Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Matrix Biol ; 102: 70-84, 2021 08.
Article in English | MEDLINE | ID: mdl-34274450

ABSTRACT

The formation of elastic fibers is active only in the perinatal period. How elastogenesis is developmentally regulated is not fully understood. Citrullination is a unique form of post-translational modification catalyzed by peptidylarginine deiminases (PADs), including PAD1-4. Its physiological role is largely unknown. By using an unbiased proteomic approach of lung tissues, we discovered that FBLN5 and LTBP4, two key elastogenic proteins, were temporally modified in mouse and human lungs. We further demonstrated that PAD2 citrullinated FBLN5 preferentially in young lungs compared to adult lungs. Genetic ablation of PAD2 resulted in attenuated elastogenesis in vitro and age-dependent emphysema in vivo. Mechanistically, citrullination protected FBLN5 from proteolysis and subsequent inactivation of its elastogenic activity. Furthermore, citrullinated but not native FBLN5 partially rescued in vitro elastogenesis in the absence of PAD activity. Our data uncover a novel function of citrullination, namely promoting elastogenesis, and provide additional insights to how elastogenesis is regulated.


Subject(s)
Citrullination , Elastic Tissue/growth & development , Extracellular Matrix Proteins/metabolism , Protein-Arginine Deiminase Type 2/metabolism , Recombinant Proteins/metabolism , Animals , Calcium-Binding Proteins , Humans , Mice , Protein Processing, Post-Translational , Protein-Arginine Deiminases/genetics , Protein-Arginine Deiminases/metabolism , Proteomics
2.
Circ Res ; 125(11): 1006-1018, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31590613

ABSTRACT

RATIONALE: Elastin is an important ECM (extracellular matrix) protein in large and small arteries. Vascular smooth muscle cells (SMCs) produce the layered elastic laminae found in elastic arteries but synthesize little elastin in muscular arteries. However, muscular arteries have a well-defined internal elastic lamina (IEL) that separates endothelial cells (ECs) from SMCs. The extent to which ECs contribute elastin to the IEL is unknown. OBJECTIVE: To use targeted elastin (Eln) deletion in mice to explore the relative contributions of SMCs and ECs to elastic laminae formation in different arteries. METHODS AND RESULTS: We used SMC- and EC-specific Cre recombinase transgenes with a novel floxed Eln allele to focus gene inactivation in mice. Inactivation of Eln in SMCs using Sm22aCre resulted in depletion of elastic laminae in the arterial wall with the exception of the IEL and SMC clusters in the outer media near the adventitia. Inactivation of elastin in ECs using Tie2Cre or Cdh5Cre resulted in normal medial elastin and a typical IEL in elastic arteries. In contrast, the IEL was absent or severely disrupted in muscular arteries. Interruptions in the IEL resulted in neointimal formation in the ascending aorta but not in muscular arteries. CONCLUSIONS: Combined with lineage-specific fate mapping systems, our knockout results document an unexpected heterogeneity in vascular cells that produce the elastic laminae. SMCs and ECs can independently form an IEL in most elastic arteries, whereas ECs are the major source of elastin for the IEL in muscular and resistance arteries. Neointimal formation at IEL disruptions in the ascending aorta confirms that the IEL is a critical physical barrier between SMCs and ECs in the large elastic arteries. Our studies provide new information about how SMCs and ECs contribute elastin to the arterial wall and how local elastic laminae defects may contribute to cardiovascular disease.


Subject(s)
Elastic Tissue/metabolism , Elastin/metabolism , Endothelial Cells/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Arteries/growth & development , Arteries/metabolism , Blood Pressure , Cell Lineage , Cell Proliferation , Elastic Tissue/growth & development , Elastic Tissue/ultrastructure , Elastin/deficiency , Elastin/genetics , Endothelial Cells/ultrastructure , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/growth & development , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Neointima , Signal Transduction
3.
Cell Signal ; 62: 109333, 2019 10.
Article in English | MEDLINE | ID: mdl-31176018

ABSTRACT

Arterial wall elastic fibers, made of 90% elastin, are arranged into elastic lamellae which are responsible for the resilience and elastic properties of the large arteries (aorta and its proximal branches). Elastin is synthesized only in early life and adolescence mainly by the vascular smooth muscles cells (VSMC) through the cross-linking of its soluble precursor, tropoelastin. In normal aging, the elastic fibers become fragmented and the mechanical load is transferred to collagen fibers, which are 100-1000 times stiffer than elastic fibers. Minoxidil, an ATP-dependent K+ channel opener, has been shown to stimulate elastin expression in vitro, and in vivo in the aorta of male aged mice and young adult hypertensive rats. Here, we have studied the effect of a 3-month chronic oral treatment with minoxidil (120 mg/L in drinking water) on the abdominal aorta structure and function in adult (6-month-old) and aged (24-month-old) male and female mice. Our results show that minoxidil treatment preserves elastic lamellae integrity at both ages, which is accompanied by the formation of newly synthesized elastic fibers in aged mice. This leads to a generally decreased pulse pressure and a significant improvement of the arterial biomechanical properties in female mice, which present an increased distensibility and a decreased rigidity of the aorta. Our studies show that minoxidil treatment reversed some of the major adverse effects of arterial aging in mice and could be an interesting anti-arterial aging agent, also potentially usable for female-targeted therapies.


Subject(s)
Aorta/growth & development , Arteries/growth & development , Elastic Tissue/growth & development , Minoxidil/pharmacology , Adenosine Triphosphate/genetics , Aging/genetics , Aging/metabolism , Animals , Aorta/drug effects , Arteries/drug effects , Biomechanical Phenomena/genetics , Elastic Tissue/drug effects , Elastin/genetics , Female , Humans , Male , Mice , Potassium Channels/genetics , Protective Agents/pharmacology
4.
Proc Natl Acad Sci U S A ; 116(12): 5350-5355, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30819899

ABSTRACT

Tissue growth is a fundamental aspect of development and is intrinsically noisy. Stochasticity has important implications for morphogenesis, precise control of organ size, and regulation of tissue composition and heterogeneity. However, the basic statistical properties of growing tissues, particularly when growth induces mechanical stresses that can in turn affect growth rates, have received little attention. Here, we study the noisy growth of elastic sheets subject to mechanical feedback. Considering both isotropic and anisotropic growth, we find that the density-density correlation function shows power law scaling. We also consider the dynamics of marked, neutral clones of cells. We find that the areas (but not the shapes) of two clones are always statistically independent, even when they are adjacent. For anisotropic growth, we show that clone size variance scales like the average area squared and that the mode amplitudes characterizing clone shape show a slow [Formula: see text] decay, where n is the mode index. This is in stark contrast to the isotropic case, where relative variations in clone size and shape vanish at long times. The high variability in clone statistics observed in anisotropic growth is due to the presence of two soft modes-growth modes that generate no stress. Our results lay the groundwork for more in-depth explorations of the properties of noisy tissue growth in specific biological contexts.


Subject(s)
Elastic Tissue/growth & development , Animals , Epithelium/growth & development , Feedback , Models, Biological , Morphogenesis/physiology , Organ Size/physiology , Stress, Mechanical
5.
Endocrinology ; 156(1): 90-102, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25353182

ABSTRACT

Glucagon-like peptide 1 (GLP-1) is a metabolic hormone involved in the stimulation of insulin biosynthesis and secretion. It has been recently reported that GLP-1 also exerts cardioprotective effects and facilitates functional recovery after myocardial infarction through GLP-1 receptor-mediated signaling in cardiomyocytes. GLP-1 treatment has been also demonstrated to produce sustained improvement in cardiac function in long-term studies, suggesting the involvement of mechanisms beyond the acute metabolic and cytoprotective effects. For example, the possible interaction of GLP-1 with the cardiac fibroblasts, which are responsible for the postinfarct remodeling and extracellular matrix production, has not been previously explored. Here, we report that cultures of human cardiac fibroblasts treated with GLP-1 peptides display a selective up-regulation in elastin gene expression and a consequent increase in elastic fibers production, in the absence of the classic GLP-1 receptor. Importantly, we provide experimental evidence that this GLP-1-induced elastogenesis is triggered through the cross-activation of the IGF-I receptor. Because GLP-1 does not stimulate deposition of collagen I, nor promote the proliferation or apoptosis of cultured cardiac fibroblasts, we speculate that its elastogenic effect may also contribute to the beneficial remodeling of the human heart after myocardial infarction.


Subject(s)
Elastic Tissue/growth & development , Fibroblasts/physiology , Glucagon-Like Peptide 1/metabolism , Myocardium/cytology , Receptor, IGF Type 1/metabolism , Amino Acid Sequence , Apoptosis , Cell Proliferation , Female , Fibroblasts/cytology , Gene Expression Regulation , Glucagon-Like Peptide 1/genetics , Humans , Male , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics
6.
Tissue Eng Part A ; 20(9-10): 1403-15, 2014 May.
Article in English | MEDLINE | ID: mdl-24313750

ABSTRACT

In vitro tissue engineering of vascular conduits requires a synergy between several external factors, including biochemical supplementation and mechanotranductive stimulation. The goal of this study was to improve adult human vascular smooth muscle cell orientation and elastic matrix synthesis within 3D tubular collagen gel constructs. We used a combination of elastogenic factors (EFs) previously tested in our lab, along with cyclic circumferential strains at low amplitude (2.5%) delivered at a range of frequencies (0.5, 1.5, and 3 Hz). After 21 days of culture, the constructs were analyzed for elastic matrix outcomes, activity of matrix metalloproteinases (MMPs)-2 and -9, cell densities and phenotype, and mechanical properties of constructs. While cell densities remained unaffected by the addition of stretch, contractile phenotypic markers were elevated in all stretched constructs relative to control. Constructs cultured with EFs stretched at 1.5 Hz exhibited the maximum elastin mRNA expression and total matrix elastin (over sixfold vs. the static EFs control). MMP-2 content was comparable in all treatment conditions, but MMP-9 levels were elevated at the higher frequencies (1.5 and 3 Hz). Minimal circumferential orientation was achieved and the mechanical properties remained comparable among the treatment conditions. Overall, constructs treated with EFs and stretched at 1.5 Hz exhibited the most elastogenic outcomes.


Subject(s)
Elastic Tissue/growth & development , Endothelial Cells/physiology , Extracellular Matrix/physiology , Mechanotransduction, Cellular/physiology , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/physiology , Tissue Scaffolds , Cell Proliferation , Cells, Cultured , Elastic Modulus/physiology , Elastic Tissue/cytology , Elastin/metabolism , Endothelial Cells/cytology , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Physical Stimulation/methods , Stress, Mechanical
7.
PLoS One ; 7(6): e38475, 2012.
Article in English | MEDLINE | ID: mdl-22685574

ABSTRACT

Spinal ligaments, such as the ligamentum flavum (LF), are prone to degeneration and iatrogenic injury that can lead to back pain and nerve dysfunction. Repair and regeneration strategies for these tissues are lacking, perhaps due to limited understanding of spinal ligament formation, the elaboration of its elastic fibers, maturation and homeostasis. Using immunohistochemistry and histology, we investigated murine LF elastogenesis and tissue formation from embryonic to mature postnatal stages. We characterized the spatiotemporal distribution of the key elastogenic proteins tropoelastin, fibrillin-1, fibulin-4 and lysyl oxidase. We found that elastogenesis begins in utero with the microfibril constituent fibrillin-1 staining intensely just before birth. Elastic fibers were first detected histologically at postnatal day (P) 7, the earliest stage at which tropoelastin and fibulin-4 stained intensely. From P7 to P28, elastic fibers grew in diameter and became straighter along the axis. The growth of elastic fibers coincided with intense staining of tropoelastin and fibulin-4 staining, possibly supporting a chaperone role for fibulin-4. These expression patterns correlated with reported skeletal and behavioral changes during murine development. This immunohistochemical characterization of elastogenesis of the LF will be useful for future studies investigating mechanisms for elastogenesis and developing new strategies for treatment or regeneration of spinal ligaments and other highly elastic tissues.


Subject(s)
Extracellular Matrix Proteins/metabolism , Ligamentum Flavum/metabolism , Microfilament Proteins/metabolism , Protein-Lysine 6-Oxidase/metabolism , Tropoelastin/metabolism , Animals , Elastic Tissue/embryology , Elastic Tissue/growth & development , Elastic Tissue/metabolism , Fibrillin-1 , Fibrillins , Immunohistochemistry , Ligamentum Flavum/embryology , Ligamentum Flavum/growth & development , Mice , Mice, Inbred C57BL , Mice, Transgenic , Time Factors
8.
Mol Genet Metab ; 106(1): 99-103, 2012 May.
Article in English | MEDLINE | ID: mdl-22386972

ABSTRACT

Galactosialidosis is a lysosomal storage disorder caused by loss of function of protective protein cathepsin A, which leads to secondary deficiencies of ß-galactosidase and neuraminidase-1. Emphysema has not been previously reported as a possible complication of this disorder, but we now describe this condition in a 41-year-old, non-smoking male. Our patient did not display deficiency in α-1-antitrypsin, the most common cause of emphysema in non-smokers, which brings about disseminated elastolysis. We therefore hypothesized that loss of cathepsin A activity was responsible because of previously published evidence showing it is prerequisite for normal elastogenesis. We now present experimental evidence to support this theory by demonstrating impaired primary elastogenesis in cultures of dermal fibroblasts from our patient. The obtained data further endorse our previous finding that functional integrity of the cell surface-targeted molecular complex of cathepsin A, neuraminidase-1 and the elastin-binding protein (spliced variant of ß-galactosidase) is prerequisite for the normal assembly of elastic fibers. Importantly, we also found that elastic fiber production was increased after exposure either to losartan, spironolactone, or dexamethasone. Of immediate clinical relevance, our data suggest that surviving patients with galactosialidosis should have periodic assessment of their pulmonary function. We also encourage further experimental exploration of therapeutic potential of the afore-mentioned elastogenesis-stimulating drugs for the alleviation of pathological processes in galactosialidosis that could be mechanistically linked to impaired deposition of elastic fibers.


Subject(s)
Cathepsin A , Elastic Tissue , Emphysema , Lysosomal Storage Diseases , Adult , Cathepsin A/genetics , Cathepsin A/metabolism , Cells, Cultured , Elastic Tissue/enzymology , Elastic Tissue/growth & development , Elastic Tissue/ultrastructure , Elastin/genetics , Elastin/metabolism , Emphysema/etiology , Emphysema/pathology , Fibrillins , Fibroblasts , Gene Expression/genetics , Humans , Lysosomal Storage Diseases/complications , Lysosomal Storage Diseases/pathology , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neuraminidase/genetics , Neuraminidase/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
9.
J Periodontal Res ; 45(1): 52-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19602118

ABSTRACT

BACKGROUND AND OBJECTIVE: Elastic system fibers are a major component of the periodontal ligament, but little information is available about their detailed composition or the mechanism of elastogenesis in the developing periodontal ligament. The purpose of this study was to investigate immunolocalization of elastin, fibrillins and microfibril-associated glycoprotein-1 (MAGP-1) in the developing periodontal ligament of the rat molar. MATERIAL AND METHODS: Frozen sections of demineralized as well as non-demineralized periodontal ligament of Wistar rats of various ages from 19 days to 7 weeks were incubated with anti-elastin, anti-fibrillin-1 and -2 and anti-MAGP-1 antibodies followed by peroxidase-conjugated secondary antibodies. After incubation with diaminobenzidine solution, immunoreaction products were observed with a light microscope. RESULTS: In the developing periodontal ligament of 19-day-old rats, fibers immunopositive to elastin were not present, but fibers positively stained for fibrillin-2 and MAGP-1 were widely distributed throughout the ligament. The latter fibers were arranged in the apico-occlusal direction along with blood vessels. In 3-week-old rats, fibers stained for elastin were observed for the first time in the apical region of the ligament. The number and distribution pattern of these elastin-positive fibers was basically the same as those in rats aged 5 and 7 weeks. In contrast, fibrillin-2- and MAGP-1-positive fibers were more extensively distributed in the ligament, and their pattern of distribution was comparable to that of reported oxytalan fibers. Fibrillin-1 was, however, not detected either in demineralized sections or in non-demineralized sections, indicating its absence in periodontal ligament. CONCLUSION: Elastin expressed in the periodontal ligament assembled into elaunin fibers in the vicinity of blood vessels. Both fibrillin-2 and MAGP-1 are structural components not only of the elastin-associated microfibrils but also of elastin-free microfibrils, with possible roles in elastogenesis and in periodontal ligament homeostasis.


Subject(s)
Contractile Proteins/analysis , Elastic Tissue/growth & development , Elastin/analysis , Extracellular Matrix Proteins/analysis , Microfilament Proteins/analysis , Molar/anatomy & histology , Periodontal Ligament/growth & development , Animals , Fibrillin-1 , Fibrillin-2 , Fibrillins , Immunohistochemistry , Male , Odontogenesis/physiology , Periodontal Ligament/blood supply , RNA Splicing Factors , Rats , Rats, Wistar , Tooth Crown/growth & development , Tooth Eruption/physiology , Tooth Root/growth & development
10.
Age (Dordr) ; 31(4): 305-25, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19588272

ABSTRACT

The ability of elastic tissues to deform under physiological forces and to subsequently release stored energy to drive passive recoil is vital to the function of many dynamic tissues. Within vertebrates, elastic fibres allow arteries and lungs to expand and contract, thus controlling variations in blood pressure and returning the pulmonary system to a resting state. Elastic fibres are composite structures composed of a cross-linked elastin core and an outer layer of fibrillin microfibrils. These two components perform distinct roles; elastin stores energy and drives passive recoil, whilst fibrillin microfibrils direct elastogenesis, mediate cell signalling, maintain tissue homeostasis via TGFß sequestration and potentially act to reinforce the elastic fibre. In many tissues reduced elasticity, as a result of compromised elastic fibre function, becomes increasingly prevalent with age and contributes significantly to the burden of human morbidity and mortality. This review considers how the unique molecular structure, tissue distribution and longevity of elastic fibres pre-disposes these abundant extracellular matrix structures to the accumulation of damage in ageing dermal, pulmonary and vascular tissues. As compromised elasticity is a common feature of ageing dynamic tissues, the development of strategies to prevent, limit or reverse this loss of function will play a key role in reducing age-related morbidity and mortality.


Subject(s)
Aging/physiology , Elastic Tissue/growth & development , Elastin/analysis , Extracellular Matrix Proteins/analysis , Extracellular Matrix/metabolism , Microfibrils/chemistry , Animals , Elastic Tissue/metabolism , Elasticity , Humans
11.
Connect Tissue Res ; 49(5): 321-7, 2008.
Article in English | MEDLINE | ID: mdl-18991085

ABSTRACT

This work was designed to determine the course of development of elastic fibers in myocardial scars in rats and their relationship to other components of such structures. Light and electron microscopic observations were made on tissues from 24 rats, killed at sequent stages from 4 to 24 days postinjury. By both techniques, elastic fibers, shown to be forming by 4 days, had increased in size and number with maturation of the scar. At later stages they became interdigitated with the stumps of viable myocytes. We also saw that these fibers often had formed close contacts with the cell surfaces of myofibroblasts and nonvascular smooth muscle cells; a process found in some other situations but not previously in myocardial scars. This information is relevant, in particular, to the dynamics of myocardial scars and thus to the maintenance of function in the injured heart, but also to elastic fiber behavior in general. The integral role of elastic fibers in cell-matrix interactions as well as their biomechanical function is emphasized.


Subject(s)
Cicatrix/pathology , Elastic Tissue/growth & development , Elastic Tissue/ultrastructure , Heart Injuries/pathology , Myocardium/ultrastructure , Animals , Fibroblasts/ultrastructure , Male , Microscopy, Electron, Transmission , Muscle Cells/ultrastructure , Rats , Rats, Sprague-Dawley
13.
Mol Cell Biol ; 28(3): 1061-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18070922

ABSTRACT

Fibulin-2 is an extracellular matrix protein belonging to the five-member fibulin family, of which two members have been shown to play essential roles in elastic fiber formation during development. Fibulin-2 interacts with two major constituents of elastic fibers, tropoelastin and fibrillin-1, in vitro and localizes to elastic fibers in many tissues in vivo. The protein is prominently expressed during morphogenesis of the heart and aortic arch vessels and at early stages of cartilage development. To examine its role in vivo, we generated mice that do not express the fibulin-2 gene (Fbln2) through homologous recombination of embryonic stem cells. Unexpectedly, the fibulin-2-null mice were viable and fertile and did not display gross and anatomical abnormalities. Histological and ultrastructural analyses revealed that elastic fibers assembled normally in the absence of fibulin-2. No compensatory up-regulation of mRNAs for other fibulin members was detected in the aorta and skin tissue. However, in the fibulin-2 null aortae, fibulin-1 immunostaining was increased in the inner elastic lamina, where fibulin-2 preferentially localizes. The results demonstrate that fibulin-2 is not required for mouse development and elastic fiber formation and suggest possible functional redundancy between fibulin-1 and fibulin-2.


Subject(s)
Calcium-Binding Proteins/physiology , Elastic Tissue/growth & development , Extracellular Matrix Proteins/physiology , Animals , Aorta/chemistry , Aorta/cytology , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/deficiency , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/deficiency , Growth and Development , Mice , Mice, Knockout , Phenotype
14.
J Nutr ; 137(8): 1895-900, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17634261

ABSTRACT

Elastic fibers play a key role in the structure and function of numerous organs that require elasticity. Elastogenesis is a complex process in which cells first produce a microfibrillar scaffold, composed of numerous structural proteins, upon which tropoelastin assembles to be cross-linked into polymeric elastin. Recently, it was demonstrated that low concentrations of free iron upregulate elastin gene expression in cultured fibroblasts. The present studies were conducted to assess whether low-iron diets would affect the deposition of elastic fibers in an in vivo model. One-day-old chicks were fed semipurified diets containing 1.3 (low), 12 (moderate), and 24 (control) mg/kg of iron. After 3 wk, chicks in the low-iron group were underweight and anemic. Their aortas were smaller with significantly thinner walls than control chicks, yet elastin or collagen content did not decrease relative to total protein. They also demonstrated a significantly lower stress-strain resistance than the controls. Electron microscopy demonstrated that aortic and lung smooth muscle cells were vacuolated and surrounded by loose extracellular matrix and disorganized elastic lamellae with diffuse and fragmented networks of elastic fibers and microfibrils. Immunohistology demonstrated that fibrillin-3 (FBN3) was disorganized and markedly reduced in amount in aortas of the low-iron chicks. Elastin messenger RNA levels were not downregulated in the tissues from the low-iron-fed chicks; however, there was a significant reduction in expression of the FBN1 and FBN3 genes compared with control chicks. The studies indicate that iron deficiency had a pronounced negative effect on elastic fiber development and suggests that fibrillin may have an important role in this pathology.


Subject(s)
Aorta/growth & development , Chickens/metabolism , Elastic Tissue/growth & development , Iron Deficiencies , Iron, Dietary/administration & dosage , Lung/growth & development , Animal Feed/analysis , Animals , Aorta/drug effects , Aorta/ultrastructure , Body Weight/drug effects , Diet , Elastic Tissue/drug effects , Iron, Dietary/metabolism , Lung/drug effects , Lung/ultrastructure
15.
Birth Defects Res C Embryo Today ; 81(4): 229-40, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18228265

ABSTRACT

Elastic fibers provide recoil to tissues that undergo repeated stretch, such as the large arteries and lung. These large extracellular matrix (ECM) structures contain numerous components, and our understanding of elastic fiber assembly is changing as we learn more about the various molecules associated with the assembly process. The main components of elastic fibers are elastin and microfibrils. Elastin makes up the bulk of the mature fiber and is encoded by a single gene. Microfibrils consist mainly of fibrillin, but also contain or associate with proteins such as microfibril associated glycoproteins (MAGPs), fibulins, and EMILIN-1. Microfibrils were thought to facilitate alignment of elastin monomers prior to cross-linking by lysyl oxidase (LOX). We now know that their role, as well as the overall assembly process, is more complex. Elastic fiber formation involves elaborate spatial and temporal regulation of all of the involved proteins and is difficult to recapitulate in adult tissues. This report summarizes the known interactions between elastin and the microfibrillar proteins and their role in elastic fiber assembly based on in vitro studies and evidence from knockout mice. We also propose a model of elastic fiber assembly based on the current data that incorporates interactions between elastin, LOXs, fibulins and the microfibril, as well as the pivotal role played by cells in structuring the final functional fiber.


Subject(s)
Elastic Tissue/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Contractile Proteins/genetics , Contractile Proteins/metabolism , Elastic Tissue/growth & development , Elastic Tissue/ultrastructure , Elastin/deficiency , Elastin/genetics , Elastin/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibrillins , Gene Expression Regulation, Developmental , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Models, Biological , Mutation , Protein-Lysine 6-Oxidase/metabolism , RNA Splicing Factors , Rats , Tropoelastin/genetics , Tropoelastin/metabolism
16.
Hum Mol Genet ; 15(23): 3379-86, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17035250

ABSTRACT

To elucidate the molecular mechanisms of impaired elastic fiber formation in recessive cutis laxa, we have investigated two disease-causing missense substitutions in fibulin-5, C217R and S227P. Pulse-chase immunoprecipitation experiments indicated that S227P mutant fibulin-5 was synthesized and secreted by skin fibroblasts at a reduced rate when compared with the wild-type protein. Both mutants failed to be incorporated into elastic fibers by transfected rat lung fibroblasts. Purified recombinant fibulin-5 with either mutation showed reduced affinity for tropoelastin in solid-phase binding assays. Furthermore, S227P mutant fibulin-5 also showed impaired association with fibrillin-1 microfibrils. The same mutation triggered an endoplasmic reticulum (ER) stress response, as indicated by the strong co-localization of this mutant protein with folding chaperones in the ER, including calreticulin, immunoglobulin-binding protein and protein disulfide isomerase, and by increased rates of apoptosis in patient fibroblasts. Histological analysis of skin sections from a cutis laxa patient with a homozygous S227P mutation showed a lack of fibulin-5 in the extracellular matrix and a concomitant disorganization of dermal elastic fibers. By electron microscopy, elastic fibers in the skin of this patient showed a failure of elastin globules to fuse into a continuous elastic fiber core. We conclude that recessive cutis laxa mutations in fibulin-5 result in misfolding, decreased secretion and a reduced interaction with elastin and fibrillin-1 leading to impaired elastic fiber development. These findings support the hypothesis that fibulin-5 is necessary for elastic fiber formation by facilitating the deposition of elastin onto a microfibrillar scaffold via direct molecular interactions.


Subject(s)
Cutis Laxa/genetics , Cutis Laxa/pathology , Elastic Tissue/metabolism , Elastic Tissue/ultrastructure , Extracellular Matrix Proteins/genetics , Skin/pathology , Amino Acid Substitution , Animals , Calreticulin/analysis , Calreticulin/metabolism , Cutis Laxa/metabolism , Elastic Tissue/growth & development , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/metabolism , Fibrillin-1 , Fibrillins , Fibroblasts/metabolism , Fibroblasts/pathology , Genes, Recessive , Humans , Immunoprecipitation , Microfilament Proteins/analysis , Microfilament Proteins/metabolism , Mutation, Missense , Prostatic Secretory Proteins/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Folding , Rats , Skin/metabolism , Tropoelastin/metabolism
17.
Otolaryngol Head Neck Surg ; 134(5): 843-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16647545

ABSTRACT

OBJECTIVE: To determine if subglottic development is at least partially under local control and to determine which tissue layer(s) is predominantly responsible. DESIGN: The suglottises of 12 day-3 CD1 mice were grown in whole organ culture. The 12 subglottises were divided into 3 individual groups: +++, -++, and ---. Group+++ had all tissue layers of the subglottis intact: luminal epithelium, cricoid cartilage, inner and outer perichondrium. Group-++ had all layers intact with the exception of luminal epithelium. Group--- had all layers removed (luminal epithelium, inner and outer perichondrium) resulting in cricoid cartilage-only rings. All rings were grown in basic medium without the use of growth factors or serum for 15 days. Measurements of the rings were taken before and after organ culture growth. RESULTS: Group+++ was the only group that experienced growth. Only luminal growth was statistically significant although all rings experienced growth in both the luminal and external diameter. Group-++ did not experience any growth. Group--- lost structural integrity with collapse of the ring and did not experience growth of any dimension of the cartilage. CONCLUSIONS: Growth of the subglottis is under local control but may have additional influences from the outside that were not investigated here. Removal of just the epithelium stunts growth of the entire ring, but preferentially the lumen more so than the external diameter. Removal of all tissue layers around the cricoid cartilage results in a structural collapse of the ring, suggesting that the cartilage in this age group is dependent on surrounding tissues for structural integrity.


Subject(s)
Cricoid Cartilage/growth & development , Glottis/growth & development , Animals , Animals, Newborn , Elastic Tissue/growth & development , Follow-Up Studies , Laryngeal Mucosa/growth & development , Mice , Organ Culture Techniques
18.
J Cell Physiol ; 207(1): 97-106, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16331676

ABSTRACT

Elastic fibers are responsible for the extensibility and resilience of many vertebrate tissues, and improperly assembled elastic fibers are implicated in a number of human diseases. It was recently demonstrated that in vitro, cells first secrete tropoelastin into a punctate pattern of globules. To study the dynamics of macroassembly, that is, the assembly of the secreted tropoelastin globules into elastic fibers, we utilized long-term time-lapse immunofluorescence imaging and a tropoelastin p Timer fusion protein, which shifts its fluorescence spectrum over time. Pulse-chase immunolabeling of the fibroblast-like RFL-6 cells demonstrates that tropoelastin globules aggregate in a hierarchical manner, creating progressively larger fibrillar structures. By analyzing the correlation between cell and extracellular matrix movements, we show that both the aggregation process and shaping the aggregates into fibrillar form is coupled to cell motion. We also show that the motion of non-adjacent cells becomes more coordinated as the physical size of elastin-containing aggregates increases. Our data imply that the formation of elastic fibers involves the concerted action and motility of multiple cells.


Subject(s)
Cell Movement/physiology , Elastic Tissue/metabolism , Extracellular Matrix/metabolism , Animals , Cattle , Cell Line , Elastic Tissue/growth & development , Elastin/metabolism , Extracellular Matrix/ultrastructure , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fluorescent Antibody Technique , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Protein Transport , Rats , Rats, Sprague-Dawley , Time Factors , Transfection , Tropoelastin/genetics , Tropoelastin/immunology , Tropoelastin/metabolism
19.
J Cell Physiol ; 207(1): 87-96, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16261592

ABSTRACT

To study the dynamics of elastic fiber assembly, mammalian cells were transfected with a cDNA construct encoding bovine tropoelastin in frame with the Timer reporter. Timer is a derivative of the DsRed fluorescent protein that changes from green to red over time and, hence, can be used to distinguish new from old elastin. Using dynamic imaging microscopy, we found that the first step in elastic fiber formation is the appearance of small cell surface-associated elastin globules that increased in size with time (microassembly). The elastin globules are eventually transferred to pre-existing elastic fibers in the extracellular matrix where they coalesce into larger structures (macroassembly). Mechanical forces associated with cell movement help shape the forming, extracellular elastic fiber network. Time-lapse imaging combined with the use of Timer constructs provides unique tools for studying the temporal and spatial aspects of extracellular matrix formation by live cells.


Subject(s)
Elastic Tissue/metabolism , Extracellular Matrix/metabolism , Animals , Cattle , Cell Line , Cell Movement/physiology , Chondrocytes/ultrastructure , Elastic Tissue/growth & development , Elastin/analysis , Elastin/metabolism , Elastin/ultrastructure , Extracellular Matrix/ultrastructure , Fibroblasts/chemistry , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Myocytes, Smooth Muscle/ultrastructure , Rats , Time Factors , Transfection , Tropoelastin/genetics , Tropoelastin/metabolism
20.
J Biol Chem ; 279(28): 29185-94, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15131124

ABSTRACT

The interactions of microfibril-associated glycoprotein (MAGP)-2 have been investigated with fibrillins and fibrillin-containing microfibrils. Solid phase binding assays were conducted with recombinant fragments covering fibrillin-1 and most of fibrillin-2. MAGP-2, and its structure relative MAGP-1, were found to bind two fragments spanning the N-terminal half of fibrillin-1 and an N-terminal fragment of fibrillin-2. Blocking experiments indicated that MAGP-2 had a binding site(s) close to the N terminus of the fibrillin-1 molecule that was distinct from that for MAGP-1 and an additional, more central binding site(s) that may be shared by the two MAGPs. Immunogold labeling of developing nuchal ligament tissue showed that MAGP-2 had regular covalent and periodic (about 56 nm) association with fibrillin-containing microfibrils of elastic fibers in this tissue. Further analysis of isolated microfibrils indicated that MAGP-2 was attached at two points along the microfibril substructure, "site 1" on the "beads" and "site 2" at the "shoulder" of the interbead region close to where the two "arms" fuse. In contrast, MAGP-1 was located only on the beads. Comparison of the MAGP-2 binding data with known fibrillin epitope maps of the microfibrils showed that site 1 correlated with the N-terminal MAGP-2 binding region, and site 2 correlated with the second, more central, MAGP-2 binding region on the fibrillin-1 molecule. Of particular note, immunolabeling at site 2 was markedly decreased, relative to that at site 1, on extended microfibrils with bead-to-bead periods over 90 nm, suggesting that site 2 may move toward the beads when the microfibril is stretched. The study points to MAGP-2 being an integral component of some populations of fibrillin-containing microfibrils. Moreover, the identification of multiple MAGP-binding sequences on fibrillins supports the concept that MAGPs may function as molecular cross-linkers, stabilizing fibrillin monomers in folded conformation within or between the microfibrils, and thus MAGPs may be implicated in the modulation of the elasticity of these structures.


Subject(s)
Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Microfibrils/metabolism , Microfilament Proteins/metabolism , Animals , Binding Sites , Cattle , Cell Line , Elastic Tissue/growth & development , Elastic Tissue/metabolism , Elastic Tissue/ultrastructure , Epitopes , Fibrillin-1 , Fibrillin-2 , Fibrillins , Humans , Immunohistochemistry , Ligaments/growth & development , Ligaments/metabolism , Ligaments/ultrastructure , Microfilament Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , RNA Splicing Factors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...