Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cells ; 11(5)2022 03 02.
Article in English | MEDLINE | ID: mdl-35269476

ABSTRACT

Severe acute respiratory syndrome virus 2 (SARS-CoV2) has infected an estimated 400 million people world-wide, causing approximately 6 million deaths from severe coronavirus disease 2019 (COVID-19). The SARS-CoV2 Spike protein plays a critical role in viral attachment and entry into host cells. The recent emergence of highly transmissible variants of SARS-CoV2 has been linked to mutations in Spike. This review provides an overview of the structure and function of Spike and describes the factors that impact Spike's ability to mediate viral infection as well as the potential limits to how good (or bad) Spike protein can become. Proposed here is a framework that considers the processes of Spike-mediated SARS-CoV2 attachment, dissociation, and cell entry where the role of Spike, from the standpoint of the virus, is to maximize cell entry with each viral-cell collision. Key parameters are identified that will be needed to develop models to identify mechanisms that new Spike variants might exploit to enhance viral transmission. In particular, the importance of considering secondary co-receptors for Spike, such as heparan sulfate proteoglycans is discussed. Accurate models of Spike-cell interactions could contribute to the development of new therapies in advance of the emergence of new highly transmissible SARS-CoV2 variants.


Subject(s)
COVID-19 , Pandemics , Humans , RNA, Viral , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry
2.
Int J Mol Sci ; 22(2)2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33466887

ABSTRACT

The binding of vascular endothelial growth factor A (VEGF) to VEGF receptor-2 (VEGFR-2) stimulates angiogenic signaling. Lipid rafts are cholesterol-dense regions of the plasma membrane that serve as an organizational platform for biomolecules. Although VEGFR2 has been shown to colocalize with lipid rafts to regulate its activation, the effect of lipid rafts on non-activated VEGFR2 has not been explored. Here, we characterized the involvement of lipid rafts in modulating the stability of non-activated VEGFR2 in endothelial cells using raft disrupting agents: methyl-ß-cyclodextrin, sphingomyelinase and simvastatin. Disrupting lipid rafts selectively decreased the levels of non-activated VEGFR2 as a result of increased lysosomal degradation. The decreased expression of VEGFR2 translated to reduced VEGF-activation of the extracellular signal-regulated protein kinases (ERK). Overall, our results indicate that lipid rafts stabilize VEGFR2 and its associated signal transduction activities required for angiogenesis. Thus, modulation of lipid rafts may provide a means to regulate the sensitivity of endothelial cells to VEGF stimulation. Indeed, the ability of simvastatin to down regulate VEGFR2 and inhibit VEGF activity suggest a potential mechanism underlying the observation that this drug improves outcomes in the treatment of certain cancers.


Subject(s)
Endothelial Cells/metabolism , Membrane Microdomains/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Anticholesteremic Agents/pharmacology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiology , Cattle , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Membrane Microdomains/drug effects , Protein Binding/drug effects , Protein Stability/drug effects , Signal Transduction/drug effects , Simvastatin/pharmacology , Vascular Endothelial Growth Factor A/metabolism
3.
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
4.
J Biol Chem ; 294(46): 17603-17611, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31601651

ABSTRACT

Vascular endothelial growth factor-A (VEGF) plays a critical role in stimulating angiogenesis in normal and disease states. Anti-VEGF antibodies have been developed to manage pathological angiogenesis. Bevacizumab, sold under the brand name Avastin, is a humanized mAb that binds VEGF and blocks its binding to its signaling receptor, VEGF receptor 2, and is used to treat patients with a variety of cancers or retinal disorders. The ability of Avastin to modulate other nonreceptor interactions of VEGF has not been fully defined. In this study, we investigated Avastin's capacity to modulate VEGF165 binding to porcine aortic endothelial cells and to heparin and fibronectin (FN) across a range of pH values (pH 5-8). We observed that Avastin slightly enhanced VEGF binding to heparin and that heparin increased VEGF binding to Avastin. In contrast, Avastin inhibited VEGF binding to cells and FN, yet Avastin could still bind to VEGF that was bound to FN, indicating that these binding events are not mutually exclusive. Avastin binding to VEGF was dramatically reduced at acidic pH values (pH 5.0-6.5), whereas VEGF binding to FN and nonreceptor sites on cells was enhanced. Interestingly, the reduced Avastin-VEGF binding at acidic pH was rescued by heparin, as was Avastin's ability to inhibit VEGF binding to cells. These results suggest that heparin might be used to expand the clinical utility of Avastin. Our findings highlight the importance of defining the range of VEGF interactions to fully predict antibody activity within a complex biological setting.


Subject(s)
Bevacizumab/pharmacology , Neovascularization, Pathologic/genetics , Vascular Endothelial Growth Factor A/genetics , Acids/chemistry , Animals , Aorta/drug effects , Aorta/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibronectins/genetics , Fibronectins/immunology , Heparin/pharmacology , Humans , Hydrogen-Ion Concentration , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Protein Binding/drug effects , Swine , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology
5.
Anal Biochem ; 564-565: 21-31, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30292477

ABSTRACT

Angiogenesis is a highly regulated process orchestrated, in large part, by the vascular endothelial growth factor-A (VEGF-A) system of ligands and receptors. Considerable effort has been invested in finding optimal ways to modulate VEGF-A activity to treat disease, however, the mechanisms by which the various components interact remain poorly understood. This is in part because of the difficulty of analyzing the various interactions in an intercomparable manner. In the present study, we established conditions to allow for the detailed characterization of the molecular interactions between VEGF and its receptors and the co-receptor NRP-1 using surface plasmon resonance (SPR). We found that VEGF dissociated 25-times faster from its major signaling receptor, VEGF receptor-2 (VEGFR-2) than from its "decoy" receptor, VEGF receptor-1 (VEGFR-1). Using a systematic approach, we obtained kinetic parameters for each individual interaction under a consistent set of experimental conditions allowing for comparison between various receptors. The set of quantitative kinetic parameters and experimental conditions reported herein will provide valuable tools for developing comprehensive models of the VEGF system.


Subject(s)
Neuropilins/metabolism , Surface Plasmon Resonance/methods , Vascular Endothelial Growth Factor A/metabolism , Animals , Humans , Kinetics , Neuropilin-1/metabolism , Signal Transduction
6.
J Cell Physiol ; 231(9): 2026-39, 2016 09.
Article in English | MEDLINE | ID: mdl-26773314

ABSTRACT

Vascular endothelial growth factor A (VEGF) drives endothelial cell maintenance and angiogenesis. Endothelial cell behavior is altered by the stiffness of the substrate the cells are attached to suggesting that VEGF activity might be influenced by the mechanical cellular environment. We hypothesized that extracellular matrix (ECM) stiffness modifies VEGF-cell-matrix tethering leading to altered VEGF processing and signaling. We analyzed VEGF binding, internalization, and signaling as a function of substrate stiffness in endothelial cells cultured on fibronectin (Fn) linked polyacrylamide gels. Cell produced extracellular matrices on the softest substrates were least capable of binding VEGF, but the cells exhibited enhanced VEGF internalization and signaling compared to cells on all other substrates. Inhibiting VEGF-matrix binding with sucrose octasulfate decreased cell-internalization of VEGF and, inversely, heparin pre-treatment to enhance Fn-matrix binding of VEGF increased cell-internalization of VEGF regardless of matrix stiffness. ß1 integrins, which connect cells to Fn, modulated VEGF uptake in a stiffness dependent fashion. Cells on hard surfaces showed decreased levels of activated ß1 and inhibition of ß1 integrin resulted in a greater proportional decrease in VEGF internalization than in cells on softer matrices. Extracellular matrix binding is necessary for VEGF internalization. Stiffness modifies the coordinated actions of VEGF-matrix binding and ß1 integrin binding/activation, which together are critical for VEGF internalization. This study provides insight into how the microenvironment may influence tissue regeneration and response to injury and disease. J. Cell. Physiol. 231: 2026-2039, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Cell Movement , Cells, Cultured , Fibronectins/metabolism , Humans
7.
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
8.
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
9.
Integr Biol (Camb) ; 7(9): 1011-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26183123

ABSTRACT

Vascular disease and its associated complications are the number one cause of death in the Western world. Both extracellular matrix stiffening and dysfunctional endothelial cells contribute to vascular disease. We examined endothelial cell calcium signaling in response to VEGF as a function of extracellular matrix stiffness. We developed a new analytical tool to analyze both population based and individual cell responses. Endothelial cells on soft substrates, 4 kPa, were the most responsive to VEGF, whereas cells on the 125 kPa substrates exhibited an attenuated response. Magnitude of activation, not the quantity of cells responding or the number of local maximums each cell experienced distinguished the responses. Individual cell analysis, across all treatments, identified two unique cell clusters. One cluster, containing most of the cells, exhibited minimal or slow calcium release. The remaining cell cluster had a rapid, high magnitude VEGF activation that ultimately defined the population based average calcium response. Interestingly, at low doses of VEGF, the high responding cell cluster contained smaller cells on average, suggesting that cell shape and size may be indicative of VEGF-sensitive endothelial cells. This study provides a new analytical tool to quantitatively analyze individual cell signaling response kinetics, that we have used to help uncover outcomes that are hidden within the average. The ability to selectively identify highly VEGF responsive cells within a population may lead to a better understanding of the specific phenotypic characteristics that define cell responsiveness, which could provide new insight for the development of targeted anti- and pro-angiogenic therapies.


Subject(s)
Calcium Signaling/physiology , Cell Communication/physiology , Endothelial Cells/physiology , Extracellular Matrix/physiology , Mechanotransduction, Cellular/physiology , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Calcium Signaling/drug effects , Cattle , Cell Communication/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Elastic Modulus/drug effects , Elastic Modulus/physiology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Extracellular Matrix/drug effects , Mechanotransduction, Cellular/drug effects , Stress, Mechanical
10.
J Biol Chem ; 290(26): 16451-62, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25979342

ABSTRACT

Angiogenesis is a highly regulated process orchestrated by the VEGF system. Heparin/heparan sulfate proteoglycans and neuropilin-1 (NRP-1) have been identified as co-receptors, yet the mechanisms of action have not been fully defined. In the present study, we characterized molecular interactions between receptors and co-receptors, using surface plasmon resonance and in vitro binding assays. Additionally, we demonstrate that these binding events are relevant to VEGF activity within endothelial cells. We defined interactions and structural requirements for heparin/HS interactions with VEGF receptor (VEGFR)-1, NRP-1, and VEGF165 in complex with VEGFR-2 and NRP-1. We demonstrate that these structural requirements are distinct for each interaction. We further show that VEGF165, VEGFR-2, and monomeric NRP-1 bind weakly to heparin alone yet show synergistic binding to heparin when presented together in various combinations. This synergistic binding appears to translate to alterations in VEGF signaling in endothelial cells. We found that soluble NRP-1 increases VEGF binding and activation of VEGFR-2 and ERK1/2 in endothelial cells and that these effects require sulfated HS. These data suggest that the presence of HS/heparin and NRP-1 may dictate the specific receptor type activated by VEGF and ultimately determine the biological output of the system. The ability of co-receptors to fine-tune VEGF responsiveness suggests the possibility that VEGF-mediated angiogenesis can be selectively stimulated or inhibited by targeting HS/heparin and NRP-1.


Subject(s)
Heparin/metabolism , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Kinetics , Mice , Neuropilin-1/chemistry , Neuropilin-1/genetics , Protein Binding , Signal Transduction , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/chemistry , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-2/genetics
11.
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
12.
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
13.
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
14.
J Chem Inf Model ; 54(7): 2068-78, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24974889

ABSTRACT

Many proteins of widely differing functionality and structure are capable of binding heparin and heparan sulfate. Since crystallizing protein-heparin complexes for structure determination is generally difficult, computational docking can be a useful approach for understanding specific interactions. Previous studies used programs originally developed for docking small molecules to well-defined pockets, rather than for docking polysaccharides to highly charged shallow crevices that usually bind heparin. We have extended the program PIPER and the automated protein-protein docking server ClusPro to heparin docking. Using a molecular mechanics energy function for scoring and the fast Fourier transform correlation approach, the method generates and evaluates close to a billion poses of a heparin tetrasaccharide probe. The docked structures are clustered using pairwise root-mean-square deviations as the distance measure. It was shown that clustering of heparin molecules close to each other but having different orientations and selecting the clusters with the highest protein-ligand contacts reliably predicts the heparin binding site. In addition, the centers of the five most populated clusters include structures close to the native orientation of the heparin. These structures can provide starting points for further refinement by methods that account for flexibility such as molecular dynamics. The heparin docking method is available as an advanced option of the ClusPro server at http://cluspro.bu.edu/ .


Subject(s)
Heparin/metabolism , Molecular Docking Simulation , Proteins/chemistry , Proteins/metabolism , Binding Sites , Heparitin Sulfate/metabolism , Humans , Monte Carlo Method , Protein Conformation , Solvents/chemistry
15.
Am J Physiol Cell Physiol ; 306(10): C972-85, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24671101

ABSTRACT

The process of wound healing must be tightly regulated to achieve successful restoration of injured tissue. Previously, we demonstrated that when corneal epithelium is injured, nucleotides and neuronal factors are released to the extracellular milieu, generating a Ca(2+) wave from the origin of the wound to neighboring cells. In the present study we sought to determine how the communication between epithelial cells in the presence or absence of neuronal wound media is affected by hypoxia. A signal-sorting algorithm was developed to determine the dynamics of Ca(2+) signaling between neuronal and epithelial cells. The cross talk between activated corneal epithelial cells in response to neuronal wound media demonstrated that injury-induced Ca(2+) dynamic patterns were altered in response to decreased O2 levels. These alterations were associated with an overall decrease in ATP and changes in purinergic receptor-mediated Ca(2+) mobilization and localization of N-methyl-d-aspartate receptors. In addition, we used the cornea in an organ culture wound model to examine how hypoxia impedes reepithelialization after injury. There was a change in the recruitment of paxillin to the cell membrane and deposition of fibronectin along the basal lamina, both factors in cell migration. Our results provide evidence that complex Ca(2+)-mediated signaling occurs between sensory neurons and epithelial cells after injury and is critical to wound healing. Information revealed by these studies will contribute to an enhanced understanding of wound repair under compromised conditions and provide insight into ways to effectively stimulate proper epithelial repair.


Subject(s)
Calcium/metabolism , Cornea/metabolism , Epithelial Cells/metabolism , Oxygen/metabolism , Trigeminal Ganglion/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Communication , Cell Hypoxia/genetics , Cell Line , Cell Movement/drug effects , Coculture Techniques , Cornea/drug effects , Corneal Injuries , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Oxygen/pharmacology , Paxillin/genetics , Paxillin/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Re-Epithelialization/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/injuries
16.
J Biol Chem ; 289(5): 2526-36, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24344132

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal lung disease characterized by the overgrowth, hardening, and scarring of lung tissue. The exact mechanisms of how IPF develops and progresses are unknown. IPF is characterized by extracellular matrix remodeling and accumulation of active TGFß, which promotes collagen expression and the differentiation of smooth muscle α-actin (SMA)-positive myofibroblasts. Aortic carboxypeptidase-like protein (ACLP) is an extracellular matrix protein secreted by fibroblasts and myofibroblasts and is expressed in fibrotic human lung tissue and in mice with bleomycin-induced fibrosis. Importantly, ACLP knockout mice are significantly protected from bleomycin-induced fibrosis. The goal of this study was to identify the mechanisms of ACLP action on fibroblast differentiation. As primary lung fibroblasts differentiated into myofibroblasts, ACLP expression preceded SMA and collagen expression. Recombinant ACLP induced SMA and collagen expression in mouse and human lung fibroblasts. Knockdown of ACLP slowed the fibroblast-to-myofibroblast transition and partially reverted differentiated myofibroblasts by reducing SMA expression. We hypothesized that ACLP stimulates myofibroblast formation partly through activating TGFß signaling. Treatment of fibroblasts with recombinant ACLP induced phosphorylation and nuclear translocation of Smad3. This phosphorylation and induction of SMA was dependent on TGFß receptor binding and kinase activity. ACLP-induced collagen expression was independent of interaction with the TGFß receptor. These findings indicate that ACLP stimulates the fibroblast-to-myofibroblast transition by promoting SMA expression via TGFß signaling and promoting collagen expression through a TGFß receptor-independent pathway.


Subject(s)
Carboxypeptidases/metabolism , Fibroblasts/cytology , Idiopathic Pulmonary Fibrosis/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Carboxypeptidases/genetics , Cell Differentiation/physiology , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Fibroblasts/metabolism , HEK293 Cells , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mink , Primary Cell Culture , Repressor Proteins/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...