Hostname: page-component-8448b6f56d-jr42d Total loading time: 0 Render date: 2024-04-16T10:03:47.314Z Has data issue: false hasContentIssue false

Elastic fibres in health and disease

Published online by Cambridge University Press:  20 August 2013

Andrew K. Baldwin
Affiliation:
Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, UK
Andreja Simpson
Affiliation:
Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, UK
Ruth Steer
Affiliation:
Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, UK
Stuart A. Cain
Affiliation:
Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, UK
Cay M. Kielty*
Affiliation:
Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, UK
*
*Corresponding author: Cay M. Kielty, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of ManchesterM13 9PT, UK. E-mail: cay.kielty@manchester.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Elastic fibres are insoluble components of the extracellular matrix of dynamic connective tissues such as skin, arteries, lungs and ligaments. They are laid down during development, and comprise a cross-linked elastin core within a template of fibrillin-based microfibrils. Their function is to endow tissues with the property of elastic recoil, and they also regulate the bioavailability of transforming growth factor β. Severe heritable elastic fibre diseases are caused by mutations in elastic fibre components; for example, mutations in elastin cause supravalvular aortic stenosis and autosomal dominant cutis laxa, mutations in fibrillin-1 cause Marfan syndrome and Weill–Marchesani syndrome, and mutations in fibulins-4 and -5 cause autosomal recessive cutis laxa. Acquired elastic fibre defects include dermal elastosis, whereas inflammatory damage to fibres contributes to pathologies such as pulmonary emphysema and vascular disease. This review outlines the latest understanding of the composition and assembly of elastic fibres, and describes elastic fibre diseases and current therapeutic approaches.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2013 

Elastic fibres are insoluble components of the extracellular matrix (ECM) of extensible connective tissues such as large arteries, skin, lungs, ligaments and auricular cartilage (Ref. Reference Kielty1). They endow these tissues with the mechanical properties necessary to withstand repeated cycles of stretch and recoil through life. They also influence the bioavailability of transforming growth factor β (TGFβ) family growth factors (Ref. Reference Nistala2). Although fibrillin microfibrils arose in early metazoans (Refs Reference Özbek3, Reference Piha-Gossack, Sossin and Reinhardt4) (Fig. 1), elastin and elastic fibres emerged only during vertebrate evolution as an essential requirement to reinforce high-pressure circulatory systems (Ref. Reference Faury5). Elastic fibres comprise an inner core of cross-linked elastin ensheathed within fibrillin microfibrils. These long-lasting structures begin to assemble during mid-gestation, with little adult elastic fibre assembly.

Figure 1. Evolutionary analysis of fibrillin. A list of species from the fibrillin (FBN) sequence alignment was generated, and used to create a common tree using the National Center for Biotechnology Information (NCBI) Taxonomy Browser. The resulting tree was imported into Cytoscape version 2.7, and generated using the organic layout. FBN types were identified using phylogenetic analysis and domain analysis of the FBN multiple sequence alignments and FBN types were mapped onto the common tree. Identified sequences were grouped into 5 categories: vertebrate FBNs 1-3, invertebrate FBN and fibrillin-like epidermal growth factor (EGF) array-containing proteins. The latter sequences had a high identity to FBN sequences but only contained EGF domains organised in arrays. Also indicated are the sequences, from the appearance of chordates that contain the arg-gly-asp (RGD) cell binding motif (branches shown in light brown). By comparison, branches shown in light purple precede chordates and RGD. The first emergence of other key extracellular proteins that interact with FBN is shown (dark blue arrows), along with the evolutionary timescale in million years ago (MYA).

Mature elastic fibres have tissue-specific architectural arrangements that reflect different elastic requirements. Thus, arterial elastic fibres form concentric lamellar layers that support vascular elastic recoil. Dermal elasticity is based on integrated networks of thick reticular elastic fibres and thin fibres in the papillary dermis, and alveolar elastic fibres form fine networks that allow respiratory expansion and contraction (Ref. Reference Kielty1).

Although tropoelastin and fibrillin are the two principal structural components of elastic fibres, several ‘accessory’ molecules contribute to elastic fibre assembly and function. From initial microfibril formation to the deposition and cross-linking of tropoelastin monomers upon microfibrils, these accessory proteins support the progression of elastic fibre formation in a spatially and temporally appropriate manner. For example, small fibulins influence the association of elastin with its cross-linking enzymes, thereby facilitating its stable deposition on microfibrils (see the Assembly of elastic fibres section).

Aberrant elastic fibre formation and/or altered homoeostasis cause many inherited and acquired diseases, with phenotypes ranging from mild (e.g. loose skin) to severe and potentially life-threatening (e.g. vascular defects) (see the Elastic fibre disorders section). Heritable elastic fibre disorders include Marfan syndrome (MFS) caused by fibrillin-1 mutations (Ref. Reference Robinson6) and autosomal dominant cutis laxa (ADCL) caused by elastin mutations (Ref. Reference Sugitani7). Mutations in accessory glycoproteins such as fibulins-4 or -5 cause autosomal recessive cutis laxa (ARCL) (Ref. Reference Berk8), and in a disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin-10 (ADAMTS-10) cause Weill–Marchesani syndrome (WMS) (Ref. Reference Dagoneau9). Acquired elastic fibre disorders arise wherever elastic fibre structure and/or function are compromised; for example, actinic elastosis and vascular degeneration, as well as acquired forms of cutis laxa (CL) and pseudoxanthoma elasticum (PXE).

In this review, we describe the current understanding of microfibril and elastic fibre composition and formation, delineate the diseases associated with elastic fibre defects and summarise the latest therapeutic advances.

Composition of elastic fibres

The structural and associated components of elastic fibres are outlined below; their roles in elastic fibre formation and function are further delineated below (see the Assembly of elastic fibres section).

Structural molecules

Elastin

Elastin is the most abundant protein of elastic fibres, comprising approximately 90% of the mature structure. It is encoded by a gene on chromosome 7q11.2, and secreted as the soluble precursor tropoelastin (60–70 kDa) from elastogenic cells such as fibroblasts, smooth muscle cells and auricular chondrocytes. Tropoelastin has a multi-domain structure comprising alternating hydrophobic and lysine-containing (lysine–alanine and lysine–proline) cross-linking domains. The lysine–alanine-rich domains preferentially participate in desmosine cross-link formation (Ref. Reference Dyksterhuis and Weiss10). The primary transcript can undergo tissue-specific alternative splicing that modifies elastin incorporation into fibres (Ref. Reference Sugitani7).

Tropoelastin is an asymmetric molecule, which has two functionally distinct regions (Ref. Reference Baldock11): an N-terminal elastic coil that endows spring-like properties on tropoelastin, and a C-terminal cell interactive module that facilitates cell adhesion via an association between the αvβ3 integrin and a GRKRK motif (Ref. Reference Bax12). They are separated by a bridge region, within which are domains shown to contain sites of contact for coacervation (Ref. Reference Dyksterhuis13). Destabilisation of this region by mutagenesis has been shown to impair elastogenesis (Ref. Reference Yeo14).

Fibrillins

Fibrillin molecules assemble to form microfibrils (Refs Reference Hubmacher, Tiedemann and Reinhardt15, Reference Ramirez and Sakai16). In man, there are three fibrillin genes, each encoding 350 kDa multi-domain glycoproteins (fibrillins 1–3) (Ref. Reference Piha-Gossack, Sossin and Reinhardt4). Each fibrillin comprises 43 calcium-binding epidermal growth factor-like (cbEGF) domains, five EGF-like domains, seven eight-cysteine-containing (TB) motifs and two hybrid domains with similarities to TB and cbEGF-like domains (Fig. 2). There is a unique proline-rich region in the N-terminal region of fibrillin-1, with corresponding regions of fibrillin-2 and fibrillin-3 being richer in glycine.

Figure 2. Domain structures of elastic fibre molecules. The domain organisations of fibrillins, latent TGFβ-binding protein (LTBPs), fibulins, a disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin (ADAMTS) and ADAMTS-like (ADAMTSL) molecules and tropoelastin are shown, with keys for domain types.

Fibrillin-1 (gene on chromosome 15q21.1) is the most abundant fibrillin isoform; it is expressed throughout life and is required for microfibril homoeostasis, whereas fibrillin-2 (gene on chromosome 5q23-q31) and fibrillin-3 (gene on chromosome 19p3) are expressed predominantly during development (Refs Reference Charbonneau17, Reference Zhang18, Reference Corson19, Reference Sabatier20). Overlapping expression of fibrillins occurs in developing blood vessels, cartilage, bone and lungs, but tissue-specific differences are also apparent, e.g. fibrillin-1 in dermis, fibrillin-2 in peripheral nerves and fibrillin-3 in cartilage, perichondrium and developing bronchi (Ref. Reference Sabatier20). Fibrillin-2 may form the inner core of many fibrillin-1 containing microfibrils (Ref. Reference Charbonneau21). Although rarely detected in adult skin, fibrillin-2 expression can increase during wound healing and in sclerosis (Ref. Reference Brinckmann22). Mouse models confirm the critical importance of fibrillin-1 to vascular development and function. Fibrillin-1 null mice die perinatally from ruptured aortic aneurysm, whereas fibrillin-2 null mice have seemingly normal formation of the aorta, although double-null mice have a much more severe aortic phenotype than fibrillin-1 null mice (Ref. Reference Carta23). Both fibrillins thus perform partially overlapping functions during the development of the aorta and great arteries, with fibrillin-1 able to compensate for loss of fibrillin-2 but not vice versa. The fibrillin-3 gene is disrupted in rodents; so fibrillin-3 is not critical to all mammalian life (Ref. Reference Corson19).

Accessory molecules associated with microfibrils and elastic fibres

Many microfibril and elastic fibre-associated molecules have been identified by microscopy and immunochemical approaches (reviewed in Ref. Reference Kielty1; updated in Table 1). We summarise here selected molecules shown by functional analysis, mouse models and/or heritable diseases to contribute to elastic fibre formation and function; microfibril-associated molecules include the latent TGFβ binding proteins (LTBPs), ADAMTS isoforms and microfibril-associated glycoprotein (MAGPs) and elastic fibre-associated molecules include the small fibulins 3–5 and lysyl oxidase (LOX).

Table 1. Microfibril and elastic fibre-associated molecules (updated from Ref. Reference Kielty1)

Abbreviations: βIgH3, also known as transforming growth factor-β-inducible gene-H3 and as keratoepithelin, on chromosome 5q31; HS, heparan sulphate; LOX, lysyl oxidase; LOXL1, lysyl oxidase-like 1; LTBP, latent TGFβ-binding protein; MAGP, microfibril-associated glycoprotein; MFAP-1, microfibril-associated protein-1; ADAMTSL, a disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin type-I motif-like.

LTBPs: LTBPs 1–4 are large extracellular glycoproteins with a multi-domain structure that has similarities to the fibrillins, comprising cbEGF and TB domains, with an N-terminal cysteine-rich domain. They are expressed in many tissues including heart, placenta, lung, kidney, skeletal muscle and ovary and have cbEGF and TB domain homology with fibrillins (Refs Reference Piha-Gossack, Sossin and Reinhardt4, Reference Todorovic54) (Fig. 2). The third TB domain of LTBPs 1–4 can covalently bind the propeptide (latency-associated peptide, LAP) of the cytokine TGFβ (Refs Reference Gleizes55, Reference Saharinen, Taipale and Keski-Oja56), which is rendered inactive when associated with its propeptide (forming the small latent complex). Association of this complex with LTBP forms the large latent complex, which can be sequestered within the ECM, thereby regulating TGFβ bioavailability (Refs Reference Todorovic54, Reference Gleizes55, Reference Saharinen, Taipale and Keski-Oja56). C-terminal regions of LTBPs 1, 2 and 4 can interact directly with fibrillin-1 through TB4 (Ref. Reference Saharinen, Taipale and Keski-Oja56), thus implicating fibrillin microfibrils in regulating TGFβ activity. LTBP-1 (and thus latent TGFβ) can be deposited in the absence of fibrillins-1 and -2 (Ref. Reference Zilberberg26), although depletion of fibrillin-1 disrupts its pattern of extracellular deposition (Refs Reference Massam-Wu28, Reference Isogai57).

ADAMTS ( a disintegrin-like and metalloprotease (reprolysin-type) with thrombo spondin type-I motif) molecules: Several members of this superfamily, including ADAMTS-10 and ADAMTSL 2–6, have been genetically and/or functionally implicated in microfibril biology (see the Assembly of elastic fibres section). The full superfamily comprises 19 zinc metalloproteases and seven noncatalytic ADAMTS-like (ADAMTSL) proteins, varying greatly in size; many members of this family play roles in morphogenesis, angiogenesis and ovulation and in ECM deposition (Ref. Reference Apte58). Briefly, each ADAMTS protease has an N-terminal protease domain, containing a catalytic module, a disintegrin-like module, a cysteine-rich module and a single thrombospondin type I repeat (TSR) (Fig. 2). The C-terminal (or ancillary) domain is variable between family members, although many contain multiple TSRs and may confer substrate-binding specificity, with the N-terminal domain providing catalytic activity. ADAMTS-10 lacks an optimal furin cleavage site (necessary for activation of ADAMTS enzymes) and is unlikely to be catalytically active in vivo. ADAMTSL family members, including those implicated in microfibril biology, contain multiple TSRs but lack disintegrin-like and catalytic domains. Thus, low/no catalytic activity is a hallmark of ADAMTS family members with identified roles in microfibril biology.

MAGPs: MAGP-1 (a ~31 kDa glycoprotein) was originally detected in microfibril preparations from elastic and nonelastic tissues (Ref. Reference Gibson, Kumaratilake and Cleary59). It has an acidic N-terminal region enriched in proline and glutamine residues, and a cysteine-rich C-terminal portion. It colocalises widely with microfibrils (Ref. Reference Gibson, Kumaratilake and Cleary59), and has been detected in purified microfibrils by mass spectrometry (Ref. Reference Cain60). Surprisingly, mice lacking the MAGP-1 gene show normal microfibrils and elastic fibre assembly (Refs Reference Craft24, Reference Weinbaum61), so it is not essential for elastogenesis. Structurally related glycoprotein MAGP-2 (~25 kDa) has a cysteine-rich central region, is rich in serine and threonine and binds integrins. It colocalises with microfibrils in some tissues (Refs Reference Lemaire27, Reference Gibson62, Reference Penner63, Reference Hanssen64). Both MAGPs can bind fibrillin and microfibrils, and may enhance the deposition of elastin on microfibrils (see the Assembly of elastic fibres section).

Fibulins: Fibulins are extracellular glycoproteins that are classified as Class I or II on the basis of their domain structure and length (Ref. Reference Yanagisawa and Davis39). The closely related Class II (or short) fibulins (fibulins 3–5; 50–60 kDa) each comprise six cbEGF domains, including an atypical N-terminal cbEGF and a C-terminal fibulin (‘FC’) module (Fig. 2) and are widely expressed in developing embryos, particularly cardiovascular and skeletal tissues (Ref. Reference Yanagisawa and Davis39). Fibulins-4 and -5 can bind tropoelastin and LOX or LOX-like (LOXL) cross-linking enzymes, as well as fibrillin-1; by juxtaposing these molecules, they are thought to support the cross-linking of elastin and its deposition on microfibrils.

LOX family: The five members of the LOX family (LOX, LOXL1-4) are secreted as zymogens, and are activated by removal of N-terminal propeptides by bone morphogenetic protein-1 (BMP-1) (Ref. Reference Molnar65). LOX especially, but also its homologue LOXL1 catalyses the oxidative deamination of peptidyl lysine residues in elastin to generate α-aminoadipic-δ-semialdehydes that then spontaneously condense to form covalent desmosine and isodesmosine cross-links, and a very stable insoluble elastin core (Ref. Reference Molnar65). Their C-terminal regions show sequence homology but their N-terminal ‘pro’ regions vary greatly, and may impart specificity to the extracellular targeting of LOX enzymes.

Assembly of elastic fibres

Microfibril formation

Assembly of fibrillin monomers into microfibrils is a complex process. ‘Pro'fibrillin molecules are processed N- and C-terminally by furin/PACE proprotein convertases, either immediately before or upon secretion, thereby facilitating terminal interactions that enable linear and lateral multimerisation (Ref. Reference Marson66). Homotypic N-terminal interactions are enhanced by heparan sulphate (HS) (Ref. Reference Cain67), whereas C-terminal interactions may underpin bead formation (Ref. Reference Hubmacher68). In this way, microfibrils are thought to assemble at the cell surface (Fig. 3a). Microfibril formation is unlikely to be solely a self-assembly process, and there is some evidence for cellular involvement. We reported that, in fibroblasts, microfibril deposition requires fibronectin arg–gly–asp (RGD)-dependent α5β1 integrins (Ref. Reference Kinsey70). Fibrillin-1 can also interact with cells through integrins α5β1 and αvβ3, and αvβ6 in epithelial cells (Refs Reference Bax71, Reference Jovanovic72). It remains to be determined whether direct fibrillin-1 interactions with cells are necessary for microfibril assembly and/or for specific tissue-specific functions of microfibrils (and elastic fibres) such as association with smooth muscle cells in the aorta.

Figure 3. Schematic diagram of microfibril and elastic fibre assembly: elastin assembly. (a) Microfibril assembly occurs pericellularly, and requires fibronectin, integrins and heparan sulphate proteoglycans (HSPG). Fibrillin molecules are secreted and, after processing N- and C-terminally by furin, interact homotypically at N- and C-termini leading to axial and lateral assembly to form microfibrils. Beads may arise from folding of terminal regions. Microfibrils may be stabilised by transglutaminase cross-links. The reason why fibronectin is needed for microfibril deposition is unclear, but it may act as a template for assembly and/or it may stimulate cytoskeletal tension through the α5β1 integrin, thereby facilitating assembly at fibrillar adhesions. Fibrillin-1 also interacts with α5β1, αvβ3 and αvβ6 integrins; however, it is not known whether these interactions are essential for microfibril assembly. Heparin inhibits microfibril assembly, and HSPGs may contribute by facilitating cell surface fibrillin-1 interactions. (b) Elastin assembly occurs pericellularly on ‘microassembly’ and on microfibrils ‘macroaggregates’ (Ref. Reference Wagenseil and Mecham69). Secreted tropoelastin forms globules at the cell surface which become cross-linked by lysyl oxidase; this process may involve αvβ3 integrin interactions with tropoelastin, and integrin interactions with heparan sulphate proteoglycans (HSPGs) which can interact with tropoelastin. Fibulin-4 and fibulin-5 contribute to elastin cross-linking by lysyl oxidase, and probably direct the deposition of elastin globules onto preformed fibrillin microfibrils, to form elastic fibres. Microfibrils and elastic fibres are important matrix storage sites for BMPs and latent TGFβ1.

Assembled microfibrils comprise fibrillin monomers aligned in a head-to-tail arrangement and laterally associated (probably eight molecules in cross-section) (Refs Reference Reinhardt73, Reference Baldock74, Reference Wang, Lu and Baldock75). Fibrillin molecules within tissue microfibrils can be cross-linked by transglutaminase (Ref. Reference Qian and Glanville76). The ‘beads-on-a-string’ appearance of individual microfibrils, their untensioned periodicity (50–60 nm) in tissue sections (Refs Reference Reinhardt73, Reference Baldock74) and their altered structural organisation upon extension (Refs Reference Wang, Lu and Baldock75, Reference Baldock77) has given rise to unstaggered (Refs Reference Baldock74, Reference Wang, Lu and Baldock75) and staggered (Refs Reference Jensen, Robertson and Handford78, Reference Sabatier79) models of fibrillin alignment within microfibrils. The precise alignment of fibrillin molecules within microfibrils awaits further insights, perhaps from approaches such as experimental cross-linking.

Role for fibronectin

In mesenchymal cultures, fibronectin is an essential prerequisite for microfibril formation (Refs Reference Zilberberg26, Reference Kinsey70, Reference Sabatier79). This relationship is surprising because fibrillin microfibrils arose in early metazoans and fibronectin-like molecules only in chordates (Refs Reference Özbek3, Reference Piha-Gossack, Sossin and Reinhardt4, Reference Tucker and Chiquet-Ehrismann80). Fibronectin is able to bind several regions of fibrillin-1, as well as fibrillin multimers, whereas newly deposited microfibrils and fibronectin networks show initial colocalisation (Refs Reference Kinsey70, Reference Sabatier79, Reference Hubmacher81). It has been suggested that fibronectin could act as a template for microfibril deposition (Refs Reference Kinsey70, Reference Sabatier79); however fibrillin microfibrils occur in many lower organisms that lack fibronectin (Ref. Reference Reber-Müller82), and in fibronectin-null cultures (Ref. Reference Dallas83), implying that fibronectin enhances rather than underpins microfibril assembly.

Role for HS

The glycosaminoglycan HS is strongly implicated in microfibril and elastic fibre assembly. It is a component of syndecan and glypican receptors, and of the basement membrane molecule perlecan. Supplementation of cell cultures with heparin effectively ablates microfibril assembly (Refs Reference Tiedemann84, Reference Ritty85). At least six high affinity-binding sites between heparin and fibrillin-1 have been identified (Refs Reference Cain67, Reference Tiedemann84, Reference Ritty85, Reference Cain86), some of which direct N-terminal multimerisation during microfibril assembly (Ref. Reference Cain67) or interactions with cell surface HS (Ref. Reference Bax71). Although it is not known precisely how HS contributes to microfibril deposition, these HS-directed interactions may be essential. Heparin does not disrupt N- and C-terminal fibrillin-1 interactions and so these interactions are compatible with microfibril assembly (Ref. Reference Cain86), and may directly support these homotypic interactions. Murine perlecan (recombinant fragment or molecules isolated from Engelbreth-Holm-Swarm (EHS) sarcoma; Ref. Reference Tiedemann35) or human perlecan expressed by ARPE-19 retinal epithelial cells (Ref. Reference Cain60) binds fibrillin-1 through protein and HS interactions, and directs microfibril-basement membrane interactions. HS competes with MAGP-1 and tropoelastin (Ref. Reference Cain67) to bind fibrillin-1; hence, it may regulate elastin deposition onto microfibrils. HS can also mediate smooth muscle cell interactions with tropoelastin, thus influencing cell phenotype (Ref. Reference Akhtar87). It also directly influences elastin multimerisation (Ref. Reference Gheduzzi88).

Roles for microfibril-associated molecules

LTBP-1 co-localises with microfibrils in some tissues (Ref. Reference Reber-Müller82). This juxtaposition, together with the ability of LTBPs (Refs Reference Kielty1, Reference Özbek3, Reference Piha-Gossack, Sossin and Reinhardt4) to form large latent TGFβ complexes, and to bind fibrillin-1 (Ref. Reference Ono89), implicates microfibrils in TGFβ sequestration. Recently, it was shown that fibrillins are needed for matrix deposition of LTBP-3, but not LTBP-1 (Ref. Reference Zilberberg26). LTBP-1 deposition in the ECM is however, such as fibrillin-1, dependent on fibronectin and LTBP-1 colocalises with microfibrils but not fibronectin over time in culture (Refs Reference Zilberberg26, Reference Reber-Müller82). Fibronectin is also required for LTBP-4 deposition (Ref. Reference Kantola90). However, incorporation of LTBP-2, which does not bind TGFβ, as well as LTBP-3 and LTBP-4, into ECM is dependent on microfibrils (Refs Reference Zilberberg26, Reference Vehvilainen91). LTBP-2 also competes with LTBP-1 to interact with the N-terminal region of fibrillin-1 (Ref. Reference Hirani92). Thus, while none of the LTBPs is necessary for fibrillin microfibril assembly, microfibrils and fibronectin both profoundly influence the extracellular deposition of LTBPs.

Several members of the ADAMTS superfamily are genetically and functionally implicated in microfibril biology (Ref. Reference Hubmacher and Apte93). ADAMTS-10, which causes autosomal recessive WMS (see the Elastic fibre disorders section) may not be physiologically cleaved by furin and is probably not an active enzyme in vivo. ADAMTSL-5 binds both fibrillin-1 and -2, as well as heparin, and co-localises with microfibrils (Ref. Reference Bader51). ADAMTSL-6 binds to the N-terminal region of fibrillin-1, whereas the conditioned medium-containing recombinant ADAMTSL-6 suggested potential to promote fibrillin-1 matrix assembly (Ref. Reference Tsutsui52), and it enhanced microfibrils in an MFS mouse model (Ref. Reference Saito94). ADAMTSL-3 is implicated in a molecular pathway involving fibrillin-1 and ADAMTS-10 (Ref. Reference Sengle49); it can bind fibrillin-1 and may participate in microfibril biogenesis (Ref. Reference Kutz95). Supplementation with recombinant ADAMTSL-4 accelerated fibrillin-1 deposition in culture, and co-localisation with microfibrils (Ref. Reference Gabriel50). It is interesting to speculate that inactive ADAMTS-10 and the ADAMTSL 3–6 molecules could act as ‘decoys’ to protect fibrillins from active ADAMTS enzymes.

MAGP-1 can interact with elastin and may contribute to elastin deposition on microfibrils (Refs Reference Jensen25, Reference Rock96). It strongly binds an N-terminal sequence of fibrillin-1, thereby inhibiting N- and C-terminal fibrillin-1 interactions (Ref. Reference Jensen25), and thus has the potential to fine-tune fibrillin multimerisation. MAGP-1 can also bind TGFβ and BMP-7 (Ref. Reference Weinbaum61), and can activate pSmad2 signalling in culture (Ref. Reference Massam-Wu28), so it may contribute to regulating TGFβ growth factors in association with microfibrils (Ref. Reference Weinbaum61). Intracellular MAGP-1 can modulate expression of versican (Ref. Reference Segade97), which interacts with microfibrils (Ref. Reference Isogai34) and is a regulator of matrix. MAGP-1 can also bind fibronectin (Ref. Reference Werneck98) and could influence fibronectin-mediated microfibril deposition. None of these putative functional contributions to elastic fibre formation can be essential since MAGP-1 null mice have functional elastic fibres (Ref. Reference Craft24). MAGP-2 can interact with fibrillins-1 and -2, has covalent periodic association with isolated microfibrils and co-localises with microfibrils in certain tissues (Refs Reference Lemaire27, Reference Penner63, Reference Hanssen64). MAGP-2 expression peaks during elastic fibre assembly; evidence shows that its overexpression increases elastic fibre formation and that it can stimulate elastic fibre assembly, probably by targeting elastin onto microfibrils (Ref. Reference Lemaire27).

Elastic fibre formation

Tropoelastin has the propensity to self-assemble through a process termed coacervation, which involves rapid molecular association, at increasing temperatures (Ref. Reference Yeo, Keeley and Weiss99). Exposed hydrophobic domains interact in an entropically-driven process, resulting in lysine residues aligning in readiness for LOX-mediated cross-linking. In vitro, the sizes and properties of coacervation ‘droplets’, and the rate at which coacervation and maturation processes proceed, are dependent on tropoelastin concentration, pH, temperature and solution conditions (Ref. Reference Cirulis and Keeley100), as well as the presence of accessory proteins. Fibulin-5 and also fibulin-4, limit aggregation of tropoelastin and slows the maturation phase (Ref. Reference Choi101). The N-terminal region of fibrillin-1 and MAGP-1 both increase maturation velocity and induce clustering of elastin droplets, and the fibrillin-1 fragment induces ‘strings’ of linear droplets (Ref. Reference Cirulis and Keeley100).

In culture and tissues, elastin can self-associate pericellularly into globules that become cross-linked prior to deposition onto microfibrils (Refs Reference Wagenseil and Mecham69, Reference Czirok102, Reference Sato103) (Fig. 3b). This initial stage in elastic fibre assembly, termed ‘microassembly’, may involve interaction of the C-terminal region of tropoelastin with αvβ3 integrin (Ref. Reference Bax12) and HS (Refs Reference Akhtar87, Reference Broekelmann104). Certainly, this region of elastin is critical for elastic fibre assembly (Ref. Reference Sugitani7). In culture, pericellular globule aggregation is coupled with cell motion (Ref. Reference Czirok102), with globule size probably limited by accessory proteins (Ref. Reference Wagenseil and Mecham69) and biophysical constraints (Ref. Reference Cirulis and Keeley100). Once transferred to the microfibril scaffold, the globules coalesce and are stabilised by formation of cross-links facilitated by LOX or LOXL1, probably facilitated by fibulins-4 or -5 (see below), thus forming the insoluble elastin core in a process termed ‘macroassembly’ (Fig. 3b) (Refs Reference Yeo, Keeley and Weiss99, Reference Cirulis and Keeley100).

Roles for fibulins

The homologous small fibulins (Reference Özbek3Reference Faury5) are critical regulators of elastic fibre assembly (Refs Reference Yanagisawa and Davis39, Reference Kobayashi41). Fibulin-4 colocalises with microfibrils (Ref. Reference Kobayashi41), and binds fibrillin-1 in vitro (Ref. Reference Choudhury42). Fibulin-4 depletion decreases elastic fibre formation but not microfibrillogenesis (Ref. Reference McLaughlin43). Its involvement in the pathogenesis of the disease CL (see the Elastinopathies section) identifies it as an essential element of elastic fibre formation. Fibulin-4 null mice, which die perinatally, have a drastic reduction in desmosine cross-links and grossly impaired elastic fibre formation (Ref. Reference McLaughlin43). Fibulin-4 can bind LOX concurrently with tropoelastin (Ref. Reference Choudhury42) and influence LOX-tropoelastin interactions (Ref. Reference Horiguchi44). Thus, it is considered that fibulin-4 is essential for LOX-mediated elastin cross-linking, probably by forming complexes that juxtapose LOX and elastin (Refs Reference Choudhury42, Reference Horiguchi44). Fibulin-4 may also play a, as yet poorly defined, role in the sequestration of LTBPs (and thus latent TGFβ) within the ECM (Ref. Reference Ono89). Fibulin-5 null mice exhibit a less dramatic elastic fibre phenotype than fibulin-4 null mice, with only 16% decrease in desmosine levels; mice survive well into adulthood but have loose skin (Refs Reference Yanagisawa45, Reference Nakamura46). Fibulin-5 localises at the interface between the elastin core and microfibrils (Ref. Reference Kobayashi41). It can interact with LOXL enzymes, and it enhances tropoelastin self-association and elastic fibre formation (Ref. Reference Hirai47). Fibulin-5 is also capable of integrin-mediated cell attachment, but does not activate α5β1 integrin (Ref. Reference Lomas48). Fibulin-5 may act as a molecular adapter that directs the deposition of elastin microaggregates onto microfibrils (Refs Reference Choudhury42, Reference Wagenseil and Mecham69, Reference El-Hallous105, Reference Zheng106). Indeed, fibulin-5 knockdown in culture-altered elastin aggregates (Ref. Reference Choudhury42). Like its homologues, fibulin-3 null mice exhibit elastic fibre deficiencies in some tissues as well as herniation (Ref. Reference McLaughlin40). A fibulin-3 point mutation (R345W) caused malattia leventinese, a dominantly inherited macular degenerative disease with sub-retinal pigment epithelial deposits (Refs Reference Michaelides107, Reference Marmorstein108) with similarities to age-related macular degeneration (ARMD).

The different severities and tissue defects induced by depletion of fibulins 3–5 may reflect, in part, their tissue-specific expression patterns. It is also likely that some evolutionary diversification of their roles in elastic fibre assembly has occurred, e.g. fibulin-4 preferentially interacts with LOX and fibulin-5 with LOXL enzymes (Refs Reference Choudhury42, Reference Horiguchi44, Reference Hirai47).

Elastic fibre disorders

Inherited and acquired diseases of elastic fibres are also reviewed in Ref. Reference Kielty1 (updated in Table 2).

Table 2. Heritable disorders of elastic fibres (updated from Ref. Reference Kielty1)

Abbreviations: ADAMTSL, a disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin like; ADCL, autosomal dominant cutis laxa; ARCL, autosomal recessive cutis laxa; ARMD, age-related macular degeneration; CCA, congenital contractural arachnactyly; ECM, extracellular matrix; GD, geleophysic dysplasias; HS, heparan sulphate; LOX, lysyl oxidase; LTBP, latent TGFβ-binding protein; MACS, macrocephaly, alopaecia, CL and scoliosis; MFS, Marfan syndrome; P5CS, Δ1-pyrroline-5-carboxylate synthase; PXE, pseudoxanthoma elasticum, SVAS, supravalvular aortic stenosis; TGFβ, transforming growth factor β; WBS, Williams-Beuren syndrome; WMS, Weill-Marchesani syndrome; XLCL, X-linked cutis laxa.

Fibrillinopathies

Heritable connective tissue disorders termed fibrillinopathies arise from mutations in fibrillin genes. Fibrillin-1 disorders include MFS, ectopia lentis, MASS (mitral valve prolapse, aortic enlargement, skin and skeletal findings) syndrome, Shprintzen–Goldberg syndrome, Stiff skin syndrome, autosomal dominant WMS and acromicric and geleophysic dysplasias (AD, GD) (Refs Reference Robinson6, Reference Dietz and Pagon109, Reference Loeys116, Reference Faivre115). Fibrillin-1 is also implicated in the pathogenesis of homocystinuria, which exhibits MFS-like symptoms (Ref. Reference Dallas83). Congenital contractural arachnodactyly (CCA; Beal's syndrome) arises from mutations in fibrillin-2, and polycystic ovary syndrome is associated with mutations in fibrillin-3 (Ref. Reference Jordan113). In addition, mutations in LTBP-2 and the ADAMTS family members ADAMTS-10, ADAMTS-17, ADAMTSL-2 and ADAMTSL-4 are associated with the fibrillin-related disorders WMS, a Weill–Marchesani-like syndrome, acromicric and GD and ectopia lentis, demonstrating genetic and functional links between these proteins and fibrillin microfibrils (Refs Reference Dagoneau9, Reference Haji-Seyed-Javadi147, Reference Morales148, Reference Le Goff149).

MFS and related pathologies

MFS (OMIM 154700) is the most prevalent fibrillinopathy, with an incidence of 2–3 per 10,000 people. A complex and phenotypically heterogeneous systemic disorder, MFS is characterised by severe cardiovascular, skeletal and ocular abnormalities, with symptoms ranging from isolated features through progressive classical MFS to severe early onset and rapidly progressive MFS (associated with early childhood mortality). Disease symptoms can reflect both elastic fibre deficiencies and perturbed TGFβ activity (Refs Reference Robinson6, Reference Dietz and Pagon109). A life-threatening complication of MFS is aortic rupture due to dilation of the ascending aorta.

Although commonly inherited as an autosomal dominant trait, 25% of cases arise from de novo mutations; well over 1000 causative fibrillin-1 mutations have been identified. Mutations often affect disulphide bond patterns or calcium-binding consensus sequences within cbEGF domains, resulting in structural alterations that alter assembly and/or integrity of microfibrils and elastic fibres (Ref. Reference Robinson6). There is no single mechanism from fibrillin-1 mutation to disease; mouse models and recombinant fibrillin-1 studies have shown that altered secretion, decreased expression, structural defects and increased proteolytic susceptibility of mutant fibrillin-1 proteins and perturbations in TGFβ activity can all contribute to MFS phenotypes (Refs Reference Robinson6, Reference Dietz and Pagon109, Reference Mellody150, Reference Charbonneau151, Reference Kirschner152, Reference Ashworth153).

Fibrillin-1 is highly susceptible to proteolytic degradation, and increased proteolytic susceptibility appears to play a significant role in the pathogenesis of MFS (Refs Reference Robinson6, Reference Kirschner152, Reference Ashworth153). Murine models of MFS (e.g. C1039 G heterozygote; Ref. Reference Judge154) have revealed gross disruption to the elastic lamellar structure of the aorta (Fig. 4). Matrix metalloproteinases (MMPs) -1, -2, -3 and -9 that can cleave fibrillin-1 are upregulated in thoracic aortas from MFS patients and MMPs-2 and -9 are upregulated in an MFS mouse model (Ref. Reference Chung155). MMP-1 up-regulation has been associated with a fibrillin-1 fragment containing an elastin-binding protein recognition sequence (Ref. Reference Booms156). Recombinant fibrillin-1 fragments harbouring mutations associated with severe neonatal MFS showed enhanced susceptibility to physiological and non-physiological proteases. In addition, cathepsins K and V can cleave fibrillin-1 at multiple sites and thus may play a role in the pathogenesis of MFS (Ref. Reference Kirschner152).

Figure 4. Effects of the fibrillin-1(Fbn)C1039G/+ phenotype, and Smad4 haploinsufficiency (Smad4+/−), on the aortic architecture in mice. Verhoeff-Van Giesen staining revealed architectural abnormalities in the ascending aorta of these mutant mice. Compared with wild-type littermates, FbnC1039G/+ mice had medial thickening and elastic fibre fragmentation. These defects were enhanced in Smad4+/−: FbnC1039G/+ mice. Figure taken from Ref. Reference Holm111 (with permission from the corresponding author and the publisher).

TGFβ is known to control smooth muscle cell differentiation, matrix synthesis and vascular morphogenesis, and perturbed TGFβ signalling has been identified as a primary causative feature of MFS aortic disease progression (Refs Reference Dietz and Pagon109, Reference Jaffe157) (Fig. 4). Mouse studies have shed further light on the complex contribution of TGFβ to disease progression. For example, mice with a fibrillin-1 deletion showed increased TGFβ activity in lung sections compared with wild-type littermates, coupled with impairment in alveolar septation (Reference Neptune158). Increased TGFβ activity also contributed to defective mitral valvulogenesis in a murine model of MFS. Cultured vascular smooth muscle cells from thoracic aortas of fibrillin-1 null mice showed constitutively active Smad2/3 signalling (Ref. Reference Carta159). Non-canonical (non-Smad-mediated) TGFβ signalling also enhances aortic defects in Marfan mice (Ref. Reference Holm111) (Fig. 4).

Enhanced TGFβ activity is associated with other fibrillinopathies including Stiff skin syndrome (Ref. Reference Loeys116), and acromicric and GD that can be caused by fibrillin-1 or ADAMTSL2 mutations (Refs Reference Le Goff149, Reference Le Goff160). These diseases highlight the central role of fibrillin-1 microfibrils in regulating TGFβ bioavailability. Other MFS-like diseases with perturbed TGFβ include MFS2 caused by mutations in the TGFβ receptor II, and Loeys–Dietz syndrome (Refs Reference Dietz and Pagon109, Reference Loeys161), familial aortic aneurysm (Ref. Reference Pannu162) and Furlong syndrome (Ref. Reference Ades163), which are caused by mutations in TGFβ receptors I or II. The rare disorder arterial tortuosity syndrome, which causes arterial elastic fibre fragmentation, might result from altered TGFβ activity as a consequence of mutations in the gene encoding the glucose transporter GLUT10 (Ref. Reference Coucke164). Mutations in the TGFβ repressor SKI cause the Shprintzen–Goldberg syndrome, which has many overlapping features with MFS and Loeys–Dietz syndrome (Ref. Reference Doyle165).

MFS patients are routinely prescribed beta blockers to reduce haemodynamic stress on the aorta (Ref. Reference Dietz and Pagon109). In recent years, uncovering the link between increased TGFβ activity and MFS pathogenesis has led to the development of advanced therapeutic strategies that reduce TGFβ signalling. In particular, treatment with the angiotensin II type I receptor antagonist losartan was shown in MFS mice (Ref. Reference Habashi110) to prevent aortic aneurysm and to improve non-cardiovascular symptoms (such as alveolar septation and muscle repair) (Fig. 5). Several clinical trials have since been assessing the benefits of losartan treatment for MFS patients. In a clinical trial of patients with severe and rapidly progressive MFS, losartan treatment reduced both the rate of aortic root growth to 10% of pretreatment levels (Refs Reference Dietz and Pagon109, Reference Geirsson166). A multicentre losartan trial is in progress (Ref. Reference Brooke117), alongside a trial that studies the combined effects of β-blockers and losartan in MFS patients (Ref. Reference Detaint118). Angiotensin II-receptor blockers are also being assessed for their potential to modulate pathological progression of mitral valve prolapse in MFS patients (Ref. Reference Möberg119). Levels of circulating TGFβ are elevated in MFS mice and patients; thus circulating TGFβ may prove to be a valuable prognostic marker for MFS (Ref. Reference Dietz and Pagon109). Doxycycline has also been shown to delay aneurysm rupture through inhibition of MMPs-2 and -9 (Ref. Reference Xiong120). A recent study has also revealed that treatment of MFS mice with antagonists to the elastin-binding protein motif (GxxPG) can reduce several abnormalities characteristic for MFS, suggesting a potential new method of treatment for cardiovascular manifestations of MFS (Ref. Reference Guo121).

Figure 5. Prevention of aortic aneurysm in mice by losartan. Representative murine ascending aortae (arrowheads), after therapy, are: (a) wild-type mice; (b–f) mice heterozygous for Marfan-causing fibrillin-1 mutation C1039 G (FbnC1039G/+), treated with placebo (b), propanolol (c) or losartan (d–f). Scale bars = 4 mm. Figure taken from Ref. Reference Habashi110 (with permission from the corresponding author and the publisher).

WMS and acromicric and GD

WMS is characterised by short stature, brachydactyly (short digits), eye abnormalities and joint stiffness. Both autosomal (WMS2; OMIM 608328) and recessive (WMS1; OMIM 277600) forms of WMS exist (Ref. Reference Faivre115). Clinical manifestations of the acromicric (AD; OMIM 102370) and GD (OMIM 231050) include short stature, joint defects and thickened skin (Ref. Reference Le Goff160).

Mutations causing autosomal dominant WMS, AD and GD are located within the TB5 domain of fibrillin-1 (Refs Reference Marmorstein108, Reference Jaffe157). This WMS-associated mutation abolishes binding of the TB5 domain to heparin, whereas six tested mutations associated with AD and GD reduce binding (Ref. Reference Cain112). A second N-terminal fibrillin-1 mutation associated with dominant WMS results in an in-frame deletion, abolishing a specific binding site in fibrillin-1 for ADAMTSL-2, -3, -6 and the ADAMTSL papilin (Ref. Reference Sengle49). It was recently demonstrated that ADAMTS-10 can bind fibrillin-1 (Ref. Reference Kutz95), and recessive WMS is associated with ADAMTS-10 mutations (Ref. Reference Dagoneau9). Mutations in ADAMTS-17 and ADAMTSL-2 cause a Weill-Marchesani-like syndrome (WMS-like; OMIM 613195) and GD, respectively (Refs Reference Morales148, Reference Le Goff149). These closely related genetic disorders suggest a functional relationship between ADAMTSL (-2,-4), ADAMTS (-10,-17) and fibrillin-1. It has been hypothesised that ADAMTSL-3, ADAMTS-10 and fibrillin-1 might form a ternary complex and that these interactions may influence integrin interactions with fibrillin-1 because of the proximity of the fibrillin-1 binding sites to an RGD motif (Ref. Reference Sengle49). ADAMTS molecules are also implicated in TGFβ sequestration and activation (Ref. Reference Doyle, Gerber and Dietz114); although the underlying mechanism is unknown, ADAMTS molecules could influence stable deposition of LTBP-containing large latent TGFβ complexes with fibrillin microfibrils. In addition, a mutation in LTBP-2 has recently been associated with WMS (WMS3; OMIM 614819) (Ref. Reference Haji-Seyed-Javadi147).

Elastinopathies

Mutations in elastin and elastic fibre-associated proteins cause disease through loss or gain of function, and increased susceptibility to inflammatory or proteolytic damage of elastic fibres (Refs Reference Wagenseil and Mecham122, Reference Hu123).

Supravalvular aortic stenosis (SVAS; OMIM 185500) occurs in 1 in 20 000 live births, is inherited in an autosomal dominant manner but can also occur sporadically and is characterised by narrowing or obstruction of the ascending aorta. Other defects include obstruction of pulmonary, coronary, carotid and renal arteries. Severe SVAS may lead to dyspnoea, angina, systolic murmur, left ventricular hypertrophy ultimately leading to congestive heart failure. Although congenital non-syndromic forms of SVAS exist, SVAS is typically associated with Williams–Beuren syndrome (WBS), a complex developmental disorder (below).

The spectrum of heterozygous loss-of-function mutations in the elastin gene reflects diverse SVAS phenotypes (Ref. Reference Urban124). Point mutations, translocations and partial deletions of the elastin gene lead to premature stop codons and unstable mRNA. The most widely accepted underlying mechanism of SVAS is elastin haploinsufficiency (Ref. Reference Micale125), where smooth muscle cells of large vessels generate half normal levels of elastin, leading to thinner and disorganised elastic lamellae (Ref. Reference Brooke117). That ELN gene haploinsufficiency is an underlying cause of nonsyndromic SVAS was confirmed by the identification of seven novel elastin gene mutations in 31 SVAS patients with familial and sporadic non-syndromic SVAS (Ref. Reference Urban126). Apart from its structural role, elastin inhibits smooth muscle cell proliferation and promotes organisation of actin filament bundles. In SVAS, smooth muscle cells hyper-proliferate, become hypertrophic and collagen levels increase, leading to thicker, narrower and less flexible and resilient arteries.

Human induced pluripotent stem (iPS) cells from SVAS patients, which exhibit SVAS characteristics, have been generated to study pathogenesis and patient-specific interventions (Ref. Reference Ge127). They reveal that affected smooth muscle cells have fewer organised networks of smooth muscle alpha-actin filament bundles compared with control cells. They also predict therapeutic inhibition of smooth muscle cell overproliferation by decreasing ERK1/2 signalling. Another SVAS therapy by simple sliding aortoplasty has been reported (Ref. Reference Shin128).

Williams–Beuren Syndrome (WBS; OMIM 194050) is a rare multisystem developmental genetic disorder that has a prevalence of 1 in 7500 and usually occurs sporadically (Refs Reference Morris and Mervis129, Reference Pober, Johnson and Urban167). Characteristics associated with WBS include intellectual deficit, connective tissue defects, cardiovascular abnormalities (SVAS, also peripheral pulmonary aortic stenosis and arterial hypertension), dimorphic facial features, short stature and metabolic defects (infantile hypocalcaemia and abnormal glucose intolerance). Mitral valve disease is also associated with WBS, with a prevalence of approximately 40%.

WBS results from a hemizygous deletion (1.5–1.8 megabase), and less frequently duplication, of 26–28 genes, including elastin, that localise to the Williams syndrome region of the chromosome 7q11.23 (Ref. Reference Urban131). These deletions arise as a consequence of meiotic misalignment of repetitive sequences that flank the WBS chromosome region (Ref. Reference Schubert132). Deletions cause the phenotypes outlined above, whereas duplications typically result in milder phenotypes (Refs Reference Urban131, Reference Schubert132).

Deletion of one copy of elastin gene is the most common and most recognised genetic rearrangement known in WBS, as it occurs in over 96% of WBS patients, and elastin haploinsufficiency underlies the cardiovascular manifestations in WBS, most prominently SVAS. Smooth muscle cells of WBS patients produce reduced levels of elastin (Ref. Reference Urban124). Treatments for the vascular symptoms of WBS commonly involve reparative surgery of aortic stenosis and individualised hypertension treatment (Ref. Reference Pober133).

Pseudoxanthoma elasticum (PXE; OMIM 264800) is a heritable autosomal recessive disorder with a prevalence of approximately 1 in 25 000–100 000 (Refs Reference Chassaing134, Reference Uitto135). PXE is characterised by progressive ectopic mineralisation and fragmentation of elastic fibres that leads to ocular, cutaneous and cardiovascular abnormalities. PXE becomes initially evident when the accumulation of calcium/phosphate complexes in the skin causes the appearance of yellow papule and loss of elasticity. In the eyes, these complexes cause angioid streaks and neovascularisation, leading to gradual loss of vision. Early onset of arteriosclerosis and cardiac failure are the most serious complication. PXE is caused by loss-of-function mutations in the ABCC6 gene, which encodes a putative transmembrane transporter, especially abundant in the liver (Ref. Reference Bergen136). The dysfunctional transporter may affect levels or activity of circulatory factors, such as fetuin-A and matrix Gla-protein, responsible for prevention of aberrant mineralisation in normal conditions (Ref. Reference Uitto, Li and Jiang137). Therapies for the ocular symptoms of PXE focus on treating neovascularisation of the eye by intravitreal injections of vascular endothelial growth factor (VEGF) inhibitors (Ref. Reference Georgalas138). Other approaches such as supplementation of diet with magnesium, introduction of anti-mineralising factors, regeneration of the liver and stem-cell therapy are being assessed.

Cutis laxa (CL) comprises inherited (Ref. Reference Berk8) and acquired disorders that are characterised by abnormal elastic fibres and loose sagging skin that often gives an aged appearance. CL can be inherited in ADCL, ARCL or X-linked forms (XLCL). Based on different mutations and phenotypic manifestations, autosomal recessive types can be further divided into type I (A and B), II (A and B) and III (De Barsy syndrome), Urban-Rifkin-Davis syndrome, macrocephaly, alopaecia, CL and scoliosis (MACS) syndrome and arterial tortuosity syndrome. Recessive forms of CL tend to be more severe, often resulting in childhood mortality (Ref. Reference Berk8).

ARCL type I (OMIM 219100) is caused by mutations in the genes encoding fibulin-4 (subtype A) or fibulin-5 (subtype B) (Refs Reference Hucthagowder139, Reference Renard140, Reference Loeys141, Reference Claus142). Fibulin-4 mutations usually cause more severe and often lethal phenotypes with emphysema, tortuosity, aortic aneurysms and joint laxity. Fibulin-4 mutations were previously thought to be rare, but recently, in a small population, a novel fibulin-4 mutation has been identified in 22 infants suffering from severe arteriopathy syndrome (Ref. Reference Kappanayil168).

The two subtypes of ARCL type II (OMIM 219200) are both associated with loss-of-function mutation in the ATP6VOA2 gene, which encodes a vesicular proton pump, thus affecting vesicular trafficking. The mutation indirectly leads to tropoelastin accumulation within the Golgi and thereby significantly reduced deposition of mature elastin (Ref. Reference Hucthagowder143). Mutations in the PYCR1 gene, which encodes for a mitochondrial enzyme involved in proline biosynthesis (pyrroline-5-carboxylate reductase 1), have also been associated with this form of CL (Ref. Reference Reversade169).

The genetic cause of the De Barsy syndrome/CL-corneal clouding-mental retardation syndrome (OMIM 219150) is currently unknown; however, because of the significant overlap with the phenotypic characteristics of type II ARCL, mutations in ATPVOA2 and PYCR1 seem to be the likely underlying cause of this syndrome (Ref. Reference Leao-Teles170).

Urban-Rifkin-Davis syndrome (OMIM 613177) is a newly characterised type of ARCL that is caused by mutations in LTBP-4 (Ref. Reference Urban171). A severe phenotype including developmental, craniofacial and fatal pulmonary abnormalities is common. Haploinsufficiency of LTBP-4 is likely to cause increased TGFβ activity and abnormal elastic fibre assembly.

MACS syndrome (OMIM 613075) is a multitude malformation syndrome that includes macrocephaly, alopaecia, CL and scoliosis. It is caused by mutations in RIN2, which is a guanine exchange factor for Rab5, which in turn controls endocytic trafficking. It is unlikely that these mutations underlie the connective tissue abnormalities, which more probably are attributable to fibulin-5 deficiency, which MACS patients exhibit (Ref. Reference Basel-Vanagaite172).

Arterial tortuosity syndrome (OMIM 208050) is characterised by elongated and tortuous arteries, along with CL. Mutations in the SLC2A10 gene that encodes the glucose transporter GLUT10 are associated with this syndrome. Although previously thought that the mutation may lead to abnormal TGFβ activity, stimulating smooth muscle cell proliferation, it has also been suggested that the loss of GLUT10 rather results in aberrant collagen and elastin metabolism (Ref. Reference Segade173).

XLCL (OMIM 304150) and Menkes disease (OMIM 309400) are both caused by mutations in the ATP7A gene that encodes for a copper-transporting enzyme. Defects in this enzyme reduce functionality of copper-dependent enzymes such as LOX, affecting elastin and collagen deposition. Although both disease forms have overlapping phenotypic manifestations, XLCL tends to be milder. Severe and commonly lethal Menkes disease is associated with neurological defects. Although neonatal diagnosis and early treatment with copper can improve survival, some residual activity of ATP7A is essential (Ref. Reference Berk8). Recently, gene replacement therapy has been suggested as a therapeutic approach to alleviate Menkes disease (Ref. Reference Kaler130).

ADCL (OMIM 123700) is generally defined as a milder form of the inherited CL spectrum, typically characterised by aged facial appearance. Systemic manifestations are less common and may involve gastrointestinal abnormalities, hernias, pulmonary aortic stenosis, tortuosity, emphysema and aortic root dilatation (Ref. Reference Callewaert174). ADCL-causing mutations do not lead to elastin haploinsufficiency, in contrast to SVAS, but rather have a dominant-negative effect on elastin deposition. Indeed, mutant elastin has abnormal binding to fibrillin-1 and enhanced self-association of elastin, which caused decreased mature elastin deposition (Ref. Reference Callewaert174). Increased endoplasmic reticulum stress, induced apoptosis and increased TGFβ signalling are also reported (Ref. Reference Shifren and Mecham144). Mutations in ALDH18A1, encoding Δ1-pyrroline-5-carboxylate synthase (P5CS) have overlapping symptoms with ADCL, with altered elastic fibre ultrastructure (Ref. Reference Skidmore145).

Buschke–Ollendorff syndrome (OMIM 166700) is a rare autosomal dominant condition, which includes skin lesions containing elastic fibres, and osteopoikilosis, and is caused by mutations in the LEMD3 gene, which encodes a protein involved in bone and connective tissue morphogenesis (Ref. Reference Schena146). The encoded protein can antagonise TGFβ signalling at the inner nuclear membrane.

Acquired elastic fibre disorders

Acquired elastic fibre disorders arise wherever elastic fibre structure and function are compromised, including tissues such as skin, blood vessels and lungs. Some examples are highlighted below.

Acquired CL usually develops in adulthood. Characteristic secondary destruction of elastic fibres occurs because of dermal inflammation caused by other medical conditions or reactions to medicines. Systemic manifestations, which often lead to fatal outcomes, may include emphysema, hernias and vascular dilatation (Ref. Reference Hu175).

Marshall syndrome is a type of acquired CL typically affecting children after suffering neutrophilic dermatosis (Sweet's syndrome) (Ref. Reference Timmer176).

Solar/actinic elastosis is a result of a prolonged exposure to sunlight, leading to degeneration of the elastic tissue of the dermis and consequently to premature ageing of the skin. Deposition of the elastotic material and decreased collagen synthesis lead to rough, inelastic, wrinkled and hyperpigmented skin. Current therapy of choice is a skin-rejuvenating photodynamic therapy in which photosensitisers and diverse light sources have been successfully used to reverse the effects of photodamage (Ref. Reference Karrer177).

Acquired PXE is characterised by cutaneous mineralisation and fragmentation of elastic fibres leading to lax inelastic skin. Unlike the heritable PXE, the acquired PXE is non-systemic and is limited to the skin only. Exposure to chemicals such as calcium–ammonium–nitrate salts, mechanical stress and abnormal calcium–phosphate metabolism has been implicated in the development of this disease (Ref. Reference Lewis178). There are no generally accepted therapies for the acquired PXE. Papillary dermal elastolysis is a rare condition, similar to PXE, which usually affects elderly women, and is associated with elastolytic papules on the neck and supraclavicular sites (Ref. Reference Alves179).

Ageing changes to blood vessels and lungs: loss of elastic fibre integrity and elastic recoil are common features of ageing changes to blood vessels especially the elastic arteries, whereas pulmonary emphysema is associated with loss of elasticity and pathological alveolar remodelling (Ref. Reference Pasquali-Ronchetti and Baccarani-Contri180).

ARMD (OMIM 603075) is one of the most frequent causes of loss of vision and is characterised by progressive deposition of extracellular aggregates and neovascularisation in the macula region of the eye. A multitude of genes have been associated with this disease, with mutations in complement factor H being the most common cause of the disease (Ref. Reference Klein181). Mutations in fibulins, and especially fibulin-5, have also been implicated in ARMD. Fibulin-5 mutations lead to haploinsufficiency which may cause the observed phenotype by altering the structure of the elastic Bruch's membrane of the macula (Ref. Reference Lotery182). Anti-VEGF-A therapy has been successfully used to alleviate neovascularisation in the ARMD. A recent review has highlighted other possible therapeutic approaches, and stressed the importance of supporting the coping mechanisms of the retinal pigment cells of the macula (Ref. Reference Ambati and Fowler183).

Elastosis perforans serpiginosa (EPS; OMIM 130100) is a rare degenerative skin disease, which exhibits loss of elastic fibres in the upper dermis, following extended treatment with D-penicillamine that chelates copper and inhibits cross-linking of elastin by copper-dependent lysyl oxidase (Ref. Reference Atzori184).

Research in progress and outstanding research questions

Recent progress in treating MFS and related diseases with losartan and other therapies that target TGFβ activity have led to improved patient outcomes. However, further understanding of the biology of elastic fibres and their assembly is urgently needed to advance the therapeutic prospects for elastic fibre diseases. Current research on elastic fibres aims to define how cells and their integrin and heparan sulphate proteoglycans (HSPG) receptors direct the assembly of microfibrils and elastic fibres, how fibronectin and the ADAMTS and ADAMTSL molecules influence microfibril deposition and how pericellular processing (by furin/PACE enzymes) and cross-linking (by LOX and tissue transglutaminase enzymes) regulate elastic fibre deposition. New insights into how LTBPs are deposited in the matrix, and how active TGFβ and BMPs are released from these complexes are needed to resolve precisely how TGFβ growth factor bioavailability is controlled. All this information is critical for elucidating how defects in microfibrils and elastic fibres cause distinct genetic and acquired diseases by perturbing different stages in their assembly, their structural integrity or their susceptibity to proteolytic attack. We also need to understand much better how inflammatory enzymes cause elastic fibre remodelling and loss of both microfibril and elastic fibre functional integrity in disease and ageing. There is a need to identify biomarkers of elastic fibre damage to enable early diagnosis and treatment. Much recent progress has been made in the engineering of vascular constructs based on elastin and elastic fibre components, and this approach offers great promise for the repair of damaged elastic tissues. Such advances will provide mechanistic targets to prevent elastic fibre degeneration and to encourage de novo elastic fibre formation in regenerating tissues.

Acknowledgements and funding

Current research on microfibrils and elastic fibres in our laboratory is funded by the Medical Research Council (UK) and Wellcome Trust.

References

References

1Kielty, C.M. (2006) Elastic fibres in health and disease. Expert Reviews in Molecular Medicine 8, 1-23CrossRefGoogle ScholarPubMed
2Nistala, H. et al. (2010) Fibrillin-1 and -2 differentially modulate endogenous TGFβ and BMP bioavailability during bone formation. Journal of Cell Biology 190, 1107-1121CrossRefGoogle ScholarPubMed
3Özbek, S. et al. (2010) The evolution of extracellular matrix. Molecular Biology of the Cell 21, 4300-4305Google Scholar
4Piha-Gossack, A., Sossin, W. and Reinhardt, D.P. (2012) The evolution of extracellular fibrillins and their functional domains. PLoS ONE 7, e33560CrossRefGoogle ScholarPubMed
5Faury, G. (2001) Function-structure relationship of elastic arteries in evolution: from microfibrils to elastin and elastic fibres. Pathologie Biologie (Paris) 49, 310-325CrossRefGoogle ScholarPubMed
6Robinson, P.N. et al. (2006) The molecular genetics of Marfan syndrome and related disorders. Journal of Medical Genetics 43, 769-787CrossRefGoogle ScholarPubMed
7Sugitani, H. et al. (2012) Alternative splicing and tissue-specific elastin misassembly act as biological modifiers of human elastin gene frameshift mutations associated with dominant CL. Journal of Biological Chemistry 287, 22055-22067CrossRefGoogle Scholar
8Berk, D.R. et al. (2012) CL: a review. Journal of the American Academy of Dermatology 66, 842.e1-17Google Scholar
9Dagoneau, N. et al. (2004) ADAMTS10 mutations in autosomal recessive Weill-Marchesani syndrome. American Journal of Human Genetics 75, 801-806CrossRefGoogle ScholarPubMed
10Dyksterhuis, L.B. and Weiss, A.S. (2010) Homology models for domains 21-23 of human tropoelastin shed light on lysine crosslinking. Biochemical and Biophysical Research Communications 396, 870-873CrossRefGoogle ScholarPubMed
11Baldock, C. et al. (2011) Shape of tropoelastin, the highly extensible protein that controls human tissue elasticity. Proceedings of the National Academy of Sciences USA 108, 4322-4327CrossRefGoogle ScholarPubMed
12Bax, D.V. et al. (2009) Cell adhesion to tropoelastin is mediated via the C-terminal GRKRK motif and integrin alphaVbeta3. Journal of Biological Chemistry 284, 28616-28623Google Scholar
13Dyksterhuis, L.B. et al. (2007) Domains 17-27 of tropoelastin contain key regions of contact for coacervation and contain an unusual turn-containing crosslinking domain. Matrix Biology 26, 125-135Google Scholar
14Yeo, G.C. et al. (2012) Tropoelastin bridge region positions the cell-interactive C terminus and contributes to elastic fiber assembly. Proceedings of the National Academy of Sciences USA 109, 2878-2883CrossRefGoogle ScholarPubMed
15Hubmacher, D., Tiedemann, K. and Reinhardt, D.P. (2006) Fibrillins: from biogenesis of microfibrils to signaling functions. Current Topics in Developmental Biology 75, 93-123Google Scholar
16Ramirez, F. and Sakai, L.Y. (2010) Biogenesis and function of fibrillin assemblies. Cell and Tissue Research 339, 71-82Google Scholar
17Charbonneau, N.L. et al. (2003) Fibrillins can co-assemble in fibrils, but fibrillin fibril composition displays cell-specific differences. Journal of Biological Chemistry 278, 2740-2749Google Scholar
18Zhang, H. et al. (1994) Structure and expression of fibrillin-2, a novel microfibrillar component preferentially located in elastic matrices. Journal of Cell Biology 124, 855-863CrossRefGoogle ScholarPubMed
19Corson, G.M. et al. (2004) Differential expression of fibrillin-3 adds to microfibril variety in human and avian, but not rodent, connective tissues. Genomics 83, 461-472Google Scholar
20Sabatier, L. et al. (2011) Fibrillin-3 expression in human development. Matrix Biology 30, 43-52Google Scholar
21Charbonneau, N.L. et al. (2010) Microfibril structure masks fibrillin-2 in postnatal tissues. Journal of Biological Chemistry 285, 20242-20251Google Scholar
22Brinckmann, J. et al. (2010) Enhanced fibrillin-2 expression is a general feature of wound healing and sclerosis: potential alteration of cell attachment and storage of TGF-beta. Laboratory Investigation 90, 739-752Google Scholar
23Carta, L. et al. (2006) Fibrillins 1 and 2 perform partially overlapping functions during aortic development. Journal of Biological Chemistry 281, 8016-8023Google Scholar
24Craft, C.S. et al. (2010) Microfibril-associated glycoprotein-1, an extracellular matrix regulator of bone remodeling. Journal of Biological Chemistry 285, 23858-23867Google Scholar
25Jensen, S.A. et al. (2001) Protein interaction studies of MAGP-1 with tropoelastin and fibrillin-1. Journal of Biological Chemistry 276, 39661-39666CrossRefGoogle ScholarPubMed
26Zilberberg, L. et al. (2012) Specificity of latent TGF-ß binding protein (LTBP) incorporation into matrix: role of fibrillins and FN. Journal of Cellular Physiology 227, 3828-3836Google Scholar
27Lemaire, R. et al. (2007) Microfibril-associated MAGP-2 stimulates elastic fiber assembly. Journal of Biological Chemistry 282, 800-808CrossRefGoogle ScholarPubMed
28Massam-Wu, T. et al. (2010) Assembly of fibrillin microfibrils governs extracellular deposition of latent TGF beta. Journal of Cell Science 123, 3006-3018Google Scholar
29Koli, K. et al. (2005) Sequential deposition of latent TGF-beta binding proteins (LTBPs) during formation of the extracellular matrix in human lung fibroblasts. Experimental Cell Research 310, 370-382Google Scholar
30Gibson, M.A. et al. (1995) Bovine latent transforming growth factor beta 1-binding protein 2: molecular cloning, identification of tissue isoforms, and immunolocalization to elastin-associated microfibrils. Molecular Cell Biology 15, 6932-6942Google Scholar
31Baccarani-Contri, M. et al. (1990) Immunocytochemical localization of proteoglycans within normal elastin fibers. European Journal of Cell Biology 53, 305-312Google Scholar
32Trask, B.C. et al. (2000) The microfibrillar proteins MAGP-1 and fibrillin-1 form a ternary complex with the chondroitin sulfate proteoglycan decorin. Biology of the Cell 11, 1499-1507Google Scholar
33Reinboth, B. et al. (2002) Molecular interactions of biglycan and decorin with elastic fiber components: biglycan forms a ternary complex with tropoelastin and microfibril-associated glycoprotein 1. Journal of Biological Chemistry 277, 3950-3957Google Scholar
34Isogai, Z. et al. (2002) Versican interacts with fibrillin-1 and links extracellular microfibrils to other connective tissue networks. Journal of Biological Chemistry 277, 4565-4572Google Scholar
35Tiedemann, K. et al. (2005) Microfibrils at basement membrane zones interact with perlecan via fibrillin-1. Journal of Biological Chemistry 280, 11404-11412Google Scholar
36Hirano, E. et al. (2002) Expression of 36-kDa microfibril-associated glycoprotein (MAGP-36) in human keratinocytes and its localization in skin. Journal of Dermatological Science 28, 60-67CrossRefGoogle ScholarPubMed
37Gibson, M.A., Kumaratilake, J.S. and Cleary, E.G. (1997) Immunohistochemical and ultrastructural localization of MP78/70 (betaig-h3) in extracellular matrix of developing and mature bovine tissues. Journal of Histochemistry Cytochemistry 45, 1683-1696Google Scholar
38Hayashi, K. et al. (2004) Comparative immunocytochemical localization of lysyl oxidase (LOX) and the lysyl oxidase-like (LOXL) proteins: changes in the expression of LOXL during development and growth of mouse tissues. Journal of Molecular Histology 35, 845-55Google Scholar
39Yanagisawa, H. and Davis, E.C. (2010) Unraveling the mechanism of elastic fiber assembly: the roles of short fibulins. International Journal of Biochemical and Cell Biology 42, 1084-1093Google Scholar
40McLaughlin, P.J. et al. (2007) Lack of fibulin-3 causes early aging and herniation, but not macular degeneration in mice. Human Molecular Genetics 16, 3059-3070Google Scholar
41Kobayashi, N. et al. (2007) A comparative analysis of the fibulin protein family. Biochemical characterization, binding interactions, and tissue localization. Journal of Biological Chemistry 282, 11805-11816CrossRefGoogle ScholarPubMed
42Choudhury, R. et al. (2009) Differential regulation of elastic fiber formation by fibulin-4 and -5. Journal of Biological Chemistry 284, 24553-24567CrossRefGoogle ScholarPubMed
43McLaughlin, P.J. et al. (2006) Targeted disruption of fibulin-4 abolishes elastogenesis and causes perinatal lethality in mice. Molecular and Cell Biology 26, 1700-1709CrossRefGoogle ScholarPubMed
44Horiguchi, M. et al. (2009) Fibulin-4 conducts proper elastogenesis via interaction with cross-linking enzyme lysyl oxidase. Proceedings of the National Academy of Sciences USA 106, 19029-19034Google Scholar
45Yanagisawa, H. et al. (2002) Fibulin-5 is an elastin-binding protein essential for elastic fibre development in vivo. Nature 415, 168-171Google Scholar
46Nakamura, T. et al. (2002) Fibulin-5/DANCE is essential for elastogenesis in vivo. Nature 415, 171-175Google Scholar
47Hirai, M. et al. (2007) Fibulin-5/DANCE has an elastogenic organizer activity that is abrogated by proteolytic cleavage in vivo. Journal of Cell Biology 176, 1061-1071Google Scholar
48Lomas, A.C. et al. (2007) Fibulin-5 binds human smooth-muscle cells through alpha5beta1 and alpha4beta1 integrins, but does not support receptor activation. Biochemical Journal 405, 417-428Google Scholar
49Sengle, G. et al. (2012) Microenvironmental regulation by fibrillin-1. PLoS Genetics 8, e1002425Google Scholar
50Gabriel, L.A. et al. (2012) ADAMTSL4, a secreted glycoprotein widely distributed in the eye, binds fibrillin-1 microfibrils and accelerates microfibril biogenesis. Investigative Ophthalmology and Vision Sciences 53, 461-469CrossRefGoogle ScholarPubMed
51Bader, H.L. et al. (2012) A disintegrin-like and metalloprotease domain containing thrombospondin type 1 motif-like 5 (ADAMTSL5) is a novel fibrillin-1-, fibrillin-2-, and heparin-binding member of the ADAMTS superfamily containing a netrin-like module. Matrix Biology 31, 398-411CrossRefGoogle ScholarPubMed
52Tsutsui, K. et al. (2010) ADAMTSL-6 is a novel extracellular matrix protein that binds to fibrillin-1 and promotes fibrillin-1 fibril formation. Journal of Biological Chemistry 285, 4870-4882CrossRefGoogle ScholarPubMed
53Privitera, S. et al. (1998) The 67-kDa enzymatically inactive alternatively spliced variant of beta-galactosidase is identical to the elastin/laminin-binding protein. Journal of Biological Chemistry 273, 6319-6326CrossRefGoogle Scholar
54Todorovic, V. et al. (2005) Latent TGF-beta binding proteins. International Journal of Biochemistry and Cell Biology 37, 38-41Google Scholar
55Gleizes, P.E. et al. (1996) Identification and characterization of an eight-cysteine repeat of the latent transforming growth factor-beta binding protein-1 that mediates bonding to the latent transforming growth factor-beta1. Journal of Biological Chemistry 271, 29891-29896Google Scholar
56Saharinen, J., Taipale, J. and Keski-Oja, J. (1996) Association of the small latent transforming growth factor-beta with an eight cysteine repeat of its binding protein LTBP-1. EMBO Journal 15, 245-253CrossRefGoogle ScholarPubMed
57Isogai, Z. et al. (2003) Latent transforming growth factor beta-binding protein 1 interacts with fibrillin and is a microfibril-associated protein. Journal of Biological Chemistry 278, 2750-2757CrossRefGoogle ScholarPubMed
58Apte, S.S. (2009) A disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin type 1 motif (ADAMTS) superfamily: functions and mechanisms. Journal of Biological Chemistry 284, 31493-31497Google Scholar
59Gibson, M.A., Kumaratilake, J.S. and Cleary, E.G. (1989) The protein components of the 12-nanometer microfibrils of elastic and nonelastic tissues. Journal of Biological Chemistry 264, 4590-4598Google Scholar
60Cain, S.A. et al. (2006) Proteomic analysis of fibrillin-rich microfibrils. Proteomics 6, 111-122Google Scholar
61Weinbaum, J.S. et al. (2008) Deficiency in microfibril-associated glycoprotein-1 leads to complex phenotypes in multiple organ systems. Journal of Biological Chemistry 283, 25533-25543Google Scholar
62Gibson, M.A. et al. (1998) Microfibril-associated glycoprotein-2 (MAGP-2) is specifically associated with fibrillin-containing microfibrils but exhibits more restricted patterns of tissue localization and developmental expression than its structural relative MAGP-1. Journal of Histochemistry and Cytochemistry 46, 871-886Google Scholar
63Penner, A.S. et al. (2002) Microfibril-associated glycoprotein-2 interacts with fibrillin-1 and fibrillin-2 suggesting a role for MAGP-2 in elastic fiber assembly. Journal of Biological Chemistry 277, 35044-35049Google Scholar
64Hanssen, E. et al. (2004) MAGP-2 has multiple binding regions on fibrillins and has covalent periodic association with fibrillin-containing microfibrils. Journal of Biological Chemistry 279, 29185-29194CrossRefGoogle ScholarPubMed
65Molnar, J. et al. (2003) Structural and functional diversity of lysyl oxidase and the LOX-like proteins. Biochimica Biophysica Acta 1647, 220-224Google Scholar
66Marson, A. et al. (2005) Homotypic fibrillin-1 interactions in microfibril assembly. Journal of Biological Chemistry 280, 5013-5021Google Scholar
67Cain, S.A. et al. (2008) HS regulates fibrillin-1 N- and C-terminal interactions. Journal of Biological Chemistry 283, 27017-27027Google Scholar
68Hubmacher, D. et al. (2008) Biogenesis of extracellular microfibrils: multimerization of the fibrillin-1 C terminus into bead-like structures enables self-assembly. Proceedings of the National Academy of Sciences USA 105, 6548-6553Google Scholar
69Wagenseil, J.E. and Mecham, R.P. (2007) New insights into elastic fiber assembly. Birth Defects Research C Embryo Today 81, 229-240Google Scholar
70Kinsey, R. et al. (2008) Fibrillin-1 microfibril deposition is dependent on FN assembly. Journal of Cell Science 121, 2696-2704Google Scholar
71Bax, D.V. et al. (2007) Cell adhesion to fibrillin-1: identification of an Arg-Gly-Asp-dependent synergy region and a heparin-binding site that regulates focal adhesion formation. Journal of Cell Science 120, 1383-1392Google Scholar
72Jovanovic, J. et al. (2007) AlphaVbeta6 is a novel receptor for human fibrillin-1. Comparative studies of molecular determinants underlying integrin-rgd affinity and specificity. Journal of Biological Chemistry 282, 6743-6751Google Scholar
73Reinhardt, D.P. et al. (1996) Fibrillin-1: organization in microfibrils and structural properties. Journal of Molecular Biology 258, 104-116CrossRefGoogle ScholarPubMed
74Baldock, C. et al. (2001) The supramolecular organization of fibrillin-rich microfibrils. Journal of Cell Biology 152, 1045-1056Google Scholar
75Wang, M.C., Lu, Y. and Baldock, C. (2009) Fibrillin microfibrils: a key role for the interbead region in elasticity. Journal of Molecular Biology 388, 168-179Google Scholar
76Qian, R.Q. and Glanville, R.W. (1997) Alignment of fibrillin molecules in elastic microfibrils is defined by transglutaminase-derived cross-links. Biochemistry 36, 15841-15817Google Scholar
77Baldock, C. et al. (2006) Nanostructure of fibrillin-1 reveals compact conformation of EGF arrays and mechanism for extensibility. Proceedings of the National Academy of Sciences USA 103, 11922-11927Google Scholar
78Jensen, S.A., Robertson, I.B. and Handford, P.A. (2012) Dissecting the fibrillin microfibril: structural insights into organization and function. Structure 20, 215-225Google Scholar
79Sabatier, L. et al. (2009) Fibrillin assembly requires fibronectin. Molecular Biology of the Cell 20, 846-858Google Scholar
80Tucker, R.P. and Chiquet-Ehrismann, R. (2009) Evidence for the evolution of tenascin and FN early in the chordate lineage. International Journal of Biochemistry and Cell Biology 41, 424-434Google Scholar
81Hubmacher, D. et al. (2011) Homocysteine modifies structural and functional properties of FN and interferes with the FN-fibrillin-1 interaction. Biochemistry 50, 5322-5332Google Scholar
82Reber-Müller, S. et al. (1995) An extracellular matrix protein of jellyfish homologous to mammalian fibrillins forms different fibrils depending on the life stage of the animal. Developmental Biology 169, 662-672Google Scholar
83Dallas, S.L. et al. (2005) FN regulates latent transforming growth factor-beta (TGF beta) by controlling matrix assembly of latent TGF beta-binding protein-1. Journal of Biological Chemistry 280, 18871-18880Google Scholar
84Tiedemann, K. et al. (2001) Interactions of fibrillin-1 with heparin/heparan sulfate, implications for microfibrillar assembly. Journal of Biological Chemistry 276, 36035-36042CrossRefGoogle ScholarPubMed
85Ritty, T.M. et al. (2003) Fibrillin-1 and -2 contain heparin-binding sites important for matrix deposition and that support cell attachment. Biochemical Journal 375, 425-432Google Scholar
86Cain, S.A. et al. (2005) Fibrillin-1 interactions with heparin. Implications for microfibril and elastic fiber assembly. Journal of Biological Chemistry 280, 30526-30537Google Scholar
87Akhtar, K. et al. (2011) Oxidative modifications of the C-terminal domain of tropoelastin prevent cell binding. Journal of Biological Chemistry 286, 13574-13582Google Scholar
88Gheduzzi, D. et al. (2005) Heparan sulphate interacts with tropoelastin, with some tropoelastin peptides and is present in human dermis elastic fibers. Matrix Biology 24, 15-25CrossRefGoogle ScholarPubMed
89Ono, R.N. et al. (2009) Latent transforming growth factor beta-binding proteins and fibulins compete for fibrillin-1 and exhibit exquisite specificities in binding sites. Journal of Biological Chemistry 284, 16872-16881Google Scholar
90Kantola, A.K. et al. (2008) Fibronectin and heparin binding domains of latent TGF-beta binding protein (LTBP)-4 mediate matrix targeting and cell adhesion. Experimental Cell Research 314, 2488-2500Google Scholar
91Vehvilainen, P. et al. (2009) Matrix association of latent TGF-beta binding protein-2 (LTBP-2) is dependent on fibrillin-1. Journal of Cellular Physiology 221, 586-593Google Scholar
92Hirani, R. et al. (2007) LTBP-2 specifically interacts with the amino-terminal region of fibrillin-1 and competes with LTBP-1 for binding to this microfibrillar protein. Matrix Biology 26, 213-223Google Scholar
93Hubmacher, D. and Apte, S.S. (2011) Genetic and functional linkage between ADAMTS superfamily proteins and fibrillin-1: a novel mechanism influencing microfibril assembly and function. Cellular and Molecular Life Science 68, 3137-3148Google Scholar
94Saito, M. et al. (2011) ADAMTSL6β protein rescues fibrillin-1 microfibril disorder in a Marfan syndrome mouse model through the promotion of fibrillin-1 assembly. Journal of Biological Chemistry 286, 38602-38613Google Scholar
95Kutz, W.E. et al. (2011) ADAMTS10 protein interacts with fibrillin-1 and promotes its deposition in extracellular matrix of cultured fibroblasts. Journal of Biological Chemistry 286, 17156-17167Google Scholar
96Rock, M.J. et al. (2004) Molecular basis of elastic fibre formation. Critical interactions and a tropoelastin-fibrillin-1 cross-link. Journal of Biological Chemistry 279, 23748-23758CrossRefGoogle Scholar
97Segade, F. et al. (2007) The intracellular form of human MAGP1 elicits a complex and specific transcriptional response. International Journal of Biochemistry and Cell Biology 39, 2303-2313Google Scholar
98Werneck, C.C. et al. (2008) Mice lacking the extracellular matrix protein MAGP1 display delayed thrombotic occlusion following vessel injury. Blood 111, 4137-4144Google Scholar
99Yeo, G.C., Keeley, F.W. and Weiss, A.S. (2011) Coacervation of tropoelastin. Advances in Colloid Interface Science 167, 94-103Google Scholar
100Cirulis, J.T. and Keeley, F.W. (2010) Kinetics and morphology of self-assembly of an elastin-like polypeptide based on the alternating domain arrangement of human tropoelastin. Biochemistry 49, 5726-5733Google Scholar
101Choi, J. et al. (2009) Analysis of dermal elastic fibres in the absence of fibulin-5 reveals potential roles for fibulin-5 in elastic fibre assembly. Matrix Biology 28, 211-220CrossRefGoogle Scholar
102Czirok, A. et al. (2006) Elastic fiber macro-assembly is a hierarchical, cell motion-mediated process. Journal of Cellular Physiology 207, 97-106Google Scholar
103Sato, F. et al. (2007) Distinct steps of cross-linking, self-association, and maturation of tropoelastin are necessary for elastic fiber formation. Journal of Molecular Biology 369, 841-851Google Scholar
104Broekelmann, T.J. et al. (2005) Tropoelastin interacts with cell-surface glycosaminoglycans via its COOH-terminal domain. Journal of Biologcal Chemistry 280, 40939-40947CrossRefGoogle ScholarPubMed
105El-Hallous, E. et al. (2007) Fibrillin-1 interactions with fibulins depend on the first hybrid domain and provide an adaptor function to tropoelastin. Journal of Biological Chemistry 282, 8935-8946Google Scholar
106Zheng, Q. et al. (2007) Molecular analysis of fibulin-5 function during de novo synthesis of elastic fibres. Molecular and Cellular Biology 27, 1083-1095Google Scholar
107Michaelides, M. et al. (2006) Maculopathy due to the R345W substitution in fibulin-3: distinct clinical features, disease variability, and extent of retinal dysfunction. Investigative Ophthalmology and Vision Sciences 47, 3085-3097Google Scholar
108Marmorstein, L.Y. et al. (2010) Formation and progression of sub-retinal pigment epithelium deposits in Efemp1 mutation knock-in mice: a model for the early pathogenic course of macular degeneration. Human Molecular Genetics 16, 2423-2432Google Scholar
109Dietz, H.C. (2001, updated 2011) Marfan síndrome. Gene Reviews (ed. Pagon, R.A. et al. ) NCBI Bookshelf ID NBK1335 (PMID:20301510) Seattle WA.Google Scholar
110Habashi, J.P. et al. (2006) Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse model of Marfan syndrome. Science 312, 117-121Google Scholar
111Holm, T.M. et al. (2011) Noncanonical TGFβ signaling contributes to aortic aneurysm progression in Marfan syndrome mice. Science 332, 358-361CrossRefGoogle ScholarPubMed
112Cain, S.A. et al. (2012) Fibrillin-1 mutations causing Weill-Marchesani syndrome and acromicric and geleophysic dysplasias disrupt heparan sulfate interactions. PLoS ONE, 7, e48634Google Scholar
113Jordan, C.D. et al. (2010) Fibrillins in adult human ovary and polycystic ovary syndrome: is fibrillin-3 affected in PCOS? Journal of Histochemistry and Cytochemistry 58, 903-915Google Scholar
114Doyle, J.J., Gerber, E.E. and Dietz, H.C. (2012) Matrix-dependent perturbation of TGFβ signaling and disease. FEBS Letters 586, 2003-2015CrossRefGoogle ScholarPubMed
115Faivre, L. et al. (2003) In frame fibrillin-1 gene deletion in autosomal dominant Weill-Marchesani syndrome. Journal of Medical Genetics 40, 34-36Google Scholar
116Loeys, B.L. et al. (2010) Mutations in fibrillin-1 cause congenital scleroderma: stiff skin syndrome. Science Translational Medicine 2, 23ra20Google Scholar
117Brooke, B.S. et al. (2008) Angiotensin II blockade and aortic-root dilation in Marfan's syndrome. New England Journal of Medicine. 358, 2878-2795Google Scholar
118Detaint, D. et al. (2010) Rationale and design of a randomized clinical trial (Marfan Sartan) of angiotensin II receptor blocker therapy versus placebo in individuals with Marfan syndrome. Archives of Cardiovascular Diseases 103, 317-325Google Scholar
119Möberg, K. et al. (2010) The Ghent Marfan trial – a randomised, double-blind placebo controlled trial with losartan in Marfan patients treated with β-blockers. International Jounrla of Cardiology 157, 354-358Google Scholar
120Xiong, W. et al. (2008) Doxycycline delays aneurysm rupture in a mouse model of Marfan syndrome. Journal of Vascular Surgery 47, 166-172Google Scholar
121Guo, G. et al. (2012) Antagonism of GxxPG fragments ameliorates manifestations of aortic disease in Marfan syndrome mice. Human Molecular Genetics 22, 433-443Google Scholar
122Wagenseil, J.E. and Mecham, R.P. (2012) Elastin in large artery stiffness and hypertension. Journal of Cardiovascular Translational Research 5, 264-273Google Scholar
123Hu, Q. et al. (2006) Inflammatory destruction of elastic fibres in acquired CL is associated with missense alleles in the elastin and fibulin-5 genes. Journal of Investigative Dermatology 126, 283-290Google Scholar
124Urban, Z. et al. (2001) Supravalvular aortic stenosis: genetic and molecular dissection of a complex mutation in the elastin gene. Human Genetics 109, 512-520Google Scholar
125Micale, L. et al. (2010) Identification and characterization of seven novel mutations of elastin gene in a cohort of patients affected by supravalvular aortic stenosis. European Journal of Human Genetics 18, 317-323Google Scholar
126Urban, Z. et al. (2002) Connection between elastin haploinsufficiency and increased cell proliferation in patients with supravalvular aortic stenosis and Williams-Beuren syndrome. American Journal of Human Genetics 71, 30-44CrossRefGoogle ScholarPubMed
127Ge, X. et al. (2012) Modeling supravalvular aortic stenosis syndrome with human induced pluripotent stem cells. Circulation 126, 1695-1704Google Scholar
128Shin, H.J. et al. (2011) Modified simple sliding aortoplasty for preserving the sinotubular junction without using foreign material for congenital supravalvar aortic stenosis. European Journal of Cardio-thoracic Surgery 40, 598-602Google Scholar
129Morris, C.A. and Mervis, C.B. (2000) Williams syndrome and related disorders. Annual Reviews in Genomics and Human Genetics 1, 461-484Google Scholar
130Kaler, S.G. (2011) ATP7A-related copper transport diseases-emerging concepts and future trends. Nature Reviews in Neurology 7, 15-29Google Scholar
131Urban, Z. et al. (2000) Elastin gene deletions in Williams syndrome patients result in altered deposition of elastic fibres in skin and a subclinical dermal phenotype. Pediatric Dermatology 17, 12-20Google Scholar
132Schubert, C. (2009) The genomic basis of the Williams-Beuren syndrome. Cell and Molecular Life Sciences 66, 1178-1197Google Scholar
133Pober, B.R. (2010) Williams-Beuren syndrome. New England Journal of Medicine 362, 239-252Google Scholar
134Chassaing, N. et al. (2005) Pseudoxanthoma elasticum: a clinical, pathophysiological and genetic update including 11 novel ABCC6 mutations. Journal of Medical Genetics 42, 881-892Google Scholar
135Uitto, J. et al. (2011) Pseudoxanthoma elasticum: progress in diagnostics and research towards treatment: summary of the 2010 PXE International Research Meeting. American Journal of Medical Genetics A 7, 10Google Scholar
136Bergen, A.A. et al. (2000) Mutations in ABCC6 cause pseudoxanthoma elasticum. Nature Genetics 25, 228-231Google Scholar
137Uitto, J., Li, Q., and Jiang, Q. (2010) Pseudoxanthoma elasticum: molecular genetics and putative pathomechanisms. Journal of Investigative Dermatology 130, 661-670Google Scholar
138Georgalas, I. et al. (2011) Pseudoxanthoma elasticum, ocular manifestations, complications and treatment. Clinical and Experimental Optometry 94, 169-180Google Scholar
139Hucthagowder, V. et al. (2006) Fibulin-4: a novel gene for an autosomal recessive CL syndrome. American Journal of Human Genetics 78, 1075-1080Google Scholar
140Renard, M. et al. (2010) Altered TGFβ signalling and cardiovascular manifestations in patients with autosomal recessive CL type I caused by fibulin-4 deficiency. European Journal of Human Genetics 18, 895-901Google Scholar
141Loeys, B. et al. (2002) Homozygosity for a missense mutation in fibulin-5 (FBLN5) results in a severe form of CL. Human Molecular Genetics 11, 2113-2118Google Scholar
142Claus, S. et al. (2008) A p.C217R mutation in fibulin-5 from CL patients is associated with incomplete extracellular matrix formation in a skin equivalent model. Journal of Investigative Dermatology 128, 1442-1450Google Scholar
143Hucthagowder, V. et al. (2009) Loss-of-function mutations in ATP6V0A2 impair vesicular trafficking, tropoelastin secretion and cell survival. Human Molecular Genetics 18, 2149-2165Google Scholar
144Shifren, A. and Mecham, R.P. (2006) The stumbling block in lung repair of emphysema: elastic fiber assembly. Proceedings of the American Thoracic Society 3, 428-433Google Scholar
145Skidmore, D.L. et al. (2011) Further expansion of the phenotypic spectrum associated with mutations in ALDH18A1, encoding Δ1-pyrroline-5-carboxylate synthase (P5CS). American Journal of Medical Genetics A. 155A, 1848-1856Google Scholar
146Schena, D. et al. (2008) Buschke-Ollendorff syndrome. International Journal of Dermatology 47, 1159-1161Google Scholar
147Haji-Seyed-Javadi, R. et al. (2012) LTBP2 mutations cause Weill-Marchesani and Weill-Marchesani-like syndrome and affect disruptions in the extracellular matrix. Human Mutation 33, 1182-1187Google Scholar
148Morales, J. et al. (2009) Homozygous mutations in ADAMTS10 and ADAMTS17 cause lenticular myopia, ectopia lentis, glaucoma, spherophakia, and short stature. American Journal of Human Genetics 85, 558-568Google Scholar
149Le Goff, C. et al. (2008) ADAMTSL2 mutations in geleophysic dysplasia demonstrate a role for ADAMTS-like proteins in TGF-beta bioavailability regulation. Nature Genetics 40, 1119-1123Google Scholar
150Mellody, K. et al. (2006) Marfan syndrome-causing mutations in fibrillin-1 result in gross morphological alterations and highlight the structural importance of the second hybrid domain. Journal of Biological Chemistry 281, 31854-31862Google Scholar
151Charbonneau, N.L. et al. (2010) In vivo studies of mutant fibrillin-1 microfibrils. Journal of Biological Chemistry 285, 24943-24955Google Scholar
152Kirschner, R. et al. (2011) Classical and neonatal Marfan syndrome mutations in fibrillin-1 cause differential protease susceptibilities and protein function. Journal of Biological Chemistry 286, 32810-32823Google Scholar
153Ashworth, J.L. et al. (1999) Fibrillin degradation by matrix metalloproteinases: implications for connective tissue remodelling. Biochemical Journal 340, 171-181Google Scholar
154Judge, D.P. et al. (2004) Evidence for a critical contribution of haploinsufficiency in the complex pathogenesis of Marfan syndrome. Journal of Clinical Investigation 114, 172-181Google Scholar
155Chung, A.W. et al. (2007) Loss of elastic fiber integrity and reduction of vascular smooth muscle contraction resulting from the upregulated activities of matrix metalloproteinase-2 and -9 in the thoracic aortic aneurysm in Marfan syndrome. Circulation Research 101, 512-522Google Scholar
156Booms, P. et al. (2006) A fibrillin-1-fragment containing the elastin-binding-protein GxxPG consensus sequence upregulates matrix metalloproteinase-1: biochemical and computational analysis. Journal of Molecular and Cellular Cardiology 40, 234-246Google Scholar
157Jaffe, M. et al. (2012) Transforming growth factor-beta signaling in myogenic cells regulates vascular morphogenesis, differentiation, and matrix synthesis. Arteriosclerosis, Thrombosis and Vascular Biology 32, e1-11Google Scholar
158Neptune, E. et al. (2003) Dysregulation of TGF-beta activation contributes to pathogenesis in Marfan syndrome. Nature Genetics 33, 407-411Google Scholar
159Carta, L. et al. (2009) p38 MAPK is an early determinant of promiscuous Smad2/3 signaling in the aortas of fibrillin-1 (Fbn1)-null mice. Journal of Biological Chemistry 284, 5630-5636Google Scholar
160Le Goff, C. et al. (2011) Mutations in the TGFbeta binding-protein-like domain 5 of FBN1 are responsible for acromicric and geleophysic dysplasias. American Journal of Human Genetics 89, 7-14Google Scholar
161Loeys, B.L. et al. (2006) Aneurysm syndromes caused by mutations in the TGF-beta receptor. New England Journal of Medicine 355, 788-798CrossRefGoogle ScholarPubMed
162Pannu, H. et al. (2005) Mutations in transforming growth factor-beta receptor type II cause familial thoracic aortic aneurysms and dissections. Circulation 112, 513-520CrossRefGoogle ScholarPubMed
163Ades, L.C. et al. (2006) FBN1, TGFBR1, and the Marfan-craniosynostosis/mental retardation disorders revisited. American Journal of Medical Genetics A 140, 1047-1058Google Scholar
164Coucke, P.J. et al. (2006) Mutations in the facilitative glucose transporter GLUT10 alter angiogenesis and cause arterial tortuosity syndrome. Nature Genetics 38, 452-457Google Scholar
165Doyle, A.J. et al. (2012) Mutations in the TGF-β repressor SKI cause Shprintzen-Goldberg syndrome with aortic aneurysm. Nature Genetics 44, 1249-1254Google Scholar
166Geirsson, A. et al. (2012) Modulation of transforming growth factor-β signaling and extracellular matrix production in myxomatous mitral valves by angiotensin II receptor blockers. Circulation 126, S189-197Google Scholar
167Pober, B.R., Johnson, M., and Urban, Z. (2008) Mechanisms and treatment of cardiovascular disease in Williams-Beuren syndrome. Journal of Clinical Investigation 118, 1606-1615Google Scholar
168Kappanayil, M. et al. (2012) Characterization of a distinct lethal arteriopathy syndrome in twenty-two infants associated with an identical, novel mutation in FBLN4 gene, confirms fibulin-4 as a critical determinant of human vascular elastogenesis. Orphanet Journal of Rare Diseases 7, 61.Google Scholar
169Reversade, B. et al. (2009) Mutations in PYCR1 cause CL with progeroid features. Nature Genetics 41, 1016-1021Google Scholar
170Leao-Teles, E. et al. (2009) De Barsy syndrome and ATP6V0A2-CDG. European Journal of Human Genetics 18, 526Google Scholar
171Urban, Z. et al. (2009) Mutations in LTBP4 cause a syndrome of impaired pulmonary, gastrointestinal, genitourinary, musculoskeletal, and dermal development. American Journal of Human Genetics 85, 593-605Google Scholar
172Basel-Vanagaite, L. et al. (2009) RIN2 deficiency results in macrocephaly, alopecia, CL, and scoliosis: MACS syndrome. American Journal of Human Genetics 85, 254-263Google Scholar
173Segade, F. (2010) Glucose transporter 10 and arterial tortuosity syndrome: the vitamin C connection. FEBS Letters 584, 2990-2994Google Scholar
174Callewaert, B. et al. (2011) New insights into the pathogenesis of autosomal – dominant CL with report of five ELN mutations. Human Mutation 32, 445-455Google Scholar
175Hu, Q. et al. (2006) Inflammatory destruction of elastic fibers in acquired cutis laxa is associated with missense alleles in the elastin and fibulin-5 genes. Journal of Investigative Dermatology 126, 283-290Google Scholar
176Timmer, D.E.M.L. et al. (2009) Acquired CL in childhood Sweet's syndrome. Pediatric Dermatology 26, 358-360Google Scholar
177Karrer, S. et al. (2012) Photodynamic therapy for skin rejuvenation: review and summary of the literature – results of a consensus conference of an expert group for aesthetic photodynamic therapy. JDDG (Journal of the German Society of Dermatology) 11, 137-148Google Scholar
178Lewis, K.G. et al. (2004) Acquired disorders of elastic tissue: part I. Increased elastic tissue and solar elastotic syndromes. Journal of the American Academy of Dermatology 51, 1-21Google Scholar
179Alves, R. et al. (2010) Pseudoxanthoma elasticum papillary dermal elastolysis: a case report. Dermatology Research Practice pii, 352724Google Scholar
180Pasquali-Ronchetti, I. and Baccarani-Contri, M. (1997) Elastic fiber during development and aging. Microscopy Research Technique 38, 428-435Google Scholar
181Klein, R.J. et al. (2005) Complement factor H polymorphism in age-related macular degeneration. Science 308, 385-389Google Scholar
182Lotery, A.J. (2006) Reduced Secretion of fibulin 5 in age-related macular degeneration and cutis laxa. Human Mutation 27, 568-574Google Scholar
183Ambati, J. and Fowler, B.J. (2012) Mechanisms of Age-related Macular Degeneration. Neuron 75, 26-39Google Scholar
184Atzori, L. et al. (2011) D-penicillamine elastosis perforans serpiginosa: description of two cases and review of the literature. Dermatology Online Journal 17, 3Google Scholar
Figure 0

Figure 1. Evolutionary analysis of fibrillin. A list of species from the fibrillin (FBN) sequence alignment was generated, and used to create a common tree using the National Center for Biotechnology Information (NCBI) Taxonomy Browser. The resulting tree was imported into Cytoscape version 2.7, and generated using the organic layout. FBN types were identified using phylogenetic analysis and domain analysis of the FBN multiple sequence alignments and FBN types were mapped onto the common tree. Identified sequences were grouped into 5 categories: vertebrate FBNs 1-3, invertebrate FBN and fibrillin-like epidermal growth factor (EGF) array-containing proteins. The latter sequences had a high identity to FBN sequences but only contained EGF domains organised in arrays. Also indicated are the sequences, from the appearance of chordates that contain the arg-gly-asp (RGD) cell binding motif (branches shown in light brown). By comparison, branches shown in light purple precede chordates and RGD. The first emergence of other key extracellular proteins that interact with FBN is shown (dark blue arrows), along with the evolutionary timescale in million years ago (MYA).

Figure 1

Figure 2. Domain structures of elastic fibre molecules. The domain organisations of fibrillins, latent TGFβ-binding protein (LTBPs), fibulins, a disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin (ADAMTS) and ADAMTS-like (ADAMTSL) molecules and tropoelastin are shown, with keys for domain types.

Figure 2

Table 1. Microfibril and elastic fibre-associated molecules (updated from Ref. 1)

Figure 3

Figure 3. Schematic diagram of microfibril and elastic fibre assembly: elastin assembly. (a) Microfibril assembly occurs pericellularly, and requires fibronectin, integrins and heparan sulphate proteoglycans (HSPG). Fibrillin molecules are secreted and, after processing N- and C-terminally by furin, interact homotypically at N- and C-termini leading to axial and lateral assembly to form microfibrils. Beads may arise from folding of terminal regions. Microfibrils may be stabilised by transglutaminase cross-links. The reason why fibronectin is needed for microfibril deposition is unclear, but it may act as a template for assembly and/or it may stimulate cytoskeletal tension through the α5β1 integrin, thereby facilitating assembly at fibrillar adhesions. Fibrillin-1 also interacts with α5β1, αvβ3 and αvβ6 integrins; however, it is not known whether these interactions are essential for microfibril assembly. Heparin inhibits microfibril assembly, and HSPGs may contribute by facilitating cell surface fibrillin-1 interactions. (b) Elastin assembly occurs pericellularly on ‘microassembly’ and on microfibrils ‘macroaggregates’ (Ref. 69). Secreted tropoelastin forms globules at the cell surface which become cross-linked by lysyl oxidase; this process may involve αvβ3 integrin interactions with tropoelastin, and integrin interactions with heparan sulphate proteoglycans (HSPGs) which can interact with tropoelastin. Fibulin-4 and fibulin-5 contribute to elastin cross-linking by lysyl oxidase, and probably direct the deposition of elastin globules onto preformed fibrillin microfibrils, to form elastic fibres. Microfibrils and elastic fibres are important matrix storage sites for BMPs and latent TGFβ1.

Figure 4

Table 2. Heritable disorders of elastic fibres (updated from Ref. 1)

Figure 5

Figure 4. Effects of the fibrillin-1(Fbn)C1039G/+ phenotype, and Smad4 haploinsufficiency (Smad4+/−), on the aortic architecture in mice. Verhoeff-Van Giesen staining revealed architectural abnormalities in the ascending aorta of these mutant mice. Compared with wild-type littermates, FbnC1039G/+ mice had medial thickening and elastic fibre fragmentation. These defects were enhanced in Smad4+/−: FbnC1039G/+ mice. Figure taken from Ref. 111 (with permission from the corresponding author and the publisher).

Figure 6

Figure 5. Prevention of aortic aneurysm in mice by losartan. Representative murine ascending aortae (arrowheads), after therapy, are: (a) wild-type mice; (b–f) mice heterozygous for Marfan-causing fibrillin-1 mutation C1039 G (FbnC1039G/+), treated with placebo (b), propanolol (c) or losartan (d–f). Scale bars = 4 mm. Figure taken from Ref. 110 (with permission from the corresponding author and the publisher).