Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 20(20)2019 Oct 12.
Article in English | MEDLINE | ID: mdl-31614727

ABSTRACT

Vascular endothelial growth factor-A (VEGF) is critical for the development, growth, and survival of blood vessels. Retinal pigmented epithelial (RPE) cells are a major source of VEGF in the retina, with evidence that the extracellular matrix (ECM)-binding forms are particularly important. VEGF associates with fibronectin in the ECM to mediate distinct signals in endothelial cells that are required for full angiogenic activity. Hypoxia stimulates VEGF expression and angiogenesis; however, little is known about whether hypoxia also affects VEGF deposition within the ECM. Therefore, we investigated the role of hypoxia in modulating VEGF-ECM interactions using a primary retinal cell culture model. We found that retinal endothelial cell attachment to RPE cell layers was enhanced in cells maintained under hypoxic conditions. Furthermore, we found that agents that disrupt VEGF-fibronectin interactions inhibited endothelial cell attachment to RPE cells. We also found that hypoxia induced a general change in the chemical structure of the HS produced by the RPE cells, which correlated to changes in the deposition of VEGF in the ECM, and we further identified preferential binding of VEGFR2 over VEGFR1 to VEGF laden-fibronectin matrices. Collectively, these results indicate that hypoxia-induced HS may prime fibronectin for VEGF deposition and endothelial cell recruitment by promoting VEGF-VEGFR2 interactions as a potential means to control angiogenesis in the retina and other tissues.


Subject(s)
Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Heparitin Sulfate/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Adhesion , Cell Hypoxia , Cell Line , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Oxygen/metabolism , Rats , Retinal Pigment Epithelium/cytology
2.
Langmuir ; 35(31): 10061-10067, 2019 08 06.
Article in English | MEDLINE | ID: mdl-30681875

ABSTRACT

The development of new therapies for surgical adhesions has proven to be difficult as there is no consistently effective way to assess treatment efficacy in clinical trials without performing a second surgery, which can result in additional adhesions. We have developed lipid microbubble formulations that use a short peptide sequence, CREKA, to target fibrin, the molecule that forms nascent adhesions. These targeted polymerized shell microbubbles (PSMs) are designed to allow ultrasound imaging of early adhesions for diagnostic purposes and for evaluating the success of potential treatments in clinical trials while acting as a possible treatment. In this study, we show that CREKA-targeted microbubbles preferentially bind fibrin over fibrinogen and are stable for long periods of time (∼48 h), that these bound microbubbles can be visualized by ultrasound, and that neither these lipid-based bubbles nor their diagnostic-ultrasound-induced vibrations damage mesothelial cells in vitro. Moreover, these bubbles show the potential to identify adhesionlike fibrin formations and may hold promise in blocking or breaking up fibrin formations in vivo.


Subject(s)
Contrast Media/chemistry , Fibrin/metabolism , Microbubbles , Tissue Adhesions/diagnostic imaging , Cell Line , Cell Survival/drug effects , Contrast Media/toxicity , Humans , Lab-On-A-Chip Devices , Microfluidics/instrumentation , Microfluidics/methods , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/toxicity , Phosphatidylcholines/chemistry , Phosphatidylcholines/toxicity , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/toxicity , Polyacetylene Polymer/chemical synthesis , Polyacetylene Polymer/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Theranostic Nanomedicine/methods , Ultrasonography/methods
3.
J Cell Physiol ; 231(8): 1728-36, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26621030

ABSTRACT

The extracellular matrix (ECM) is present in a range of molecular conformations and intermolecular arrangements. Fibronectin (Fn) molecules that constitute fibers within the ECM can exist in a variety of conformations that result from both mechanical stress and chemical factors such as allosteric binding partners. The long-standing hypothesis that conformational changes regulate the binding of cells to Fn fibers has only been tested for mutated molecules of Fn and has yet to be fully evaluated with Fn fibers. Using time-lapse microscopy we examined how mechanical extension of single fibers of Fn affects the adhesion and migration of endothelial cells. Using this single fiber adhesion technique, we show that high levels of mechanical strain applied to Fn fibers decreases the rates of both cell spreading and cell migration. These data indicate a fundamental cellular response to mechanical strain in the ECM that might have important implications for understanding how cells are recruited during tissue development and repair. J. Cell. Physiol. 231: 1728-1736, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Cell Adhesion , Cell Movement , Cell Shape , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Fibronectins/metabolism , Mechanotransduction, Cellular , Animals , Cattle , Cells, Cultured , Humans , Integrin alpha5beta1/metabolism , Microscopy, Fluorescence , Stress, Mechanical , Time Factors , Time-Lapse Imaging
4.
PLoS One ; 10(12): e0145115, 2015.
Article in English | MEDLINE | ID: mdl-26672607

ABSTRACT

Elastase released from neutrophils as part of the innate immune system has been implicated in chronic diseases such as emphysema and cardiovascular disease. We have previously shown that neutrophil elastase targets vascular endothelial growth factor-A (VEGF) for partial degradation to generate a fragment of VEGF (VEGFf) that has distinct activities. Namely, VEGFf binds to VEGF receptor 1 but not to VEGF receptor 2 and shows altered signaling compared to intact VEGF. In the present study we investigated the chemotactic function of VEGF and VEGFf released from cells by neutrophil elastase. We found that endothelial cells migrated in response to intact VEGF but not VEGFf whereas RAW 264.7 macrophages/monocytes and embryonic endothelial progenitor cells were stimulated to migrate by either VEGF or VEGFf. To investigate the role of elastase-mediated release of VEGF from cells/extracellular matrices, a co-culture system was established. High or low VEGF producing cells were co-cultured with macrophages, endothelial or endothelial progenitor cells and treated with neutrophil elastase. Elastase treatment stimulated macrophage and endothelial progenitor cell migration with the response being greater with the high VEGF expressing cells. However, elastase treatment led to decreased endothelial cell migration due to VEGF cleavage to VEGF fragment. These findings suggest that the tissue response to NE-mediated injury might involve the generation of diffusible VEGF fragments that stimulate inflammatory cell recruitment.


Subject(s)
Cell Movement , Endothelial Progenitor Cells/drug effects , Macrophages/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cattle , Endothelial Progenitor Cells/physiology , Humans , Macrophages/physiology , Mice , Pancreatic Elastase/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Proteolysis , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Sf9 Cells , Spodoptera , Vascular Endothelial Growth Factor A/chemistry
5.
Matrix Biol ; 41: 36-43, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448408

ABSTRACT

The development of atherosclerosis involves phenotypic changes among vascular smooth muscle cells (VSMCs) that correlate with stiffening and remodeling of the extracellular matrix (ECM). VSMCs are highly sensitive to the composition and mechanical state of the surrounding ECM, and ECM remodeling during atherosclerosis likely contributes to pathology. We hypothesized that ECM mechanics and biochemistry are interdependent in their regulation of VSMC behavior and investigated the effect of ligand presentation on certain stiffness-mediated processes. Our findings demonstrate that substrate stiffening is not a unidirectional stimulus-instead, the influence of mechanics on cell behavior is highly conditioned on ligand biochemistry. This "stiffness-by-ligand" effect was evident for VSMC adhesion, spreading, cytoskeletal polymerization, and focal adhesion assembly, where VSMCs cultured on fibronectin (Fn)-modified substrates showed an augmented response to increasing stiffness, whereas cells on laminin (Ln) substrates showed a dampened response. By contrast, cells on Fn substrates showed a decrease in myosin light chain (MLC) phosphorylation and elongation with increasing stiffness, whereas Ln supported an increase in MLC phosphorylation and no change in cell shape with increasing stiffness. Taken together, these findings show that identical cell populations exhibit opposing responses to substrate stiffening depending on ECM presentation. Our results also suggest that the shift in VSMC phenotype in a developing atherosclerotic lesion is jointly regulated by stromal mechanics and biochemistry. This study highlights the complex influence of the blood vessel wall microenvironment on VSMC phenotype and provides insight into how cells may integrate ECM biochemistry and mechanics during normal and pathological tissue function.


Subject(s)
Aorta/cytology , Extracellular Matrix/physiology , Mechanotransduction, Cellular , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Animals , Animals, Newborn , Aorta/physiology , Cell Adhesion , Cell Proliferation , Cells, Cultured , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myosin Light Chains/metabolism , Rats
6.
Ann Biomed Eng ; 43(3): 762-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25515314

ABSTRACT

Complex hierarchical organization is a hallmark of tissues and their subsequent integration into organs. A major challenge in tissue engineering is to generate arrays of cells with defined structural organization that display appropriate functional properties. Given what is known about cellular responses to physiochemical cues from the surrounding environment, we can build tissue structures that mimic these microenvironments and validate these platforms using both experimental and computational approaches. Tissue generation encompasses many methods and tissue types, but here we review layering cell sheets to create scaffold-less myocardial patches. We discuss surgical criteria that can drive the design of myocardial cell sheets and the methods used to fabricate, mechanically condition, and functionally test them. We also focus on how computational and experimental approaches could be integrated to optimize tissue mechanical properties by using measurements of biomechanical properties and tissue anisotropy to create predictive computational models. Tissue anisotropy and dynamic mechanical stimuli affect cell phenotype in terms of protein expression and secretion, which in turn, leads to compositional and structural changes that ultimately impact tissue function. Therefore, a combinatorial approach of design, fabrication, testing, and modeling can be carried out iteratively to optimize engineered tissue function.


Subject(s)
Myocardium , Tissue Engineering , Animals , Cardiac Surgical Procedures , Humans , Models, Theoretical
7.
J Biol Chem ; 289(49): 34141-51, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25336655

ABSTRACT

Extracellular heparanase activity releases growth factors and angiogenic factors from heparan sulfate (HS) storage sites and alters the integrity of the extracellular matrix. These activities lead to a loss of normal cell matrix adherent junctions and correlate with invasive cellular phenotypes. Elevated expression of heparanase is associated with several human cancers and with vascular remodeling. Heparanase cleaves only a limited fraction of glucuronidic linkages in HS. There have been few investigations of the functional consequences of heparanase activity, largely due to the heterogeneity and complexity of HS. Here, we report a liquid chromatography-mass spectrometry (LC-MS)-based approach to profile the terminal structures created by heparanase digestion and reconstruct the heparanase cleavage sites from the products. Using this method, we demonstrate that heparanase cleaves at the non-reducing side of highly sulfated HS domains, exposing cryptic growth factor binding sites. This cleavage pattern is observed in HS from several tissue sources, regardless of overall sulfation degree, indicating a common recognition pattern. We further demonstrate that heparanase cleavage of HS chains leads to increased ability to support FGF2-dependent cell proliferation. These results suggest a new mechanism to explain how heparanase might potentiate the uncontrolled cell proliferation associated with cancer through its ability to activate nascent growth factor-promoting domains within HS.


Subject(s)
Extracellular Matrix/chemistry , Glucuronidase/metabolism , Heparitin Sulfate/chemistry , Lymphocytes/enzymology , Animals , Carbohydrate Sequence , Cattle , Cell Line, Tumor , Chromatography, Liquid , Extracellular Matrix/drug effects , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Glucuronidase/genetics , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Swine , Syndecan-4/genetics , Syndecan-4/metabolism , Tandem Mass Spectrometry
8.
PLoS One ; 9(8): e105143, 2014.
Article in English | MEDLINE | ID: mdl-25127119

ABSTRACT

Sulfs are extracellular endosulfatases that selectively remove the 6-O-sulfate groups from cell surface heparan sulfate (HS) chain. By altering the sulfation at these particular sites, Sulfs function to remodel HS chains. As a result of the remodeling activity, HSulf2 regulates a multitude of cell-signaling events that depend on interactions between proteins and HS. Previous efforts to characterize the substrate specificity of human Sulfs (HSulfs) focused on the analysis of HS disaccharides and synthetic repeating units. In this study, we characterized the substrate preferences of human HSulf2 using HS oligosaccharides with various lengths and sulfation degrees from several naturally occurring HS sources by applying liquid chromatography mass spectrometry based glycomics methods. The results showed that HSulf2 preferentially digests highly sulfated HS oligosaccharides with zero acetyl groups and this preference is length dependent. In terms of length of oligosaccharides, HSulf2 digestion induced more sulfation decrease on DP6 (DP: degree of polymerization) compared to DP2, DP4 and DP8. In addition, the HSulf2 preferentially digests the oligosaccharide domain located at the non-reducing end (NRE) of the HS and heparin chain. In addition, the HSulf2 digestion products were altered only for specific isomers. HSulf2 treated NRE oligosaccharides also showed greater decrease in cell proliferation than those from internal domains of the HS chain. After further chromatographic separation, we identified the three most preferred unsaturated hexasaccharide for HSulf2.


Subject(s)
Heparitin Sulfate/chemistry , Oligosaccharides/chemistry , Sulfotransferases/chemistry , Animals , Cell Line , Cell Proliferation , Chromatography, Ion Exchange , Glycomics , Heparin Lyase/chemistry , Humans , Hydrolysis , Mass Spectrometry , Substrate Specificity , Sulfatases , Sus scrofa
9.
Matrix Biol ; 34: 124-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24148804

ABSTRACT

Extracellular matrix (ECM) conformation is regulated by a variety of stimuli in vivo, including mechanical forces and allosteric binding partners, and these conformational changes contribute to the regulation of cell behavior. Heparin and heparan sulfate, for example, have been shown to regulate the sequestration and presentation of numerous growth factors, including vascular endothelial growth factor, on the heparin 2 binding domain in fibronectin (Fn). However, mechanical force also alters Fn conformation, indicating that the growth factor binding region may be co-regulated by both heparin and mechanical force. Herein, we describe a simple antibody-based method for evaluating the conformation of the heparin 2 binding domain in Fn, and use it to determine the relative contributions of heparin and mechanical strain to the regulation of Fn conformation. We achieved specificity in quantifying conformational changes in this region of Fn by measuring the ratio of two fluorescent monoclonal antibodies, one that is insensitive to Fn conformational changes and a second whose binding is reduced or enhanced by non-equilibrium conformational changes. Importantly, this technique is shown to work on Fn adsorbed on surfaces, single Fn fibers, and Fn matrix fibers in cell culture. Using our dual antibody approach, we show that heparin and mechanical strain co-regulate Fn conformation in matrix fibrils, which is the first demonstration of heparin-dependent regulation of Fn in its physiologically-relevant fibrillar state. Furthermore, the dual antibody approach utilizes commercially available antibodies and simple immunohistochemistry, thus making it accessible to a wide range of scientists interested in Fn mechanobiology.


Subject(s)
Antibodies/immunology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Heparin/metabolism , Binding Sites , Cell Adhesion/genetics , Extracellular Matrix/chemistry , Extracellular Matrix/immunology , Fibronectins/chemistry , Fibronectins/immunology , Heparin/chemistry , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Humans , Protein Binding , Protein Conformation , Protein Structure, Tertiary
10.
Tissue Cell ; 45(4): 253-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23648172

ABSTRACT

Extracellular matrix remodeling is a continuous process that is critical to maintaining tissue homeostasis, and alterations in this process have been implicated in chronic diseases such as atherosclerosis, lung fibrosis, and emphysema. Collagen and elastin are subject to ascorbate-dependent hydroxylation. While this post-translational modification in collagen is critical for function, the role of hydroxylation of elastin is not well understood. A number of studies have indicated that ascorbate leads to reduced elastin synthesis. However, these studies were limited to analysis of cells grown under traditional 2D tissue culture conditions. To investigate this process we evaluated elastin and collagen synthesis in primary rat neonatal pulmonary fibroblasts in response to ascorbate treatment in traditional 2D culture and within 3D cross-linked gelatin matrices (Gelfoam). We observed little change in elastin or collagen biosynthesis in standard 2D cultures treated with ascorbate, yet observed a dramatic increase in elastin protein and mRNA levels in response to ascorbate in 3D cell-Gelfoam constructs. These data suggest that the cell-ECM architecture dictates pulmonary cell response to ascorbate, and that approaches aimed toward stimulating ECM repair or engineering functional cell-derived matrices should consider all aspects of the cellular environment.


Subject(s)
Collagen/biosynthesis , Elastin/biosynthesis , Fibroblasts/cytology , Tissue Engineering , Animals , Ascorbic Acid/administration & dosage , Ascorbic Acid/metabolism , Embryonic Development , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Hydroxylation , Lung/cytology , Primary Cell Culture , Protein Processing, Post-Translational , Rats
11.
Biomatter ; 3(3)2013.
Article in English | MEDLINE | ID: mdl-23628870

ABSTRACT

A broad range of cells are subjected to irregular time varying mechanical stimuli within the body, particularly in the respiratory and circulatory systems. Mechanical stretch is an important factor in determining cell function; however, the effects of variable stretch remain unexplored. In order to investigate the effects of variable stretch, we designed, built and tested a uniaxial stretching device that can stretch three-dimensional tissue constructs while varying the strain amplitude from cycle to cycle. The device is the first to apply variable stretching signals to cells in tissues or three dimensional tissue constructs. Following device validation, we applied 20% uniaxial strain to Gelfoam samples seeded with neonatal rat lung fibroblasts with different levels of variability (0%, 25%, 50% and 75%). RT-PCR was then performed to measure the effects of variable stretch on key molecules involved in cell-matrix interactions including: collagen 1α, lysyl oxidase, α-actin, ß1 integrin, ß3 integrin, syndecan-4, and vascular endothelial growth factor-A. Adding variability to the stretching signal upregulated, downregulated or had no effect on mRNA production depending on the molecule and the amount of variability. In particular, syndecan-4 showed a statistically significant peak at 25% variability, suggesting that an optimal variability of strain may exist for production of this molecule. We conclude that cycle-by-cycle variability in strain influences the expression of molecules related to cell-matrix interactions and hence may be used to selectively tune the composition of tissue constructs.


Subject(s)
Equipment Design/instrumentation , Extracellular Matrix/genetics , Fibroblasts/metabolism , Mechanotransduction, Cellular , RNA, Messenger/analysis , Animals , Biomechanical Phenomena/genetics , Biomechanical Phenomena/physiology , Collagen/chemistry , Equipment Design/methods , Extracellular Matrix/physiology , Gene Expression , Lung/cytology , Rats , Reproducibility of Results , Stress, Mechanical , Tissue Engineering
12.
J Cell Physiol ; 227(11): 3693-700, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22378222

ABSTRACT

Vascular endothelial growth factor A (VEGF-A) is a promoter of neovascularization and thus a popular therapeutic target for diseases involving excessive growth of blood vessels. In this study, we explored the potential of the disaccharide sucrose octasulfate (SOS) to alter VEGF165 diffusion through Descemet's membrane. Descemet's membranes were isolated from bovine eyes and used as a barrier between two chambers of a diffusion apparatus to measure VEGF transport. Diffusion studies revealed a dramatic increase in VEGF165 transport in the presence of SOS, with little diffusion of VEGF165 across the membrane over a 10-h time course in the absence of SOS. Diffusion studies with VEGF121, a non-heparin binding variant of VEGF, showed robust diffusion with or without SOS. To determine a possible mechanism, we measured the ability of SOS to inhibit VEGF interactions with extracellular matrix (ECM), using cell-free and cell surface binding assays. Binding studies showed SOS had no effect on VEGF165 binding to either heparin-coated plates or endothelial cell surfaces at less than mg/ml concentrations. In contrast, we show that SOS inhibited VEGF165 binding to fibronectin in a dose dependent manner and dramatically accelerated the rate of release of VEGF165 from fibronectin. SOS also inhibited the binding of VEGF165 to fibronectin-rich ECM deposited by vascular smooth muscle cells. These results suggest that fibronectin-rich extracellular matrices serve as barriers to VEGF165 diffusion by providing a network of binding sites that can trap and sequester the protein. Since the content of Descemet's membrane is typical of many basement membranes it is possible that they serve throughout the body as formidable barriers to VEGF165 diffusion and tightly regulate its bioavailability and distribution within tissues.


Subject(s)
Descemet Membrane , Facilitated Diffusion/drug effects , Sucrose/analogs & derivatives , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Cells, Cultured , Descemet Membrane/drug effects , Descemet Membrane/metabolism , Diffusion Chambers, Culture , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Protein Binding/drug effects , Sucrose/chemistry , Sucrose/pharmacology , Vascular Endothelial Growth Factor A/chemistry
13.
J Biol Chem ; 286(22): 19311-9, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21471211

ABSTRACT

Human fibroblast growth factor-2 (FGF2) regulates cellular processes including proliferation, adhesion, motility, and angiogenesis. FGF2 exerts its biological function by binding and dimerizing its receptor (FGFR), which activates signal transduction cascades. Effective binding of FGF2 to its receptor requires the presence of heparan sulfate (HS), a linear polysaccharide with N-sulfated domains (NS) localized at the cell surface and extracellular matrix. HS acts as a platform facilitating the formation of a functional FGF-FGFR-HS ternary complex. Crystal structures of the signaling ternary complex revealed two conflicting architectures. In the asymmetrical model, two FGFs and two FGFRs bind a single HS chain. In contrast, the symmetrical model postulates that one FGF and one FGFR bind to the free end of the HS chain and dimerization require these ends to join, bringing the two half-complexes together. In this study, we screened a hexasaccharide HS library for compositions that are able to bind FGF2. The library was composed primarily of NS domains internal to the HS chain with minor presence of non-reducing end (NRE) NS. The binders were categorized into low versus high affinity binders. The low affinity fraction contained primarily hexasaccharides with low degree of sulfation that were internal to the HS chains. In contrast, the high affinity bound fraction was enriched in NRE oligosaccharides that were considerably more sulfated and had the ability to promote FGFR-mediated cell proliferation. The results suggest a role of the NRE of HS in FGF2 signaling and favor the formation of the symmetrical architecture on short NS domains.


Subject(s)
Fibroblast Growth Factor 2/chemistry , Heparitin Sulfate/chemistry , Oligosaccharides/chemistry , Animals , Cell Line , Cell Proliferation , Crystallography, X-Ray , Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Humans , Oligosaccharides/genetics , Oligosaccharides/metabolism , Protein Structure, Tertiary , Receptors, Fibroblast Growth Factor , Signal Transduction/physiology , Swine
14.
PLoS One ; 5(2): e9389, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20186334

ABSTRACT

BACKGROUND: Increasing evidence has revealed important roles for complex glycans as mediators of normal and pathological processes. Glycosaminoglycans are a class of glycans that bind and regulate the function of a wide array of proteins at the cell-extracellular matrix interface. The specific sequence and chemical organization of these polymers likely define function; however, identification of the structure-function relationships of glycosaminoglycans has been met with challenges associated with the unique level of complexity and the nontemplate-driven biosynthesis of these biopolymers. METHODOLOGY/PRINCIPAL FINDINGS: To address these challenges, we have devised a computational approach to predict fine structure and patterns of domain organization of the specific glycosaminoglycan, heparan sulfate (HS). Using chemical composition data obtained after complete and partial digestion of mixtures of HS chains with specific degradative enzymes, the computational analysis produces populations of theoretical HS chains with structures that meet both biosynthesis and enzyme degradation rules. The model performs these operations through a modular format consisting of input/output sections and three routines called chainmaker, chainbreaker, and chainsorter. We applied this methodology to analyze HS preparations isolated from pulmonary fibroblasts and epithelial cells. Significant differences in the general organization of these two HS preparations were observed, with HS from epithelial cells having a greater frequency of highly sulfated domains. Epithelial HS also showed a higher density of specific HS domains that have been associated with inhibition of neutrophil elastase. Experimental analysis of elastase inhibition was consistent with the model predictions and demonstrated that HS from epithelial cells had greater inhibitory activity than HS from fibroblasts. CONCLUSIONS/SIGNIFICANCE: This model establishes the conceptual framework for a new class of computational tools to use to assess patterns of domain organization within glycosaminoglycans. These tools will provide a means to consider high-level chain organization in deciphering the structure-function relationships of polysaccharides in biology.


Subject(s)
Disaccharides/chemistry , Glycosaminoglycans/chemistry , Models, Chemical , Software , Algorithms , Animals , Binding Sites , Cell Line , Cells, Cultured , Disaccharides/analysis , Disaccharides/metabolism , Epithelial Cells/chemistry , Epithelial Cells/cytology , Fibroblasts/chemistry , Fibroblasts/cytology , Fourier Analysis , Glycosaminoglycans/analysis , Glycosaminoglycans/metabolism , Heparin Lyase/metabolism , Heparitin Sulfate/analysis , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Hexuronic Acids/analysis , Hexuronic Acids/chemistry , Molecular Structure , Rats , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship
15.
Proc Natl Acad Sci U S A ; 106(4): 1081-6, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19144920

ABSTRACT

Mechanical failure of soft tissues is characteristic of life-threatening diseases, including capillary stress failure, pulmonary emphysema, and vessel wall aneurysms. Failure occurs when mechanical forces are sufficiently high to rupture the enzymatically weakened extracellular matrix (ECM). Elastin, an important structural ECM protein, is known to stretch beyond 200% strain before failing. However, ECM constructs and native vessel walls composed primarily of elastin and proteoglycans (PGs) have been found to fail at much lower strains. In this study, we hypothesized that PGs significantly contribute to tissue failure. To test this, we developed a zipper network model (ZNM), in which springs representing elastin are organized into long wavy fibers in a zipper-like formation and placed within a network of springs mimicking PGs. Elastin and PG springs possessed distinct mechanical and failure properties. Simulations using the ZNM showed that the failure of PGs alone reduces the global failure strain of the ECM well below that of elastin, and hence, digestion of elastin does not influence the failure strain. Network analysis suggested that whereas PGs drive the failure process and define the failure strain, elastin determines the peak and failure stresses. Predictions of the ZNM were experimentally confirmed by measuring the failure properties of engineered elastin-rich ECM constructs before and after digestion with trypsin, which cleaves the core protein of PGs without affecting elastin. This study reveals a role for PGs in the failure properties of engineered and native ECM with implications for the design of engineered tissues.


Subject(s)
Extracellular Matrix/chemistry , Models, Biological , Animals , Biomechanical Phenomena , Computer Simulation , Elasticity , Elastin/chemistry , Proteoglycans/chemistry , Rats , Rats, Sprague-Dawley , Time Factors
16.
Ann Biomed Eng ; 36(12): 2134-48, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18839312

ABSTRACT

Growth factor binding to transmembrane protein receptors is generally understood to initiate cell signaling. Receptor binding of heparin-binding growth factors (HB-GFs), such as fibroblast growth factor-2 (FGF-2), is regulated by interactions with heparan sulfate proteoglycans. While there is some specificity for binding to heparan sulfate, overlap in sites for different growth factors may allow for cross regulation. Here we demonstrate, using experiments and computer simulations, that the HB-GFs FGF-2 and heparin-binding EGF-like growth factor (HB-EGF) can cross regulate receptor binding of the other despite having unique receptors. The ability of HSPG to stabilize HB-GF receptor binding is critical for competing growth factors to modulate receptor binding with both enhanced and reduced binding possible depending on this stabilization process. HSPG density and affinity for HB-GF are also critical factors for HB-GF cross regulation. Simulations further reveal that HB-GF can regulate receptor binding of non-HB-GFs such as EGF even when the two proteins share no binding sites when other HB-GF are present within the network. Proliferation studies demonstrate potentiation of HB-EGF-induced growth by FGF-2 indicating that competition networks can alter biological response. Exogenous manipulation of cellular responses to growth factors in complex living systems will require understanding the HSPG-controlled network.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Models, Biological , Myocytes, Smooth Muscle/metabolism , Acid Phosphatase/metabolism , Animals , Aorta/cytology , Cattle , Cell Count , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fibroblast Growth Factor 2/genetics , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Iodine Radioisotopes/metabolism , Myocytes, Smooth Muscle/enzymology , Recombinant Proteins/metabolism
17.
J Cell Biochem ; 105(1): 108-20, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18459114

ABSTRACT

Histone acetyltransferases (HATs) are a class of enzymes that participate in modulating chromatin structure and gene expression. Altered HAT activity has been implicated in a number of diseases, yet little is known about the regulation of HATs. In this study, we report that glycosaminoglycans (GAGs) are potent inhibitors of p300 and pCAF HAT activities in vitro, with heparin and heparan sulfate proteoglycans (HSPGs) being the most potent inhibitors. The mechanism of inhibition by heparin was investigated. The ability of heparin to inhibit HAT activity was in part dependent upon its size and structure, as small heparin-derived oligosaccharides (>8 sugars) and N-desulfated or O-desulfated heparin showed reduced inhibitory activity. Heparin was shown to bind to pCAF; and enzyme assays indicated that heparin shows the characteristics of a competitive-like inhibitor causing an approximately 50-fold increase in the apparent Km of pCAF for histone H4. HSPGs isolated from corneal and pulmonary fibroblasts inhibited HAT activity with similar effectiveness as heparin. As evidence that endogenous GAGs might be involved in modulating histone acetylation, the direct addition of heparin to pulmonary fibroblasts resulted in an approximately 50% reduction of histone H3 acetylation after 6 h of treatment. In addition, Chinese hamster ovary cells deficient in GAG synthesis showed increased levels of acetylated histone H3 compared to wild-type parent cells. GAGs represent a new class of HAT inhibitors that might participate in modulating cell function by regulating histone acetylation.


Subject(s)
Glycosaminoglycans/pharmacology , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Acetylation , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cricetinae , Eye/metabolism , Fibroblasts , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Lung/drug effects , Lung/metabolism , Substrate Specificity
18.
Am J Respir Cell Mol Biol ; 31(3): 344-50, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15191913

ABSTRACT

Uncontrolled elastase activity is involved in the development of several types of lung disease. Previous reports demonstrated that growth factors are liberated from pulmonary matrix storage sites by elastase; however, release of these entities in vivo is not well defined. In the present study, we investigated the release of fibroblast growth factor-2 (FGF-2) and transforming growth factor-beta (TGF-beta), after intratracheal instillation of porcine pancreatic elastase into mice. We found that elastase promoted a time-dependent release of FGF-2 and TGF-beta1 from the lung into bronchoalveolar lavage (BAL) fluid. A large fraction of the TGF-beta1 in BAL fluid was in the active form (approximately 60%), suggesting that elastase might participate in the activation of TGF-beta1 from its latent form. Analysis of the levels of FGF-2 and TGF-beta1 in mouse blood indicated that the growth factors in BAL fluid were not entirely derived from blood. Moreover, elastase treatment of pulmonary fibroblasts cultures caused the release of TGF-beta1, suggesting that the TGF-beta1 in BAL fluid could have come from lung cells/matrix. Additional in vitro studies also indicated that TGF-beta1 plays a role in upregulating elastin mRNA levels. These data suggest that elastase releases growth factors from lung that participate in elastolytic injury responses.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Lung/enzymology , Lung/metabolism , Pancreatic Elastase/metabolism , Transforming Growth Factor beta/metabolism , Animals , Blood Proteins/metabolism , Cells, Cultured , Elastin/biosynthesis , Elastin/genetics , Female , Fibroblast Growth Factor 2/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Lung/physiopathology , Mice , Pancreatic Elastase/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Reaction Time/drug effects , Reaction Time/physiology , Transforming Growth Factor beta/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Biochem J ; 379(Pt 2): 331-41, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-14717658

ABSTRACT

We investigated how lipid raft association of HSPG (heparan sulphate proteoglycans) modulates FGF-2 (fibroblast growth factor-2/basic fibroblast growth factor) interactions with vascular smooth-muscle cells. When lipid rafts were disrupted with sterol-binding agents, methyl-beta-cyclodextrin and filipin, FGF-2 binding to HSPG was reduced 2-5-fold, yet the amount and turnover of cell-surface HSPG were unaffected [corrected]. Approx. 50-65% of bound FGF-2 was in lipid raft-associated fractions based on insolubility in cold Triton X-100 and flotation in OptiPrep density gradients, and this level was increased with higher FGF-2 concentrations [corrected]. Less FGF-2 (50-90%) was associated in raft fractions when cholesterol was depleted or HSPG were degraded with heparinase III. To investigate how lipid raft-HSPG interactions altered binding, we compared the rates of FGF-2 dissociation with native, MbetaCD (methyl-beta-cyclodextrin)- and filipin-treated cells. We found that FGF-2 dissociation rates were increased when lipid rafts were disrupted. These results suggest that localization of HSPG within lipid rafts creates high local concentrations of binding sites such that dissociation of FGF-2 is hindered. The localization of FGF-2 and HSPG to lipid rafts also correlated with the activation of protein kinase Calpha. Thus raft association of HSPG might create growth factor traps resulting in increased binding and signal transduction to enhance cell sensitivity.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Membrane Microdomains/metabolism , Muscle, Smooth, Vascular/metabolism , beta-Cyclodextrins , Animals , Binding Sites , Cattle , Cyclodextrins/pharmacology , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/pharmacology , Filipin/pharmacology , Heparan Sulfate Proteoglycans/analysis , Membrane Microdomains/chemistry , Muscle, Smooth, Vascular/cytology , Protein Kinase C/metabolism , Protein Kinase C-alpha , Proteoglycans/metabolism
20.
Am J Physiol Lung Cell Mol Physiol ; 285(5): L1106-15, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12882762

ABSTRACT

Elastase degradation of elastin within alveolar walls is an important event in the development of pulmonary emphysema. In addition to elastolytic activities, elastases release growth factors from extracellular matrices and interstitial cell surfaces that can regulate elastogenesis and other cellular responses. In the present study, we demonstrate that brief treatment of matrix-laden rat pulmonary fibroblast cultures with pancreatic elastase results in the release of soluble heparin-binding epidermal growth factor-like growth factor (HB-EGF) concomitant with a decrease in HB-EGF binding to both heparan sulfate proteoglycan and receptor sites on the cells. In undigested, matrix-laden fibroblasts, HB-EGF significantly downregulates elastin mRNA via activation of epidermal growth factor receptor. Results from nuclear run-on analyses show that HB-EGF downregulates elastin mRNA via transcriptional suppression. HBEGF treatment stimulates MAP or ERK kinase (MEK)-dependent ERK1/2 phosphorylation and leads to nuclear accumulation of Fra-1. Blocking ERK1/2 activation by MEK1/2 inhibitors (PD-98059 or U-0126) diminishes HB-EGF-induced Fra-1 accumulation and subsequent downregulation of elastin mRNA. Coaddition of two elastase-released growth factors, HB-EGF and FGF-2, results in an additive inhibitory effect on elastin mRNA levels. Furthermore, HB-EGF addition to pulmonary fibroblasts increases FGF-2 mRNA and protein levels. These data suggest that HB-EGF and FGF-2 act in concert to regulate the synthesis of elastin in injury/repair situations.


Subject(s)
Elastin/genetics , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/genetics , Gene Expression Regulation/drug effects , Lung/physiology , Animals , Animals, Newborn , Binding Sites , Cells, Cultured , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacokinetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Flavonoids/pharmacology , Heparin/pharmacology , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Kinetics , Lung/cytology , Lung/drug effects , Pancreatic Elastase/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...