible BM components can also be used to affect
primarily by the generation of mutant mice lacking differentiation of cells ex vivo for the purpose of
key BM components either globally or in selective their introduction into patients, thereby providing
organs. More recently, synthetic cytocompatible missing or overlooked functions. How can we
three-dimensional hydrogels that carry BM-like collectively accomplish these tasks?
bioactive motifs have been developed and used to In conclusion, this special edition provides an
study specific BM-cell interactions and BM degra- up-to-date and hopefully informative compendium of
Basement membrane biology 9

articles focused on this interesting subject that [4] S.A. Nichols, B.W. Roberts, D.J. Richter, S.R. Fairclough, N.
affects all levels of biology. BMs are active partici- King, Origin of metazoan cadherin diversity and the antiquity
pants in the regulation of cell functions in healthy and of the classical cadherin/beta-catenin complex, Proc. Natl.
diseased organs, a sort of cliché’ statement that is Acad. Sci. U. S. A. 109 (2012) 13046–13051.
[5] B. Fahey, B.M. Degnan, Origin and evolution of laminin gene
true: life without basement membranes is impossi-
family diversity, Mol. Biol. Evol. 29 (2012) 1823–1836.
ble, and life with an abnormal basement membrane [6] P.D. Yurchenco, P.S. Amenta, B.L. Patton, Basement
is quite difficult and full of obstacles and perils. Just membrane assembly, stability and activities observed
thinking of diabetic nephropathy and lupus nephritis. through a developmental lens, Matrix Biol. 22 (2004)
A better understanding of how BMs are assembled, 521–538.
interact with cells, cue signals to cells, and help in [7] J.H. Miner, P.D. Yurchenco, Laminin functions in tissue
the maintenance of organ shape and size might morphogenesis, Annu. Rev. Cell Dev. Biol. 20 (2004)
enable us to better target BM components in 255–284.
disease, and perhaps find new therapeutic strate- [8] N. Smyth, H.S. Vatansever, P. Murray, M. Meyer, C. Frie, M.
gies to combat genetic and acquired BM diseases. Paulsson, et al., Absence of basement membranes after
targeting the LAMC1 gene results in embryonic lethality due
to failure of endoderm differentiation, J. Cell Biol. 144 (1999)
151–160.
[9] P.D. Yurchenco, B.L. Patton, Developmental and pathogenic
mechanisms of basement membrane assembly, Curr.
Acknowledgments Pharm. Des. 15 (2009) 1277–1294.
[10] J. Khoshnoodi, V. Pedchenko, B.G. Hudson, Mammalian
The original research was supported in part by a collagen IV, Microsc. Res. Tech. 71 (2008) 357–370.
Veteran's Affairs Merit Awards 1I01BX002025 (to AP), [11] M.J. Randles, M.J. Humphries, R. Lennon, Proteomic
the National Institutes of Health Grants, R01DK095761 definitions of basement membrane composition in health
(to AP), R01/R37 DK36425 (to PDY), and by the and disease, Matrix Biol. 57-58 (2017) 12–28.
National Institutes of Health grants RO1 CA39481, [12] P.D. Yurchenco, Basement membranes: cell scaffoldings and
RO1 CA47282, and RO1 CA164462 (to RVI). AP is the signaling platforms, Cold Spring Harb. Perspect. Biol. 3 (2011).
recipient of a VA Senior Research Career Scientist. [13] A. Fleger-Weckmann, Y. Ustun, J. Kloepper, R. Paus, W.
Bloch, Z.L. Chen, et al., Deletion of the epidermis derived
laminin gamma1 chain leads to defects in the regulation of
Available online 28 December 2016 late hair morphogenesis, Matrix Biol. 56 (2016) 42–56.
[14] J. Wegner, K. Loser, G. Apsite, R. Nischt, B. Eckes, T. Krieg,
Keywords: et al., Laminin alpha5 in the keratinocyte basement mem-
Collagen; brane is required for epidermal-dermal intercommunication,
Laminin; Matrix Biol. 56 (2016) 24–41.
Heparan sulfate proteoglycan; [15] Z. Sun, S.S. Guo, R. Fassler, Integrin-mediated mechan-
Discoidin domain receptor; otransduction, J. Cell Biol. 215 (2016) 445–456.
Integrin [16] A.J. Engler, S. Sen, H.L. Sweeney, D.E. Discher, Matrix
elasticity directs stem cell lineage specification, Cell 126
(2006) 677–689.
Abbreviations used: [17] C. Jimenez-Mallebrera, S.C. Brown, C.A. Sewry, F. Muntoni,
BM, basement membrane; ECM, extracellular matrix; HS, Congenital muscular dystrophy: molecular and cellular
heparan sulfate; HSPG, heparan sulfate proteoglycan; aspects, Cell. Mol. Life Sci. 62 (2005) 809–823.
MMP, matrix metalloproteinase; MDC1A, Lmα2/merosin- [18] P.D. Yurchenco, Integrating activities of laminins that drive
deficient congenital muscular dystrophy; DDSH, dysseg- basement membrane assembly and function, Curr. Top.
mental dysplasia Silverman-Handmaker type; SJS, Membr. 76 (2015) 1–30.
Schwartz-Jampel syndrome; DDR, discodin domain re- [19] A. Nakano, S.C. Chao, L. Pulkkinen, D. Murrell, L. Bruckner-
ceptor; AChE, acetylcholinesterase; EB, epidermolysis Tuderman, E. Pfendner, et al., Laminin 5 mutations in
bullosa. junctional epidermolysis bullosa: molecular basis of Herlitz
vs. non-Herlitz phenotypes, Hum. Genet. 110 (2002) 41–51.
[20] V. Matejas, B. Hinkes, F. Alkandari, L. Al-Gazali, E.
Annexstad, M.B. Aytac, et al., Mutations in the human
References laminin beta2 (LAMB2) gene and the associated phenotypic
spectrum, Hum. Mutat. 31 (2010) 992–1002.
[21] D.P. Gale, D.D. Oygar, F. Lin, P.D. Oygar, N. Khan, T.M.
[1] W. Bowman, On the minute structure and movements of Connor, et al., A novel COL4A1 frameshift mutation in familial
voluntary muscle, Phil. Trans. R. Soc. Lond. Biol. Sci. 130 kidney disease: the importance of the C-terminal NC1
(1840) 457–494. domain of type IV collagen, Nephrol. Dial. Transplant. 31
[2] A. Naba, K.R. Clauser, H. Ding, C.A. Whittaker, S.A. Carr, (2016) 1908–1914.
R.O. Hynes, The extracellular matrix: tools and insights for [22] D.B. Gould, F.C. Phalan, G.J. Breedveld, S.E. Van Mil, R.S.
the “omics” era, Matrix Biol. 49 (2016) 10–24. Smith, J.C. Schimenti, et al., Mutations in Col4a1 cause
[3] J.E. Murphy-Ullrich, E.H. Sage, Revisiting the matricellular perinatal cerebral hemorrhage and porencephaly, Science
concept, Matrix Biol. 37 (2014) 1–14. 308 (2005) 1167–1171.
10 Basement membrane biology

[23] D.B. Gould, F.C. Phalan, S.E. VanMil, J.P. Sundberg, K. myotonic myopathy and chondrodysplasia, Am. J. Hum.
Vahedi, P. Massin, et al., Role of COL4A1 in small-vessel Genet. 70 (2002) 1368–1375.
disease and hemorrhagic stroke, N. Engl. J. Med. 354 (2006) [39] E. Arikawa-Hirasawa, W.R. Wilcox, Y. Yamada, Dysseg-
1489–1496. mental dysplasia, Silverman-Handmaker type: unexpected
[24] M.V. Alavi, M. Mao, B.T. Pawlikowski, M. Kvezereli, J.L. role of perlecan in cartilage development, Am. J. Med. Genet.
Duncan, R.T. Libby, et al., Col4a1 mutations cause progres- 106 (2001) 254–257.
sive retinal neovascular defects and retinopathy, Sci. Rep. 6 [40] S. Nicole, C.-S. Davoine, H. Topaloglu, L. Cattolico, D.
(2016) 18602. Barral, P. Beighton, et al., Perlecan, the major proteoglycan
[25] M. Jeanne, D.B. Gould, Genotype-phenotype correlations in of basement membranes, is altered in patients with
pathology caused by collagen type IV alpha 1 and 2 Schwartz-Jampel syndrome (chondrodystrophic myotonia),
mutations, Matrix Biol. 57-58 (2017) 29–44. Nat. Genet. 26 (2000) 480–483.
[26] D. Cosgrove, S. Liu, Collagen IV diseases: a focus on the [41] C. Huze, S. Bauche, P. Richard, F. Chevessier, E. Goillot, K.
glomerular basement membrane in Alport syndrome, Matrix Gaudon, et al., Identification of an agrin mutation that causes
Biol. 57-58 (2017) 45–54. congenital myasthenia and affects synapse function, Am. J.
[27] R.J. Butterfield, A.R. Foley, J. Dastgir, S. Asman, D.M. Dunn, Hum. Genet. 85 (2009) 155–167.
Y. Zou, et al., Position of glycine substitutions in the triple [42] R.A. Maselli, J.M. Fernandez, J. Arredondo, C. Navarro, M.
helix of COL6A1, COL6A2, and COL6A3 is correlated with Ngo, D. Beeson, et al., LG2 agrin mutation causing severe
severity and mode of inheritance in collagen VI myopathies, congenital myasthenic syndrome mimics functional charac-
Hum. Mutat. 34 (2013) 1558–1567. teristics of non-neural (z-) agrin, Hum. Genet. 131 (2012)
[28] M. Ryynänen, J. Ryynänen, S. Solberg, R.V. Iozzo, R.G. 1123–1135.
Knowlton, J. Uitto, Genetic linkage of Type VII collagen [43] L. Rudnicka, J. Varga, A.M. Christiano, R.V. Iozzo, S.A.
(COL7A1) to dominant dystrophic epidermolysis bullosa in Jimenez, J. Uitto, Elevated expression of type VII collagen in
families with abnormal anchoring fibrils, J. Clin. Invest. 89 the skin of patients with systemic sclerosis, J. Clin. Invest. 93
(1992) 974–980. (1994) 1709–1715.
[29] H. Vahidnezhad, L. Youssefian, S. Zeinali, A.H. Saeidian, S. [44] K.J. McCarthy, The basement membrane proteoglycans
Sotudeh, N. Mozafari, et al., Dystrophic epidermolysis perlecan and agrin: something old, something new, Curr.
bullosa: COL7A1 mutation landscape in a multi-ethnic cohort Top. Membr. 76 (2015) 255–303.
of 152 extended families with high degree of customary [45] S.S. Wijeratne, J.R. Martinez, B.J. Grindel, E.W. Frey, J. Li,
consanguineous marriages, J. Investig. Dermatol. (2016). K. Wnag, et al., Single molecule force measurements of
[30] V.F. Oliver, K.A. van Bysterveldt, M. Cadzow, B. Steger, V. perlecan/HSPG2: a key component of the osteocyte pericel-
Romano, D. Markie, et al., A COL17A1 splice-altering lular matrix, Matrix Biol. 50 (2016) 27–38.
mutation is prevalent in inherited recurrent corneal erosions, [46] M.S. Lord, C.Y. Chuang, J. Melrose, M.J. Davies, R.V. Iozzo,
Ophthalmology 123 (2016) 709–722. J.M. Whitelock, The role of vascular-derived perlecan in
[31] F. Jonsson, B. Bystrom, A.E. Davidson, L.J. Backman, T.G. modulating cell adhesion, proliferation and growth factor
Kellgren, S.J. Tuft, et al., Mutations in collagen, type XVII, signaling, Matrix Biol. 35 (2014) 112–122.
alpha 1 (COL17A1) cause epithelial recurrent erosion [47] R.V. Iozzo, Basement membrane proteoglycans: from cellar
dystrophy (ERED), Hum. Mutat. 36 (2015) 463–473. to ceiling, Nat. Rev. Mol. Cell Biol. 6 (2005) 646–656.
[32] J.W. Bauer, C. Lanschuetzer, Type XVII collagen gene [48] R.V. Iozzo, L. Schaefer, Proteoglycan form and function: a
mutations in junctional epidermolysis bullosa and prospects comprehensive nomenclature of proteoglycans, Matrix Biol.
for gene therapy, Clin. Exp. Dermatol. 28 (2003) 53–60. 42 (2015) 11–55.
[33] O. Suzuki, E. Kague, K. Bagatini, H. Tu, R. Heljasvaara, L. [49] J.J. Zoeller, A. McQuillan, J. Whitelock, S.-Y. Ho, R.V. Iozzo,
Carvalhaes, et al., Novel pathogenic mutations and skin biopsy A central function for perlecan in skeletal muscle and
analysis in Knobloch syndrome, Mol. Vis. 15 (2009) 801–809. cardiovascular development, J. Cell Biol. 181 (2008)
[34] R. Heljasvaara, M. Aikio, H. Ruotsalainen, T. Pihlajaniemi, 381–394.
Collagen XVIII in tissue homeostasis and dysregulation - [50] J.J. Zoeller, J. Whitelock, R.V. Iozzo, Perlecan regulates
lessons learned from model organisms and human patients, developmental angiogenesis by modulating the VEGF-
Matrix Biol. 57-58 (2017) 55–75. VEGFR2 axis, Matrix Biol. 28 (2009) 284–291.
[35] O.T. Suzuki, A.L. Sertie, K. Der, V.F. Kok, M. Carpenter, J. [51] R.V. Iozzo, J.J. Zoeller, A. Nyström, Basement membrane
Murray, et al., Molecular analysis of collagen XVIII reveals proteoglycans: modulators par excellence of cancer growth
novel mutations, presence of a third isoform, and possible and angiogenesis, Mol. Cell 27 (2009) 503–513.
genetic heterogeneity in Knobloch syndrome, Am. J. Hum. [52] K. Baghy, P. Tatrai, E. Regos, I. Kovalszky, Proteoglycans in
Genet. 71 (2002) 1320–1329. liver cancer, World J. Gastroenterol. 22 (2016) 379–393.
[36] Y. Lin, S. Zhang, M. Rehn, P. Itaranta, J. Tuukkanen, R. [53] M.C. Farach-Carson, C.R. Warren, D.A. Harrington, D.D.
Heljasvaara, et al., Induced repatterning of type XVIII Carson, Border patrol: insights into the unique role of
collagen expression in ureter bud from kidney to lung type: perlecan/heparan sulfate proteoglycan 2 at cell and tissue
association with sonic hedgehog and ectopic surfactant borders, Matrix Biol. 34 (2014) 64–79.
protein C, Development 128 (2001) 1573–1585. [54] G. Bix, J. Fu, E. Gonzalez, L. Macro, A. Barker, S. Campbell,
[37] E. Arikawa-Hirasawa, W.R. Wilcox, A.H. Le, N. Silverman, P. et al., Endorepellin causes endothelial cell disassembly of
Govindraj, J.R. Hassell, et al., Dyssegmental dysplasia, Silver- actin cytoskeleton and focal adhesions through the α2β1
man-Handmaker type, is caused by functional null mutations of integrin, J. Cell Biol. 166 (2004) 97–109.
the perlecan gene, Nat. Genet. 27 (2001) 431–434. [55] G. Bix, R. Castello, M. Burrows, J.J. Zoeller, M. Weech, R.A.
[38] E. Arikawa-Hirasawa, A.H. Le, I. Nishino, I. Nonaka, N.C. Ho, Iozzo, et al., Endorepellin in vivo: targeting the tumor
C.A. Francomano, et al., Structural and functional mutations vasculature and retarding cancer growth and metabolism,
of the perlecan gene cause Schwartz-Jampel syndrome, with J. Natl. Cancer Inst. 98 (2006) 1634–1646.
Basement membrane biology 11

[56] G. Bix, R.V. Iozzo, Matrix revolutions: “tails” of basement- the C terminus of perlecan, J. Biol. Chem. 278 (2003)
membrane components with angiostatic functions, Trends 4238–4249.
Cell Biol. 15 (2005) 52–60. [74] E.M. Gonzalez, C.C. Reed, G. Bix, J. Fu, Y. Zhang, B.
[57] B.P. Woodall, A. Nyström, R.A. Iozzo, J.A. Eble, S. Niland, T. Gopalakrishnan, et al., BMP-1/Tolloid-like metalloproteases
Krieg, et al., Integrin α2β1 is the required receptor for process endorepellin, the angiostatic C-terminal fragment of
endorepellin angiostatic activity, J. Biol. Chem. 283 (2008) perlecan, J. Biol. Chem. 280 (2005) 7080–7087.
2335–2343. [75] M.S. O'Reilly, T. Boehm, Y. Shing, N. Fukai, G. Vasios, W.S.
[58] A. Goyal, N. Pal, M. Concannon, M. Paulk, M. Doran, C. Lane, et al., Endostatin: an endogenous inhibitor of angio-
Poluzzi, et al., Endorepellin, the angiostatic module of genesis and tumor growth, Cell 88 (1997) 277–285.
perlecan, interacts with both the α2β1 integrin and vascular [76] B. Grindel, Q. Li, R. Arnold, J. Petros, M. Zayzafoon, M.
endothelial growth factor receptor 2 (VEGFR2), J. Biol. Muldoon, et al., Perlecan/HSPG2 and matrilysin/MMP-7 as
Chem. 286 (2011) 25947–25962. indices of tissue invasion: tissue localization and circulating
[59] S. Douglass, A. Goyal, R.V. Iozzo, The role of perlecan and perlecan fragments in a cohort of 288 radical prostatectomy
endorepellin in the control of tumor angiogenesis and patients, Oncotarget 7 (2016) 10433–10447.
endothelial cell autophagy, Connect. Tissue Res. 19 (2015) [77] M.C. Farach-Carson, A.C. Brown, M. Lynam, J.B. Safran,
1–11. D.D. Carson, A novel peptide sequence in perlecan domain
[60] C. Poluzzi, R.V. Iozzo, L. Schaefer, Endostatin and endor- IV supports cell adhesion, spreading and FAK activation,
epellin: a common route of action for similar angiostatic Matrix Biol. 27 (2008) 150–160.
cancer avengers, Adv. Drug Deliv. Rev. 97 (2016) 156–173. [78] G. Bezakova, M.A. Rüegg, New insights into the roles of
[61] C. Poluzzi, J. Casulli, A. Goyal, T.J. Mercer, T. Neill, R.V. agrin, Nat. Rev. Mol. Cell Biol. 4 (2003) 295–308.
Iozzo, Endorepellin evokes autophagy in endothelial cells, J. [79] M. Gesemann, A. Brancaccio, B. Schumacher, M.A. Ruegg,
Biol. Chem. 289 (2014) 16114–16128. Agrin is a high-affinity binding protein of dystroglycan in non-
[62] A. Goyal, M.A. Gubbiotti, D.R. Chery, L. Han, R.V. Iozzo, muscle tissue, J. Biol. Chem. 273 (1998) 600–605.
Endorepellin-evoked autophagy contributes to angiostasis, J. [80] K.K. McKee, D.H. Yang, R. Patel, Z.L. Chen, S. Strickland, J.
Biol. Chem. 291 (2016) 19245–19256. Takagi, et al., Schwann cell myelination requires integration
[63] L. Ning, Z. Xu, N. Furuya, R. Nonaka, Y. Yamada, E. of laminin activities, J. Cell Sci. 125 (2012) 4609–4619.
Arikawa-Hirasawa, Perlecan inhibits autophagy to maintain [81] J. Moll, P. Barzaghi, S. Lin, G. Bezakova, H. Lochmuller, E.
muscle homeostasis in mouse soleus muscle, Matrix Biol. 48 Engvall, et al., An agrin minigene rescues dystrophic
(2015) 26–35. symptoms in a mouse model for congenital muscular
[64] M.A. Gubbiotti, R.V. Iozzo, Proteoglycans regulate autoph- dystrophy, Nature 413 (2001) 302–307.
agy via outside-in signaling: an emerging new concept, [82] K.K. McKee, S. Capizzi, P.D. Yurchenco, Scaffold-forming
Matrix Biol. 48 (2015) 6–13. and adhesive contributions of synthetic laminin-binding
[65] A.J. Kvist, A.E. Johnson, M. Mörgelin, E. Gustafsson, E. proteins to basement membrane assembly, J. Biol. Chem.
Bengtsson, K. Lindblom, et al., Chondroitin sulfate perlecan 284 (2009) 8984–8994.
enhances collagen fibril formation. Implications for perlecan [83] L. Thadikkaran, D. Crettaz, M.A. Siegenthaler, D. Gallot, V.
chondrodysplasias, J. Biol. Chem. 281 (2006) 33127–33139. Sapin, R.V. Iozzo, et al., The role of proteomics in the
[66] V. Arpino, M. Brock, S.E. Gill, The role of TIMPs in regulation assessment of premature rupture of fetal membranes, Clin.
of extracellular matrix proteolysis, Matrix Biol. 44–46 (2015) Chim. Acta 360 (2005) 27–36.
247–254. [84] A. Stephan, J.M. Mateos, S.V. Kozlov, P. Cinelli, A.D. Kistler,
[67] J.M. Wells, A. Gaggar, J.E. Blalock, MMP generated S. Hettwer, et al., Neurotrypsin cleaves agrin locally at the
matrikines, Matrix Biol. 44–46 (2015) 122–129. synapse, FASEB J. 22 (2008) 1861–1873.
[68] E.I. Deryugina, J.P. Quigley, Tumor angiogenesis: MMP- [85] O. Oda, T. Shinzato, K. Ohbayashi, I. Takai, M. Kunimatsu,
mediated induction of intravasation- and metastasis-sustain- K. Maeda, et al., Purification and characterization of perlecan
ing neovasculature, Matrix Biol. 44–46 (2015) 94–112. fragment in urine of end-stage renal failure patients, Clin.
[69] M.G. Rohani, W.C. Parks, Matrix remodeling by MMPs during Chim. Acta 255 (1996) 119–132.
wound repair, Matrix Biol. 44–46 (2015) 113–121. [86] E. O'Riordan, T.N. Orlova, N. Mendelev, D. Patschan, R.
[70] S. Duarte, J. Baber, T. Fujii, A.J. Coito, Matrix metallopro- Kemp, P.N. Chander, et al., Urinary proteomic analysis of
teinases in liver injury, repair and fibrosis, Matrix Biol. 44–46 chronic renal allograft nephropathy, Proteomics Clin. Appl. 2
(2015) 147–156. (2008) 1025–1035.
[71] U. Eckhard, P.F. Huesgen, O. Schilling, C.L. Bellac, G.S. [87] G.T. Tsangaris, P. Karamessinis, A. Kolialexi, S.D. Garbis, A.
Butler, J.H. Cox, et al., Active site specificity profiling of the Antsaklis, A. Mavrou, et al., Proteomic analysis of amniotic
matrix metalloproteinase family: proteomic identification of fluid in pregnancies with Down syndrome, Proteomics 6
4300 cleavage sites by nine MMPs explored with structural (2006) 4410–4419.
and synthetic peptide cleavage analyses, Matrix Biol. 49 [88] J.W. Chang, U.-B. Kang, D.H. Kim, J.K. Yi, J.W. Lee, D.-Y.
(2016) 37–60. Noh, et al., Identification of circulating endorepellin LG3
[72] A.M. Muir, D. Massoudi, N. Nguyen, D.R. Keene, S.J. Lee, fragment: potential use as a serological biomarker for breast
D.E. Birk, et al., BMP1-like proteinases are essential to the cancer, Proteomics Clin. Appl. 2 (2008) 23–32.
structure and wound healing of skin, Matrix Biol. 56 (2016) [89] C.B. Marshall, Rethinking glomerular basement membrane
114–131. thickening in diabetic nephropathy: adaptive or pathogenic?
[73] M. Mongiat, S. Sweeney, J.D. San Antonio, J. Fu, R.V. Iozzo, Am. J. Physiol. Ren. Physiol. 311 (2016) F831–F843.
Endorepellin, a novel inhibitor of angiogenesis derived from

You might also like