heparitin-sulfate has been researched along with Inflammation* in 92 studies
31 review(s) available for heparitin-sulfate and Inflammation
Article | Year |
---|---|
The Challenge of Modulating Heparan Sulfate Turnover by Multitarget Heparin Derivatives.
This review comes as a part of the special issue "Emerging frontiers in GAGs and mimetics". Our interest is in the manipulation of heparan sulfate (HS) turnover by employing HS mimetics/heparin derivatives that exert pleiotropic effects and are interesting for interfering at multiple levels with pathways in which HS is implicated. Due to the important role of heparanase in HS post-biosynthetic modification and catabolism, we focus on the possibility to target heparanase, at both extracellular and intracellular levels, a strategy that can be applied to many conditions, from inflammation to cancer and neurodegeneration. Topics: Biomimetic Materials; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Neoplasms; Neurodegenerative Diseases | 2020 |
Involvement of Syndecan-1 and Heparanase in Cancer and Inflammation.
The cell surface heparan sulfate proteoglycan Syndecan-1 acts as an important co-receptor for receptor tyrosine kinases and chemokine receptors, and as an adhesion receptor for structural glycoproteins of the extracellular matrix. It serves as a substrate for heparanase, an endo-β-glucuronidase that degrades specific domains of heparan sulfate carbohydrate chains and thereby alters the functional status of the proteoglycan and of Syndecan-1-bound ligands. Syndecan-1 and heparanase show multiple levels of functional interactions, resulting in mutual regulation of their expression, processing, and activity. These interactions are of particular relevance in the context of inflammation and malignant disease. Studies in animal models have revealed a mechanistic role of Syndecan-1 and heparanase in the regulation of contact allergies, kidney inflammation, multiple sclerosis, inflammatory bowel disease, and inflammation-associated tumorigenesis. Moreover, functional interactions between Syndecan-1 and heparanase modulate virtually all steps of tumor progression as defined in the Hallmarks of Cancer. Due to their prognostic value in cancer, and their mechanistic involvement in tumor progression, Syndecan-1 and heparanase have emerged as important drug targets. Data in preclinical models and preclinical phase I/II studies have already yielded promising results that provide a translational perspective. Topics: Animals; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Neoplasms; Syndecan-1 | 2020 |
HS and Inflammation: A Potential Playground for the Sulfs?
Heparan sulfate (HS) is a complex polysaccharide abundantly found in extracellular matrices and cell surfaces. HS participates in major cellular processes, through its ability to bind and modulate a wide array of signaling proteins. HS/ligand interactions involve saccharide domains of specific sulfation pattern. Assembly of such domains is orchestrated by a complex biosynthesis machinery and their structure is further regulated at the cell surface by post-synthetic modifying enzymes. Amongst them, extracellular sulfatases of the Sulf family catalyze the selective removal of 6- Topics: Animals; Heparitin Sulfate; Humans; Inflammation; Sulfatases; Sulfotransferases | 2020 |
Low molecular weight heparan sulfate containing facial skin care for reducing inflammation and restoring aged-skin homeostasis.
Chronic exposure of the skin to environmental factors, including solar radiation and air pollution, promotes skin aging by inducing inflammation which damages the skin. Heparan sulfate (HS) is one of the glycosaminoglycans, which has a central role in modulating skin repair. In its appearance in the skin, HS is large and highly polar and therefore unable to penetrate the skin. As a component in a topical formulation, the modified to a low molecular weight heparan sulfate analog (LMW-HS) showed biological modulation activity.. This review discusses the potential role of topical formulations containing LMW-HS in restoring aged-skin homeostasis and skin health.. An expert panel of dermatologists who regularly treat clinical signs of facial photoaging explored the role of LMW-HS containing formulations for reducing inflammation and facial-aging signs. For this purpose, evidence from the conducted literature searches was used together with expert opinion and experience of the panel.. Extrinsic factors contribute to skin aging through oxidative stress, stimulating inflammation involved in extracellular matrix degradation. Evidence showed that chemokines require heparan sulfate for their full range of functional activities during innate immunity. Studies showed the LMW-HS containing topical formulation to penetrate the skin within 48 hours of once-daily application. LMW-HS used in the periorbital area improved discoloration and wrinkles at Week 2 with continuous improvement up to Week 12.. The LMW-HS containing formulation showed improvements in skin condition when applied on photo-damaged skin, indicating its therapeutic potential. Topics: Aged; Heparitin Sulfate; Homeostasis; Humans; Inflammation; Molecular Weight; Skin Aging; Skin Care | 2020 |
Heparan sulfate as a regulator of inflammation and immunity.
Heparan sulfate is found on the surface of most cell types, as well as in basement membranes and extracellular matrices. Its strong anionic properties and highly variable structure enable this glycosaminoglycan to provide binding sites for numerous protein ligands, including many soluble mediators of the immune system, and may promote or inhibit their activity. The formation of ligand binding sites on heparan sulfate (HS) occurs in a tissue- and context-specific fashion through the action of several families of enzymes, most of which have multiple isoforms with subtly different specificities. Changes in the expression levels of these biosynthetic enzymes occur in response to inflammatory stimuli, resulting in structurally different HS and acquisition or loss of binding sites for immune mediators. In this review, we discuss the multiple roles for HS in regulating immune responses, and the evidence for inflammation-associated changes to HS structure. Topics: Animals; Cellular Microenvironment; Heparitin Sulfate; Humans; Immunity; Inflammation; Leukocytes; Phagocytosis | 2019 |
Heparan Sulfate Proteoglycans as Relays of Neuroinflammation.
Heparan sulfate proteoglycans (HSPGs) are implicated as inflammatory mediators in a variety of settings, including chemokine activation, which is required to recruit circulating leukocytes to infection sites. Heparan sulfate (HS) polysaccharide chains are highly interactive and serve co-receptor roles in multiple ligand:receptor interactions. HS may also serve as a storage depot, sequestering ligands such as cytokines and restricting their access to binding partners. Heparanase, through its ability to fragment HS chains, is a key regulator of HS function and has featured prominently in studies of HS's involvement in inflammatory processes. This review focuses on recent discoveries regarding the role of HSPGs, HS, and heparanase during inflammation, with particular focus on the brain. HS chains emerge as critical go-betweens in multiple aspects of the inflammatory response-relaying signals between receptors and cells. The molecular interactions proposed to occur between HSPGs and the pathogen receptor toll-like receptor 4 (TLR4) are discussed, and we summarize some of the contrasting roles that HS and heparanase have been assigned in diseases associated with chronic inflammatory states, including Alzheimer's disease (AD). We conclude by briefly discussing how current knowledge could potentially be applied to augment HS-mediated events during sustained neuroinflammation, which contributes to neurodegeneration in AD. Topics: Alzheimer Disease; Animals; Brain; Cytokines; Glucuronidase; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Immunity, Innate; Inflammation; Neuroglia; Neuroimmunomodulation; Toll-Like Receptor 4 | 2018 |
The Role of Heparan Sulfate in Inflammation, and the Development of Biomimetics as Anti-Inflammatory Strategies.
Key events that occur during inflammation include the recruitment, adhesion, and transmigration of leukocytes from the circulation to the site of inflammation. These events are modulated by chemokines, integrins, and selectins and the interaction of these molecules with glycosaminoglycans, predominantly heparan sulfate (HS). The development of HS/heparin mimetics that interfere or inhibit the interactions that occur between glycosaminoglycans and modulators of inflammation holds great potential for use as anti-inflammatory therapeutics. This review will detail the role of HS in the events that occur during inflammation, their interaction and modulation of inflammatory mediators, and the current advances in the development of HS/heparin mimetics as anti-inflammatory biotherapeutics. Topics: Animals; Anti-Inflammatory Agents; Biomimetics; Chemokines; Drug Discovery; Glucuronidase; Heparitin Sulfate; Humans; Immunity, Innate; Inflammation | 2018 |
New Advances of Heparanase and Heparanase-2 in Human Diseases.
As we all know, heparanase plays an important role in human diseases. As a kind of endo-β-glucuronidase, heparanase is the known only enzyme in mammals which could degrade heparan sulfate(HS) specifically. HS is a vital component of extracellular matrix(ECM). Heparanase takes effect by cleaving theβ(1,4)-glycosidic between glucosamine residue and glucuronic acid of HS. This cleavage will cause ECM remodelling and HS-linked biological molecules release, including cytokines, growth factors and a lot of biological molecules regulating various pathological activities. Experiments already proved that heparanase gene over-expresses in cancers of gastrointestinal tract, esophagus, breast and so on. Various studies have demonstrated the heparanase's pro-metastatic function and the reduced survival rate of patients could be indicated by high heparanase levels. Besides, pathological processes including procoagulant activities, preeclamptic placentas and inflammation are all verified to be associated with heparanase activity. In recent years, many functions other than pro-tumor effect was found in heparanase and worldwide researchers conducted varieties of experiments to identify the new function of this significant enzyme. Also, these newly-found functions are closely connected to certain cellular activities, for example epithelial to mesenchymal transition (EMT). It has already been demonstrated that EMT is related to some clinical disorders, like renal diseases. Given that heparanase is the only enzyme capable of this function, it could be concluded that heparanase would be a potential and valuable therapy target. This mini-review aims to retrospect literatures about heparanase published in 2017 and 2018 and provide a direction for therapy methods targeting heparanase. Topics: Animals; Cytokines; Epithelial-Mesenchymal Transition; Extracellular Matrix; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Kidney Diseases; Neoplasms | 2018 |
Heparanase regulation of cancer, autophagy and inflammation: new mechanisms and targets for therapy.
Because of its impact on multiple biological pathways, heparanase has emerged as a major regulator of cancer, inflammation and other disease processes. Heparanase accomplishes this by degrading heparan sulfate which regulates the abundance and location of heparin-binding growth factors thereby influencing multiple signaling pathways that control gene expression, syndecan shedding and cell behavior. In addition, heparanase can act via nonenzymatic mechanisms that directly activate signaling at the cell surface. Clinical trials testing heparanase inhibitors as anticancer therapeutics are showing early signs of efficacy in patients further emphasizing the biological importance of this enzyme. This review focuses on recent developments in the field of heparanase regulation of cancer and inflammation, including the impact of heparanase on exosomes and autophagy, and novel mechanisms whereby heparanase regulates tumor metastasis, angiogenesis and chemoresistance. In addition, the ongoing development of heparanase inhibitors and their potential for treating cancer and inflammation are discussed. Topics: Antineoplastic Agents; Autophagy; Clinical Trials as Topic; Drug Resistance, Neoplasm; Enzyme Inhibitors; Exosomes; Gene Expression Regulation, Neoplastic; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Molecular Targeted Therapy; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Syndecans | 2017 |
The Functions of Heparanase in Human Diseases.
The study of the heparanase has long been paid wide attention. Heparanase, an endo-β-D-glucuronidase, is capable of specifically degrading heparan sulfate (HS), one of the excellular matrix (ECM) components. It exerts its enzymatic activity catalyzing the cleavage of the β (1,4)-glycosidic bond between glucuronic acid and glucosamine residue. HS cleavage results in remodelling of the extracellular matrix as well as in regulating the release of many HS-linked molecules such as growth factors, cytokines and enzymes involved in inflammation, wound healing and tumour invasion. Varieties of experiments indicated that heparanase mRNA is overexpressed in human tumors, including breast cancer, gastrointestinal tumors, and esophageal carcinomas. A pro-metastatic and pro-angiogenic role for heparanase has been widely verified and high levels of heparanase correlate with reduced survival of cancer patients. Except protumor function, heparanase also plays a role in inflammation, angiogenesis, placentas and procoagulant activities. Heparanase is found to have many other functions in recent years, since many experiments have been carried out to identify this significant enzyme's new features. These newly found functions are related to the cellular activities such as autophagy and epithelial to mesenchymal transition (EMT). And together with other heparanase functions, autophagy and EMT are verified to be involved in several clinical disorders, for example, renal diseases. Considering that, once inactivated, there are no other enzymes capable of performing the same function, it is apparent that heparanase can be an effective and promising therapy target. This short review aims to establish the currently known function of this enzyme and provide evidence for heparanase targeted therapy. Topics: Autophagy; Epithelial-Mesenchymal Transition; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Kidney Diseases; Neoplasms; Prion Diseases; Scleroderma, Systemic | 2017 |
Heparan sulphate as a regulator of leukocyte recruitment in inflammation.
A key event in inflammatory disease is the transendothelial recruitment of leukocytes from the circulation to the site of inflammation. Intense research in the past decades indicates that the polyanionic carbohydrate heparan sulphate (HS) modulates multiple steps in the leukocyte recruitment cascade. Leukocyte recruitment is initiated by endothelial cell activation and presentation of chemokines to rolling leukocytes, which, via integrin activation, results in adhesion and diapedesis through the vessel wall. Heparan sulfate proteoglycans (HSPGs) immobilize the chemokines on the luminal endothelial cells, rendering them more robust against mechanical or hydrodynamic perturbations. During inflammation, endothelial HSPGs serve as ligands to L-selectin on leukocytes, transport chemokines in a basolateral to apical direction across the endothelium, and present chemokines at the luminal surface of the endothelium to circulating cells. HSPGs also promote chemokine oligomerization, which influences chemokine receptor signaling. Furthermore, proteoglycans of the syndecan family are involved in modulating integrin-mediated tight adhesion of leukocytes to the endothelium. Creation of a chemokine gradient by a localized chemokine release influences the speed of leukocyte recruitment from the blood to the tissue by attracting crawling neutrophils to optimal sites for transmigration. The directionality of intraluminal crawling is thought to be influenced by both mechanotactic and haptotactic signals, which are modulated by HS-dependent signaling processes. Finally, diapedesis is influenced by HS regarding transendothelial chemokine gradient formation and integrin- CAM interactions, and further enhanced by heparanase-mediated degradation of the endothelial basement membrane. Overall, the multifunctional role of HS in inflammation marks it as a potential target of glycan-centered therapeutic approaches. Topics: Animals; Cell Adhesion; Chemotaxis, Leukocyte; Heparitin Sulfate; Humans; Inflammation; Leukocyte Rolling; Leukocytes; Transendothelial and Transepithelial Migration | 2015 |
Recent data concerning heparanase: focus on fibrosis, inflammation and cancer.
Heparanase (HPSE) is a multitasking protein characterized by enzymatic and non-enzymatic activities. By means of its enzymatic activity, HPSE catalyzes the cutting of the side chains of heparan sulfate (HS) proteoglycans, thereby inducing the remodeling of the extracellular matrix and basement membranes. Thanks to the cleavage of HS, HPSE also promotes the release and diffusion of several HS-linked molecules such as growth factors, cytokines and enzymes. In addition to degrading HS chains, HPSE has non-enzymatic functions that trigger several signaling pathways. This signaling activity is achieved by interacting with transmembrane proteins, activating kinases such as Akt and Src, or modulating the activity of factors such as FGF-2 and TGF-β. Several studies have recently highlighted a possible intracellular activity for HPSE, particularly at nuclear level. While HPSE activity is quite limited in physiological conditions, its demonstrated increasing involvement in various pathological conditions, such as in tumor progression and renal disease, have attracted the attention of a growing number of researchers. The fact that no other molecule is capable of performing the same function as HPSE makes this enzyme an attractive potential target of medical treatment. With this short conceptual overview, we aim to provide an update on current knowledge concerning the HPSE protein in the experimental and clinical settings, paying particular attention to its role in fibrosis, inflammation and cancer. Topics: Epithelial-Mesenchymal Transition; Fibrosis; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Kidney; Models, Biological; Neoplasms; Signal Transduction | 2015 |
Implications of heparan sulfate and heparanase in neuroinflammation.
Heparan sulfate proteoglycans (HSPGs), expressed on the cell surface and in the extracellular matrix of most animal tissues, have essential functions in development and homeostasis, and have been implicated in several pathological conditions. The functions of HSPGs are mainly mediated through interactions of the heparan sulfate (HS) polysaccharide side chains with different protein ligands. The molecular structure of HS is highly diverse, expressed in a cell-type specific manner. The flexible yet controlled structure of HS is primarily generated through a strictly regulated biosynthesis process and is further modified post-synthetically, such as desulfation by endosulfatases and fragmentation by heparanase. Heparanase is an endo-glucuronidase expressed in all tissues. The enzyme has been found up-regulated in a number of pathological conditions, implying a role in diseases mainly through degradation of HS. Emerging evidence demonstrates important roles of HS and heparanase in inflammatory reactions, particularly in the regulation of leukocyte activation and extravasation. Neuroinflammation is a common feature of various central nervous system disorders, thus it is a great interest to understand the implications of HS and heparanase in neuroinflammation. Topics: Animals; Brain; Encephalitis; Extracellular Matrix; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Mice; Models, Biological | 2014 |
Pathophysiology of heparan sulphate: many diseases, few drugs.
Heparan sulphate (HS) polysaccharides are covalently attached to the core proteins of various proteoglycans at cell surfaces and in the extracellular matrix. They are composed of alternating units of hexuronic acid and glucosamine, with sulphate substituents in complex and variable yet cell-specific patterns. Whereas HS is produced by virtually all cells in the body, heparin, a highly sulphated HS variant, is confined to connective-tissue-type mast cells. The polysaccharides interact with a multitude of proteins, mainly through ionic binding, and thereby control key processes in development and homoeostasis. Similar interactions also implicate HS in various pathophysiological settings, including cancer, amyloid diseases, infectious diseases, inflammatory conditions and some developmental disorders. Prospects for the development of HS-based drugs, which are still largely unrealized, are discussed. Topics: Animals; Cell Membrane; Extracellular Matrix; Heparitin Sulfate; Homeostasis; Humans; Inflammation | 2013 |
Heparanase: multiple functions in inflammation, diabetes and atherosclerosis.
Topics: Atherosclerosis; Cell Adhesion; Cell Movement; Diabetes Mellitus; Extracellular Matrix; Gene Expression Regulation; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Models, Molecular; Protein Multimerization; Signal Transduction; Substrate Specificity | 2013 |
Heparan sulfate inhibitors and their therapeutic implications in inflammatory illnesses.
Heparan sulfate (HS) is a polysaccharide that is ubiquitously expressed on the cell surface and in the extracellular matrix and interacts with a wide variety of proteins to mediate numerous biological and pathological functions, including inflammation.. The structural diversity and the multiple biological roles of HS in inflammation are discussed. HS is involved in the recruitment and attachment of leukocytes to the inflamed epithelium, the activation of chemokines and the transmigration of leukocytes to the underlying target tissue. The endoglycosidase heparanase plays a key role in the above processes via the degradation of HS. HS mimetics that inhibit heparanase and block HS-binding proteins have been shown to inhibit inflammation in multiple animal models.. HS plays important roles in many stages of the inflammation process, in particular the regulation of leukocyte extravasation. Compounds that can inhibit HS-protein interactions thus have considerable potential as anti-inflammatory therapeutics capable of simultaneously interfering with multiple steps of the inflammation process. There are a number of such compounds in various stages of clinical development for cancer, which should also find applications in inflammatory illnesses. Topics: Animals; Glucuronidase; Heparitin Sulfate; Humans; Inflammation | 2013 |
[Glycan ligand specificity of killer lectin receptors].
Sialyl Lewis X (sLeX) antigen, Neu5Acα2,3Galβ1,4(Fucα1,3)GlcNAc-R, is expressed on the glycoproteins in sera or the surface of the cells and the expression of sLeX is enhanced in various conditions such as the inflammation and cancer. SLeX in the serum is utilized as a tumor marker. To clarify the roles of sLeX on secreted glycoproteins in vivo, we investigate the regulation of natural killer (NK) cell-dependent cytotoxicity through sLeX. NK cells express many receptors to kill the target cells such as cancerous cells and non-self, and their protein ligands have been elucidated. Of the killer lectin-like receptors (KLRs) on NK cells, several have been reported to recognize glycans. Using recombinant extracellular domains of KLRs (rKLRs: rNKG2A, C, D and rCD94), we evaluated their glycan ligand specificity and binding affinities using EIA methods. We clarified that all of these rKLRs can bind to high sLeX-expressing glycoprotein and heparin, heparan sulfate and highly sulfated polysaccharides and that glycan binding sites on NKG2D are mostly overlapped with those of protein ligands. In this review, we show the recent findings concerning the glycan ligands of these KLRs. Topics: Animals; Biomarkers; Cytotoxicity, Immunologic; Glycoproteins; Heparin; Heparitin Sulfate; Humans; Inflammation; Killer Cells, Natural; Lewis X Antigen; Ligands; Mice; Neoplasms; Polysaccharides; Protein Binding; Receptors, NK Cell Lectin-Like; Sialyl Lewis X Antigen | 2012 |
Heparin-derived heparan sulfate mimics to modulate heparan sulfate-protein interaction in inflammation and cancer.
The heparan sulfate (HS) chains of heparan sulfate proteoglycans (HSPG) are "ubiquitous" components of the cell surface and the extracellular matrix (EC) and play important roles in the physiopathology of developmental and homeostatic processes. Most biological properties of HS are mediated by interactions with "heparin-binding proteins" and can be modulated by exogenous heparin species (unmodified heparin, low molecular weight heparins, shorter heparin oligosaccharides and various non-anticoagulant derivatives of different sizes). Heparin species can promote or inhibit HS activities to different extents depending, among other factors, on how closely their structure mimics the biologically active HS sequences. Heparin shares structural similarities with HS, but is richer in "fully sulfated" sequences (S domains) that are usually the strongest binders to heparin/HS-binding proteins. On the other hand, HS is usually richer in less sulfated, N-acetylated sequences (NA domains). Some of the functions of HS chains, such as that of activating proteins by favoring their dimerization, often require short S sequences separated by rather long NA sequences. The biological activities of these species cannot be simulated by heparin, unless this polysaccharide is appropriately chemically/enzymatically modified or biotechnologically engineered. This mini review covers some information and concepts concerning the interactions of HS chains with heparin-binding proteins and some of the approaches for modulating HS interactions relevant to inflammation and cancer. This is approached through a few illustrative examples, including the interaction of HS and heparin-derived species with the chemokine IL-8, the growth factors FGF1 and FGF2, and the modulation of the activity of the enzyme heparanase by these species. Progresses in sequencing HS chains and reproducing them either by chemical synthesis or semi-synthesis, and in the elucidation of the 3D structure of oligosaccharide-protein complexes, are paving the way for rational approaches to the development of HS-inspired drugs in the field of inflammation and cancer, as well in other therapeutic fields. Topics: Animals; Anticoagulants; Antimicrobial Cationic Peptides; Blood Proteins; Carrier Proteins; Extracellular Matrix; Fibroblast Growth Factor 1; Fibroblast Growth Factor 2; Glucuronidase; Heparan Sulfate Proteoglycans; Heparin; Heparitin Sulfate; Humans; Inflammation; Interleukin-8; Models, Molecular; Neoplasms; Oligosaccharides; Polysaccharides; Proteins | 2010 |
Heparin, heparan sulfate and heparanase in inflammatory reactions.
Heparan sulfate (HS) proteoglycans at the cell surface and in the extracellular matrix of most animal tissues are essential in development and homeostasis, and are implicated in disease processes. Emerging evidence demonstrates the important roles of HS in inflammatory reactions, particularly in the regulation of leukocyte extravasation. Heparin, a classical anticoagulant, exhibits anti-inflammatory effects in animal models and in the clinic, presumably through interference with the functions of HS, as both polysaccharides share a high similarity in molecular structure. Apart of regulation during biosynthesis, the structures of HS and heparin are significantly modulated by heparanase, an endoglycosidase that is upregulated in a number of inflammatory conditions. Exploring the physiological roles of HS and heparin and the mode of heparanase action in modulating their functions during inflammation responses is of importance for future studies. Topics: Amyloidosis; Animals; Anti-Inflammatory Agents; Carbohydrate Sequence; Chemotaxis, Leukocyte; Gene Expression Regulation, Enzymologic; Glucuronidase; Golgi Apparatus; Heparin; Heparitin Sulfate; Humans; Inflammation; Mice; Mice, Knockout; Mice, Transgenic; Models, Molecular; Molecular Sequence Data; Molecular Structure; Recombinant Fusion Proteins; Structure-Activity Relationship | 2009 |
[Mechanisms of action of the growth factor midkine].
Topics: Amino Acid Sequence; Animals; Chondroitin Sulfates; Cytokines; Heparitin Sulfate; Humans; Inflammation; Midkine; Molecular Sequence Data; Neoplasms; Nerve Growth Factor; Protein Binding; Protein Conformation; Protein Structure, Tertiary | 2008 |
Heparanase: a target for drug discovery in cancer and inflammation.
The remodelling of the extracellular matrix (ECM) has been shown to be highly upregulated in cancer and inflammation and is critically linked to the processes of invasion and metastasis. One of the key enzymes involved in specifically degrading the heparan sulphate (HS) component of the ECM is the endo-beta-glucuronidase enzyme heparanase. Processing of HS by heparanase releases both a host of bioactive growth factors anchored within the mesh of the ECM as well as defined fragments of HS capable of promoting cellular proliferation. The finding that heparanase is elevated in a wide variety of tumor types and is subsequently linked to the development of pathological processes has led to an explosion of therapeutic strategies to inhibit its enzyme activity. So far only one compound, the sulphated oligosaccharide PI88, which both inhibits heparanase activity and has effects on growth factor binding has reached clinical trials where it has shown to have promising efficacy. The scene has clearly been set however for a new generation of compounds, either specific to the enzyme or with dual roles, to emerge from the lab and enter the clinic. The aim of this review is to describe the current drug discovery status of small molecule, sugar and neutralising antibody inhibitors of heparanase enzyme activity. Potential strategies will also be discussed on the selection of suitable biomarker strategies for specific monitoring of in vivo heparanase inhibition which will be crucial for both animal model and clinical trial testing. Topics: Animals; Drug Design; Enzyme Inhibitors; Gene Expression Regulation, Enzymologic; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Neoplasms; Vascular Endothelial Growth Factor A | 2007 |
The role of heparan sulphate in inflammation.
The polysaccharide heparan sulphate is ubiquitously expressed as a proteoglycan in extracellular matrices and on cell surfaces. Heparan sulphate has marked sequence diversity that allows it to specifically interact with many proteins. This Review focuses on the multiple roles of heparan sulphate in inflammatory responses and, in particular, on its participation in almost every stage of leukocyte transmigration through the blood-vessel wall. Heparan sulphate is involved in the initial adhesion of leukocytes to the inflamed endothelium, the subsequent chemokine-mediated transmigration through the vessel wall and the establishment of both acute and chronic inflammatory reactions. Topics: Animals; Basement Membrane; Cell Adhesion; Chemokines; Heparitin Sulfate; Humans; Inflammation; Proteoglycans | 2006 |
Activation of human inflammatory cells by secreted phospholipases A2.
Secreted phospholipases A(2) (sPLA(2)s) are enzymes detected in serum and biological fluids of patients with various inflammatory, autoimmune and allergic disorders. Different isoforms of sPLA(2)s are expressed and released by human inflammatory cells, such as neutrophils, eosinophils, T cells, monocytes, macrophages and mast cells. sPLA(2)s generate arachidonic acid and lysophospholipids thus contributing to the production of bioactive lipid mediators in inflammatory cells. However, sPLA(2)s also activate human inflammatory cells by mechanisms unrelated to their enzymatic activity. Several human and non-human sPLA(2)s induce degranulation of mast cells, neutrophils and eosinophils and activate exocytosis in macrophages. In addition some, but not all, sPLA(2) isoforms promote cytokine and chemokine production from macrophages, neutrophils, eosinophils, monocytes and endothelial cells. These effects are primarily mediated by binding of sPLA(2)s to specific membrane targets (heparan sulfate proteoglycans, M-type, N-type or mannose receptors) expressed on effector cells. Thus, sPLA(2)s may play an important role in the initiation and amplification of inflammatory reactions by at least two mechanisms: production of lipid mediators and direct activation of inflammatory cells. Selective inhibitors of sPLA(2)-enzymatic activity and specific antagonists of sPLA(2) receptors are current being tested for pharmacological treatment of inflammatory and autoimmune diseases. Topics: Autoimmune Diseases; Cell Degranulation; Exocytosis; Heparitin Sulfate; Humans; Inflammation; Inflammation Mediators; Lectins, C-Type; Leukocytes; Macrophage Activation; Mannose Receptor; Mannose-Binding Lectins; Phospholipases A; Protein Binding; Proteoglycans; Receptors, Cell Surface | 2006 |
Interactions of platelet factor 4 with the vessel wall.
Platelet factor 4 (PF4) is a platelet-specific protein that is stored in platelet alpha granules and released following platelet activation. PF4 was the first chemokine that was isolated, but unlike other chemokines, it may not have a clear role in inflammation. Gathering evidence suggests that unlike other chemokines that bind to specific receptors, PF4's biology depends on its unusually high affinity for heparan sulfates and other negatively charged molecules at concentrations attained in the immediate vicinity of activated platelets. There has been one report that PF4 binds to CXCR3B, a chemokine receptor isoform that may be present in some vascular beds, but the biological relevance of this single observation is not clear. We propose that the main biological role of PF4 and the basis for its presence in the alpha granules of all known mammalian platelets is to neutralize surface heparan sulfate side-chains of glycosaminoglycans and to optimize thrombus development at sites of vascular injury. In addition, the binding of PF4 to surface glycosaminoglycans may also underlie its angiostatic and proatherogenic properties. Additionally, PF4 binds to several other proteins that are central to thrombosis, angiogenesis, and atherogenesis. These interactions may also contribute to its biological and pathobiological effects. Certainly, future studies using in vivo models to test biological relevance of each of these proposed mechanisms by which PF4 interacts with the vasculature are needed, as are studies to define the importance of PF4 binding to CXCR3B. Topics: Animals; Blood Platelets; Blood Vessels; Chemokines; Dose-Response Relationship, Drug; Endothelium, Vascular; Glycosaminoglycans; Heparitin Sulfate; Humans; Inflammation; Mice; Models, Biological; Neovascularization, Pathologic; Platelet Activation; Platelet Factor 4; Protein Binding; Protein Isoforms; Receptors, Chemokine; Receptors, CXCR3; Thrombosis | 2004 |
Glycosaminoglycans and the regulation of allergic inflammation.
Glycosaminoglycans (GAGs) are large, polyanionic molecules expressed throughout the body. The GAG heparin, co-released with histamine, is synthesised by and stored exclusively in mast cells, whereas the closely related molecule heparan sulphate is expressed, as part of a proteoglycan, on cell surfaces and throughout tissue matrices. These molecules are increasingly thought to play a role in regulation of the inflammatory response and heparin like molecules are now being seriously considered to hold potential in the treatment of inflammatory diseases such as asthma. Heparin and related molecules have been found to exert anti-inflammatory effects in a wide range of in vitro assays, animal models and in human disease. The anti-inflammatory activities of heparin are independent of the well-established anticoagulant activity of heparin, suggesting that the separation of these properties could yield novel anti-inflammatory drugs, which may be useful in the future treatment of inflammatory diseases. Topics: Animals; Anti-Inflammatory Agents; Glucuronidase; Glycosaminoglycans; Heparin; Heparitin Sulfate; Humans; Hypersensitivity; Inflammation; Mast Cells | 2004 |
Syndecans in inflammation.
Cell surface heparan sulfate (HS) influences a multitude of molecules, cell types, and processes relevant to inflammation. HS binds to cell surface and matrix proteins, cytokines, and chemokines. These interactions modulate inflammatory cell maturation and activation, leukocyte rolling, and tight adhesion to endothelium, as well as extravasation and chemotaxis. The syndecan family of transmembrane proteoglycans is the major source of cell surface HS on all cell types. Recent in vitro and in vivo data suggest the involvement of syndecans in the modulation of leukocyte-endothelial interactions and extravasation, the formation of chemokine and kininogen gradients, participation in chemokine and growth factor signaling, as well as repair processes. Thus, the complex role of HS in inflammation is reflected by multiple functions of its physiological carriers, the syndecans. Individual and common functions of the four mammalian syndecan family members can be distinguished. Recently generated transgenic and knockout mouse models will facilitate analysis of the individual processes that each syndecan is involved in. Topics: Animals; Chemokines; Cytokines; Heparitin Sulfate; Humans; Inflammation; Membrane Glycoproteins; Proteoglycans; Syndecans | 2003 |
Glycosaminoglycans, airways inflammation and bronchial hyperresponsiveness.
Glycosaminoglycans (GAGs) are large, polyanionic molecules expressed throughout the body. The GAG heparin, co-released with histamine, is synthesised by and stored exclusively in mast cells, whereas the closely related molecule heparan sulphate is expressed, as part of a proteoglycan, on cell surfaces and throughout tissue matrices. These molecules are increasingly thought to play a role in regulation of the inflammatory response and heparin, for many years, has been considered to hold potential in the treatment of diseases such as asthma. Heparin and related molecules have been found to exert antiinflammatory effects in a wide range of in vitro assays, animal models and, indeed, human patients. Moreover, the results of studies carried out to date indicate that the antiinflammatory activities of heparin are dissociable from its well-established anticoagulant nature, suggesting that the separation of these characteristics could yield novel antiinflammatory drugs which may be useful in the future treatment of diseases such as asthma Topics: Animals; Anticoagulants; Asthma; Glycosaminoglycans; Heparin; Heparitin Sulfate; Humans; Inflammation; Mast Cells | 2001 |
Human leucocyte heparan sulphate proteoglycans and their roles in inflammation.
Topics: Antigens, CD; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Hyaluronan Receptors; Inflammation; Leukocytes; Membrane Glycoproteins; Models, Structural; Monocytes; Proteoglycans; Receptors, Transforming Growth Factor beta; Syndecans; T-Lymphocytes | 1997 |
In vivo, in vitro, and molecular aspects of interleukin-8 and the interleukin-8 receptors.
Topics: Amino Acid Sequence; Animals; Binding Sites; Heparitin Sulfate; Humans; In Vitro Techniques; Inflammation; Interleukin-8; Ligands; Molecular Sequence Data; Neutrophils; Receptors, Interleukin | 1996 |
Expression of heparanase by platelets and circulating cells of the immune system: possible involvement in diapedesis and extravasation.
Interaction of T and B lymphocytes, platelets, granulocytes, macrophages and mast cells with the subendothelial extracellular matrix (ECM) is associated with degradation of heparan sulfate (HS) by a specific endoglycosidase (heparanase) activity. The enzyme is released from intracellular compartments (i.e., lysosomes, specific granules) in response to various activation signals (i.e., thrombin, calcium ionophore, immune complexes, antigens, mitogens), suggesting its regulated involvement in inflammation and cellular immunity. In contrast, various tumor cells appear to express and secrete heparanase in a constitutive manner, in correlation with their metastatic potential. Heparanase enzymes produced by different cell types may exhibit different molecular properties and substrate cleavage specificities. The platelet enzyme appears also in a latent form. It can be activated by tumor cells and thereby facilitate their extravasation in the process of metastasis. Degradation of ECM-HS by all cell types was facilitated by a proteolytic activity residing in the ECM and/or expressed by the invading cells. This proteolytic activity produced a more accessible substrate for the heparanase enzymes. Heparanase-inhibiting, nonanticoagulant species of heparin markedly reduced the incidence of lung metastasis in experimental animals. These species of heparin also significantly impaired the traffic of T lymphocytes and suppressed cellular immune reactivity and experimental autoimmune diseases. Heparanase activity expressed by intact cells (i.e., platelets, mast cells, neutrophils, lymphoma cells) was found to release active HS-bound basic fibroblast growth factor from ECM and basement membranes. Heparanase may thus elicit an indirect neovascular response in processes such as wound repair, inflammation and tumor development. The significant anticancerous effect of heparanase-inhibiting molecules may therefore be attributed to their potential inhibition of both tumor invasion and angiogenesis. Both normal leukocytic cells and metastatic tumor cells can enter the bloodstream, travel to distant sites and extravasate to the parenchyma at these sites. We suggest that heparanase is utilized for this purpose by both types of cells. Other functions (i.e., enzyme activities, adhesive interactions, chemotactic and proliferative responses) of metastatic tumor cells seem to mimic the equivalent functions of leukocytes as they migrate across blood vessels to gain access to sites of infl Topics: Animals; Blood Cells; Blood Platelets; Cell Movement; Endothelium, Vascular; Extracellular Matrix; Fibroblast Growth Factor 2; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Humans; Immunity, Cellular; Inflammation; Lymphocytes, Tumor-Infiltrating; Polysaccharide-Lyases | 1992 |
Structure of the dermal-epidermal junction and potential mechanisms for its degradation: the possible role of inflammatory cells.
Evidence was presented indicating that the DEJ as a basement membrane is highly susceptible to degradation by a variety of neutral proteolytic enzymes with different specificities. The effect of endoglycosidases which degrade heparan sulfate was also discussed. The latter enzymes are capable of removing heparan sulfate from the DEJ, but little gross alteration of structure, such as tissue detachment, appears to result from the loss of this component. Of the proteinases discussed, PMN elastase and probably type IV collagenase are the most destructive. This is likely related to their ability to degrade the type IV collagen network. Even though proteinases with chymotrypsinlike and trypsinlike specificity were not efficient at degrading the lamina densa or removing type IV collagen from intact basement membranes, these proteinases were capable of producing epidermal detachment from the lamina densa. Many inflammatory cells of the immune system contain proteinases and endoglycosidases with the potential to degrade the DEJ and other basement membrane zones, suggesting that these cells may have a significant pathologic role in basement membrane-related diseases. PMNs and mast cells may be of particular interest because they have stored within their secretory granules high concentrations of neutral serine proteinases which have been demonstrated to degrade the DEJ. Topics: Animals; Basement Membrane; Chondroitin Sulfate Proteoglycans; Endopeptidases; Epidermis; Extracellular Matrix; Extracellular Matrix Proteins; Glycoside Hydrolases; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Inflammation; Leukocytes; Parasites; Skin; Skin Diseases, Parasitic | 1989 |
2 trial(s) available for heparitin-sulfate and Inflammation
Article | Year |
---|---|
Peptide 19-2.5 inhibits heparan sulfate-triggered inflammation in murine cardiomyocytes stimulated with human sepsis serum.
Myocardial dysfunction in sepsis has been linked to inflammation caused by pathogen-associated molecular patterns (PAMPs) as well as by host danger-associated molecular patterns (DAMPs). These include soluble heparan sulfate (HS), which triggers the devastating consequences of the pro-inflammatory cascades in severe sepsis and septic shock. Thus, there is increasing interest in the development of anti-infective agents, with effectiveness against both PAMPs and DAMPs. We hypothesized that a synthetic antimicrobial peptide (peptide 19-2.5) inhibits inflammatory response in murine cardiomyocytes (HL-1 cells) stimulated with PAMPs, DAMPs or serum from patients with septic shock by reduction and/or neutralization of soluble HS. In the current study, our data indicate that the treatment with peptide 19-2.5 decreases the inflammatory response in HL-1 cells stimulated with either PAMPs or DAMPs. Furthermore, our work shows that soluble HS in serum from patients with Gram-negative or Gram-positive septic shock induces a strong pro-inflammatory response in HL-1 cells, which can be effectively blocked by peptide 19-2.5. Based on these findings, peptide 19-2.5 is a novel anti-inflammatory agent interacting with both PAMPs and DAMPs, suggesting peptide 19-2.5 may have the potential for further development as a broad-spectrum anti-inflammatory agent in sepsis-induced myocardial inflammation and dysfunction. Topics: Aged; Animals; Antimicrobial Cationic Peptides; Cell Line; Female; Heparitin Sulfate; Humans; Inflammation; Male; Mice; Middle Aged; Myocytes, Cardiac; Sepsis; Serum | 2015 |
Effect of factor X inhibition on coagulation activation and cytokine induction in human systemic inflammation.
Anticoagulants have gained increasing attention in the treatment of sepsis. This study used danaparoid to investigate the role of factor Xa in endotoxin-induced coagulation and inflammation and its effectiveness when coagulation activation has already occurred. Thirty healthy volunteers were enrolled in the randomized, placebo-controlled trial. Subjects received 2 ng/kg endotoxin and danaparoid 10 min or 3 h thereafter or placebo. Endotoxin increased prothrombin fragment 1+2 (F(1+2)) levels from 0.5 to 7.0 nmol/L at 5 h in the placebo group. Early danaparoid infusion inhibited endotoxin-induced thrombin formation: maximum F(1+2) levels reached only 1.8 nmol/L (P<.01, vs. baseline or placebo). Delayed danaparoid infusion effectively blocked further thrombin formation. However, danaparoid did not alter endotoxin-induced changes in the fibrinolytic system, cytokine levels, activation of leukocytes, or tissue factor expression on monocytes. Danaparoid therefore selectively attenuates endotoxin-induced coagulopathy, even with delayed administration when coagulation activation is well under way. Topics: Adult; alpha-2-Antiplasmin; Anticoagulants; Blood Coagulation; Chemokine CCL2; Chondroitin Sulfates; Cytokines; Dermatan Sulfate; Drug Combinations; Endotoxins; Factor X; Heparitin Sulfate; Humans; Inflammation; Interleukin-6; Kinetics; Lipopolysaccharides; Pilot Projects; Reference Values | 2002 |
59 other study(ies) available for heparitin-sulfate and Inflammation
Article | Year |
---|---|
Trajectory of plasma syndecan-1 and heparan sulphate during major surgery: A retrospective observational study.
Surgical trauma-induced inflammation during major surgery may disrupt endothelial integrity and affect plasma concentrations of glycocalyx constituents, such as syndecan-1 and heparan sulphate. To date, no studies have focused on their perioperative temporal changes.. As part of a trial, we obtained plasma and urine specimens sampled during the perioperative period in 72 patients undergoing major abdominal surgery. The plasma concentration of syndecan-1 and heparan sulphate was measured on five occasions, from baseline to the second postoperative day. Plasma and urinary creatinine and urinary syndecan-1 concentrations were measured before surgery and on the first postoperative morning.. We observed three different temporal patterns of plasma syndecan-1 concentration. Group 1 'low' (64% of patients) showed only minor changes from baseline despite a median heparan sulphate increase of 67% (p < .005). Group 2 'increase' (21% of patients) showed a marked increase in median plasma syndecan-1 from 27 μg/L to 118 μg/L during the first postoperative day (p < .001) with a substantial (+670%; p < .005) increase in median plasma heparan sulphate from 279 to 2196 μg/L. Group 3 'high' (14% of patients) showed a constant elevation of plasma syndecan-1 to >100 μg/L, but low heparan sulphate levels. The plasma C-reactive protein concentration did not differ across the three groups and 90% of colon surgeries occurred in Group 1. Treatment with dexamethasone was similar across the three groups. Surgical blood loss, duration of surgery and liver resection were greatest in Group 2.. Changes in syndecan-1 and heparan sulphate after surgery appear to show three different patterns, with the greatest increases in those patients with greater blood loss, more liver surgery and longer operations. These observations suggest that increases in syndecan-1 and heparan sulphate reflect the degree of surgical injury. Topics: Glycocalyx; Heparitin Sulfate; Humans; Inflammation; Liver; Retrospective Studies; Syndecan-1 | 2023 |
Transport of the Proinflammatory Chemokines C-C Motif Chemokine Ligand 2 (MCP-1) and C-C Motif Chemokine Ligand 5 (RANTES) across the Intact Mouse Blood-Brain Barrier Is Inhibited by Heparin and Eprodisate and Increased with Systemic Inflammation.
Topics: Animals; Blood-Brain Barrier; Chemokines; Heparin; Heparitin Sulfate; Inflammation; Ligands; Mice; Receptors, Chemokine | 2023 |
A 3-
Dysregulated inflammation and coagulation are underlying mechanisms driving organ injury after trauma and hemorrhagic shock. Heparan sulfates, cell surface glycosaminoglycans abundantly expressed on the endothelial surface, regulate a variety of cellular processes. Endothelial heparan sulfate containing a rare 3-. Male Sprague-Dawley rats were pre-treated subcutaneously with vehicle (saline) or dekaparin (2 mg/kg) and subjected to a trauma/hemorrhagic shock model through laparotomy, gut distention, and fixed-pressure hemorrhage. Vehicle and dekaparin-treated rats were resuscitated with Lactated Ringer's solution (LR) and compared to vehicle-treated fresh-frozen-plasma-(FFP)-resuscitated rats. Serial blood samples were collected at baseline, after induction of shock, and 3 hours after fluid resuscitation to measure hemodynamic and metabolic shock indicators, inflammatory mediators, and thrombin-antithrombin complex formation. Lungs and kidneys were processed for organ injury scoring and immunohistochemical analysis to quantify presence of neutrophils.. Induction of trauma and hemorrhagic shock resulted in significant increases in thrombin-antithrombin complex, inflammatory markers, and lung and kidney injury scores. Compared to vehicle, dekaparin treatment did not affect induction, severity, or recovery of shock as indicated by hemodynamics, metabolic indicators of shock (lactate and base excess), or metrics of bleeding, including overall blood loss, resuscitation volume, or hematocrit. While LR-vehicle-resuscitated rodents exhibited increased lung and kidney injury, administration of dekaparin significantly reduced organ injury scores and was similar to organ protection conferred by FFP resuscitation. This was associated with a significant reduction in neutrophil infiltration in lungs and kidneys and reduced lung fibrin deposition among dekaparin-treated rats compared to vehicle. No differences in organ injury, neutrophil infiltrates, or fibrin staining between dekaparin and FFP groups were observed. Finally, dekaparin treatment attenuated induction of thrombin-antithrombin complex and inflammatory mediators in plasma following trauma and hemorrhagic shock.. Anti-thromboinflammatory properties of a synthetic 3- Topics: Animals; Fibrin; Heparitin Sulfate; Inflammation; Male; Rats; Rats, Sprague-Dawley; Shock, Hemorrhagic; Sulfates; Thromboinflammation; Thrombosis | 2023 |
Synthesis of novel
Heparan sulfate (HS), a glycosaminoglycan related to heparin, is a linear polysaccharide, consisting of repeating disaccharide units. This compound is involved in multiple biological processes such as inflammation, coagulation, angiogenesis and viral infections. Our work focuses on the synthesis of simple HS analogs for the study of structure-activity relationships, with the aim of modulating these biological activities. Thioglycoside analogs, in which the interglycosidic oxygen is replaced by a sulfur atom, are very interesting compounds in terms of therapeutic applications. Indeed, the thioglycosidic bond leads to an improvement of their stability and can allow the inhibition of enzymes involved in physiological and pathological processes. In our previous work, we developed a synthetic sequence which led to a non-sulfated thiodisaccharide analog of HS. In this paper, we report our results of the development of a new synthetic method allowing access to the novel sulfated Topics: Disaccharides; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Sulfur | 2022 |
Acute T-Cell-Driven Inflammation Requires the Endoglycosidase Heparanase-1 from Multiple Cell Types.
It has been accepted for decades that T lymphocytes and metastasising tumour cells traverse basement membranes (BM) by deploying a battery of degradative enzymes, particularly proteases. However, since many redundant proteases can solubilise BM it has been difficult to prove that proteases aid cell migration, particularly in vivo. Recent studies also suggest that other mechanisms allow BM passage of cells. To resolve this issue we exploited heparanase-1 (HPSE-1), the only endoglycosidase in mammals that digests heparan sulfate (HS), a major constituent of BM. Initially we examined the effect of HPSE-1 deficiency on a well-characterised adoptive transfer model of T-cell-mediated inflammation. We found that total elimination of HPSE-1 from this system resulted in a drastic reduction in tissue injury and loss of target HS. Subsequent studies showed that the source of HPSE-1 in the transferred T cells was predominantly activated CD4 Topics: Animals; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Inflammation; Mammals; Peptide Hydrolases; T-Lymphocytes | 2022 |
The Influence of Heparan Sulfate on Breast Amyloidosis and the Toxicity of the Pre-fibrils Formed by β-casein.
Heparan sulfate (HS) as a mediator is usually involved in both inflammation and fibrosis. Besides, pre-fibrils are the intermediates of amyloid fibrils that usually cause cell death and tissue damage, like the amyloid-β in Alzheimer's disease, α-synuclein in Parkinson disease and islet amyloid polypeptide in type II diabetes mellitus. However, the related study was involved rarely in breast. Therefore, the combing technologies including hematoxylin-eosin staining and thioflavin S staining were used to investigate the influence of HS on breast amyloidosis. To further study the toxicity of the pre-fibrils formed by β-casein on the HC11 cells and the breast mammary gland, the combing technologies including pentamer formyl thiophene acetic acid fluorescence analysis, MTT assay, Annexin V/PI staining and hematoxylin-eosin staining were performed. The results demonstrated that HS, acted as an endogenous molecule, induced the inflammation and amyloid fibril formation at the same time, and there was a close relationship between inflammation and fibrosis of breast. In addition, the pre-fibrils formed by β-casein were toxic because they induced the death and apoptosis of HC11 cells, as well as the inflammation of mammary gland of rats. Therefore, the early examination and identify of the pre-fibrils in the breast were worth considering to prevent the disease development, and it was interesting to explore the HS mimetics to impair the breast amyloidosis and attenuate the inflammatory response in the future. Topics: Amyloid; Amyloidosis; Animals; Caseins; Diabetes Mellitus, Type 2; Eosine Yellowish-(YS); Fibrosis; Hematoxylin; Heparitin Sulfate; Inflammation; Rats | 2022 |
HA and HS Changes in Endothelial Inflammatory Activation.
Cardiovascular diseases are a group of disorders caused by the presence of a combination of risk factors, such as tobacco use, unhealthy diet and obesity, physical inactivity, etc., which cause the modification of the composition of the vessel's matrix and lead to the alteration of blood flow, matched with an inflammation condition. Nevertheless, it is not clear if the inflammation is a permissive condition or a consequent one. In order to investigate the effect of inflammation on the onset of vascular disease, we treated endothelial cells with the cytokine TNF-α that is increased in obese patients and is reported to induce cardiometabolic diseases. The inflammation induced a large change in the extracellular matrix, increasing the pericellular hyaluronan and altering the heparan sulfate Syndecans sets, which seems to be related to layer permeability but does not influence cell proliferation or migration nor induce blood cell recruitment or activation. Topics: Heparitin Sulfate; Human Umbilical Vein Endothelial Cells; Humans; Hyaluronic Acid; Inflammation | 2021 |
Heparin - An old drug with multiple potential targets in Covid-19 therapy.
Topics: Anticoagulants; COVID-19 Drug Treatment; Critical Illness; Glycosaminoglycans; Heparin; Heparitin Sulfate; Humans; Inflammation; Protein Binding; Pulmonary Gas Exchange; Sepsis | 2020 |
Characterization of Fluid Biomarkers Reveals Lysosome Dysfunction and Neurodegeneration in Neuronopathic MPS II Patients.
Mucopolysaccharidosis type II is a lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS) and characterized by the accumulation of the primary storage substrate, glycosaminoglycans (GAGs). Understanding central nervous system (CNS) pathophysiology in neuronopathic MPS II (nMPS II) has been hindered by the lack of CNS biomarkers. Characterization of fluid biomarkers has been largely focused on evaluating GAGs in cerebrospinal fluid (CSF) and the periphery; however, GAG levels alone do not accurately reflect the broad cellular dysfunction in the brains of MPS II patients. We utilized a preclinical mouse model of MPS II, treated with a brain penetrant form of IDS (ETV:IDS) to establish the relationship between markers of primary storage and downstream pathway biomarkers in the brain and CSF. We extended the characterization of pathway and neurodegeneration biomarkers to nMPS II patient samples. In addition to the accumulation of CSF GAGs, nMPS II patients show elevated levels of lysosomal lipids, neurofilament light chain, and other biomarkers of neuronal damage and degeneration. Furthermore, we find that these biomarkers of downstream pathology are tightly correlated with heparan sulfate. Exploration of the responsiveness of not only CSF GAGs but also pathway and disease-relevant biomarkers during drug development will be crucial for monitoring disease progression, and the development of effective therapies for nMPS II. Topics: Adolescent; Animals; Biomarkers; Brain; Child; Child, Preschool; Dermatan Sulfate; Enzyme Replacement Therapy; Female; Gangliosides; Glycosaminoglycans; Hematopoietic Stem Cell Transplantation; Heparitin Sulfate; Humans; Iduronate Sulfatase; Infant; Inflammation; Lipid Metabolism; Lysosomes; Male; Mass Spectrometry; Mice; Mice, Knockout; Mucopolysaccharidosis II; Neurofilament Proteins; Recombinant Proteins | 2020 |
Managing chronic, nonhealing wounds stalled in the inflammatory phase: a case series using a novel matrix therapy, CACIPLIQ20.
One of the biggest challenges faced by healthcare providers is the treatment of chronic, non-healing wounds. This paper reports for the first time in the UK the results of five case studies in which a novel regenerating matrix-based therapy, CACIPLIQ20, was used. CACIPLIQ20 is a heparan sulphate mimetic designed to replace the destroyed heparan sulphate in the extracellular matrix of wound cells. All five patients in this case series had chronic, non-healing ulcers that had not improved with conventional care. Treatment included two applications of CACIPLIQ20 per week, for a maximum of 12 weeks. Three of the five wounds healed completely, and the remaining two showed significant improvements in size and quality. The treatment was well tolerated by the patients and also led to a significant reduction in pain. Moreover, CACIPLIQ20 treatment was found to be highly cost-effective when compared to conventional care, with the potential to save healthcare systems significant resources. Further studies are needed to build a strong evidence base on the use of this product, but these preliminary findings are certainly promising. Topics: Adult; Aged; Aged, 80 and over; Amputation, Surgical; Bandages; Chronic Disease; Cost-Benefit Analysis; Extracellular Matrix; Female; Glucans; Heparitin Sulfate; Humans; Inflammation; Male; Middle Aged; Peripheral Vascular Diseases; Pressure Ulcer; Regeneration; Surgical Wound; Varicose Ulcer; Wounds and Injuries | 2019 |
Heparanase-Dependent Remodeling of Initial Lymphatic Glycocalyx Regulates Tissue-Fluid Drainage During Acute Inflammation
The glycocalyx is a dense layer of carbohydrate chains involved in numerous and fundamental biological processes, such as cellular and tissue homeostasis, inflammation and disease development. Composed of membrane-bound glycoproteins, sulfated proteoglycans and glycosaminoglycan side-chains, this structure is particularly essential for blood vascular barrier functions and leukocyte diapedesis. Interestingly, whilst the glycocalyx of blood vascular endothelium has been extensively studied, little is known about the composition and function of this glycan layer present on tissue-associated lymphatic vessels (LVs). Here, we applied confocal microscopy to characterize the composition of endothelial glycocalyx of initial lymphatic capillaries in murine cremaster muscles during homeostatic and inflamed conditions using an anti-heparan sulfate (HS) antibody and a panel of lectins recognizing different glycan moieties of the glycocalyx. Our data show the presence of HS, α-D-galactosyl moieties, α2,3-linked sialic acids and, to a lesser extent, N-Acetylglucosamine moieties. A similar expression profile was also observed for LVs of mouse and human skins. Interestingly, inflammation of mouse cremaster tissues or ear skin as induced by TNF-stimulation induced a rapid (within 16 h) remodeling of the LV glycocalyx, as observed by reduced expression of HS and galactosyl moieties, whilst levels of α2,3-linked sialic acids remains unchanged. Furthermore, whilst this response was associated with neutrophil recruitment from the blood circulation and their migration into tissue-associated LVs, specific neutrophil depletion did not impact LV glycocalyx remodeling. Mechanistically, treatment with a non-anticoagulant heparanase inhibitor suppressed LV HS degradation without impacting neutrophil migration into LVs. Interestingly however, inhibition of glycocalyx degradation reduced the capacity of initial LVs to drain interstitial fluid during acute inflammation. Collectively, our data suggest that rapid remodeling of endothelial glycocalyx of tissue-associated LVs supports drainage of fluid and macromolecules but has no role in regulating neutrophil trafficking out of inflamed tissues via initial LVs. Topics: Abdominal Muscles; Animals; Drainage; Extracellular Fluid; Female; Glucuronidase; Glycocalyx; Heparitin Sulfate; Humans; Inflammation; Lymphatic Vessels; Male; Mice; Mice, Inbred C57BL; Neutrophils; Tumor Necrosis Factor-alpha | 2019 |
Indoor nanoscale particulate matter-induced coagulation abnormality based on a human 3D microvascular model on a microfluidic chip.
A growing body of evidence shows that indoor concentrations of airborne particles are often higher than is typically encountered outdoors. Since exposure to indoor PM2.5 is thought to be associated with cardiovascular disease, the health impacts of indoor air pollution need to be explored. Based on animal models, ambient particulate matter has been proved to promote coagulation which is very likely involved in the pathogenic development of cardiovascular disease. However, animal models are insufficient to predict what will happen with any certainty in humans. For this reason, the precise pathogenic mechanisms behind the development of cardiovascular disease in humans have not yet been determined.. We generated a 3D functional human microvascular network in a microfluidic device. This model enables human vascular endothelial cells to form tissue-like microvessels that behave very similarly to human blood vessels. The perfusable microvasculature allows the delivery of particles introduced into these generated human-like microvessels to follow the fluid flow. This exposure path effectively simulates the dynamic movement of airborne nanoscale particles (ANPs) within human vessels. In this study, we first identified the existence of ANPs in indoor air pollution. We then showed that ANPs could activate endothelial cells via ROS induced inflammation, and further resulted in abnormal expression of the coagulation factors (TF, TM and t-PA) involved in coagulation cascades. In addition, we found that a protein could cover ANPs, and this biointeraction could interfere with heparan sulfate (HS). Human organotypic 3D microvessel models provide a bridge for how research outcomes can translate to humans.. The 3D human microvessel model was used to determine the physiological responses of human vessels to ANP stimulation. Based on the obtained data, we concluded that ANPs not only disrupts normal coagulation functions, but also act directly on anticoagulant factors in human vessels. These experimental observations provide a potential biological explanation for the epidemiologically established link between ANPs and coagulation abnormality. This organ-on-chip model may provide a bridge from in vitro results to human responses. Topics: Air Pollution, Indoor; Blood Coagulation Factors; Endothelial Cells; Heparitin Sulfate; Humans; Inflammation; Lab-On-A-Chip Devices; Microvessels; Models, Biological; Particle Size; Particulate Matter; Reactive Oxygen Species | 2019 |
Heparan sulfate inhibits inflammation and improves wound healing by downregulating the NLR family pyrin domain containing 3 (NLRP3) inflammasome in diabetic rats.
Heparan sulfate (HS) attenuates the inflammatory response and improves diabetic wound healing in rats. However, the specific mechanisms by which HS suppresses inflammation are not clear. Given that NLR family pyrin domain containing 3 (NLRP3) is a major receptor involved in innate immune regulation, the aim of the present study was to elucidate the effects of HS on NLRP3 and proinflammatory cytokines in diabetic wounds.. Full-thickness wounds were created on the back of diabetic rats. The experimental group received HS treatment (1 mg/kg, i.m., on Days 0 and 7), whereas the control group received vehicle (0.1% dimethylsulfoxide in 0.9% NaCl). Expression of NLRP3 and its downstream effector molecules, namely cleaved interleukin (IL)-1β, IL-18, tumor necrosis factor (TNF)-α, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), proteinase inhibitor 9, and caspase-12, in the wound tissues was examined.. Treatment with HS accelerated wound healing in diabetic rats. Rats treated with HS exhibited decreased activation of cleaved IL-1β, IL-18, and TNF-α, as well as decreased expression of NLRP3 and ASC. In addition, HS increased levels of proteinase inhibitor 9 and caspase-12.. Heparan sulfate inhibits inflammation and improves wound healing by downregulating the NLRP3 inflammasome and cleaved IL-1β during the wound healing process in diabetic rats. Topics: Animals; Apoptosis; Cytokines; Diabetes Mellitus, Experimental; Heparitin Sulfate; Inflammasomes; Inflammation; Male; NLR Family, Pyrin Domain-Containing 3 Protein; Rats; Rats, Sprague-Dawley; Wound Healing | 2018 |
Docking of Meprin α to Heparan Sulphate Protects the Endothelium from Inflammatory Cell Extravasation.
Pulmonary arterial hypertension (PAH) is a rare disease characterized by increased pulmonary pressure and vascular remodelling as a consequence of smooth muscle cell proliferation, endothelial cell dysfunction and inflammatory infiltrates. Meprin α is a metalloproteinase whose substrates include adhesion and cell-cell contact molecules involved in the process of immune cell extravasation. In this study, we aimed to unravel the role of meprin α in PAH-induced vascular remodelling. Our results showed that meprin α was present in the apical membrane of endothelial cells in the lungs and pulmonary arteries of donors and idiopathic PAH (IPAH) patients. Elevated circulating meprin α levels were detected in the plasma of IPAH patients. In vitro binding assays and electron microscopy confirmed binding of meprin α to the glycocalyx of human pulmonary artery endothelial cells (hPAECs). Enzymatic and genetic approaches identified heparan sulphate (HS) as an important determinant of the meprin α binding capacity to hPAEC. Meprin α treatment protected from excessive neutrophil infiltration and the protective effect observed in the presence of neutrophils was partially reversed by removal of HS from hPAEC. Importantly, HS levels in pulmonary arteries were decreased in IPAH patients and binding of meprin α to HS was impaired in IPAH hPAEC. In summary, our results suggest a role of HS in docking meprin α to the endothelium and thus in the modulation of inflammatory cell extravasation. In IPAH, the decreased endothelial HS results in the reduction of meprin α binding which might contribute to enhanced inflammatory cell extravasation and potentially to pathological vascular remodelling. Topics: Animals; Cells, Cultured; Endothelium, Vascular; Heparitin Sulfate; Humans; Hypertension, Pulmonary; Immune System Diseases; Inflammation; Leukocyte Disorders; Lung; Male; Metalloendopeptidases; Mice; Mice, Inbred C57BL; Mice, Knockout; Protein Binding; Pulmonary Artery; Vascular Remodeling | 2018 |
An iminosugar-based heparanase inhibitor heparastatin (SF4) suppresses infiltration of neutrophils and monocytes into inflamed dorsal air pouches.
Local infiltration of inflammatory cells is regulated by a number of biological steps during which the cells likely penetrate through subendothelial basement membranes that contain heparan sulfate proteoglycans. In the present study, we examined whether administration of heparastatin (SF4), an iminosugar-based inhibitor of heparanase, could suppress local inflammation and degradation of heparan sulfate proteoglycans in basement membranes. In a carrageenan- or formyl peptide-induced dorsal air pouch inflammation model, the number of infiltrated neutrophils and monocytes was significantly lower in mice after topical administration of heparastatin (SF4). The concentration of chemokines MIP-2 and KC in pouch exudates of drug-treated mice was similar to control. In a zymosan-induced peritonitis model, the number of infiltrated cells was not altered in drug-treated mice. To further test how heparastatin (SF4) influences transmigration of inflammatory neutrophils, its suppressive effect on migration and matrix degradation was examined in vitro. In the presence of heparastatin (SF4), the number of neutrophils that infiltrated across a Matrigel-coated polycarbonate membrane was significantly lower, while the number of neutrophils passing through an uncoated membrane was not altered. Lysate of bone marrow-derived neutrophils released sulfate-radiolabeled macromolecules from basement membrane-like extracellular matrix, which was suppressed by heparastatin (SF4). Heparan sulfate degradation activity was almost completely abolished after incubation of lysate with protein G-conjugated anti-heparanase monoclonal antibody, strongly suggesting that the activity was due to heparanase-mediated degradation. Taken together, in a dorsal air pouch inflammation model heparastatin (SF4) potentially suppresses extravasation of inflammatory cells by impairing the degradation of basement membrane heparan sulfate. Topics: Animals; Basement Membrane; Carrageenan; Cell Movement; Cells, Cultured; Enzyme Inhibitors; Glucuronidase; Heparitin Sulfate; Humans; Imino Sugars; Inflammation; Male; Mice; Mice, Inbred C57BL; Models, Animal; Monocytes; N-Formylmethionine Leucyl-Phenylalanine; Neutrophils; Nipecotic Acids | 2016 |
Neuronal heparan sulfates promote amyloid pathology by modulating brain amyloid-β clearance and aggregation in Alzheimer's disease.
Accumulation of amyloid-β (Aβ) peptide in the brain is the first critical step in the pathogenesis of Alzheimer's disease (AD). Studies in humans suggest that Aβ clearance from the brain is frequently impaired in late-onset AD. Aβ accumulation leads to the formation of Aβ aggregates, which injure synapses and contribute to eventual neurodegeneration. Cell surface heparan sulfates (HSs), expressed on all cell types including neurons, have been implicated in several features in the pathogenesis of AD including its colocalization with amyloid plaques and modulatory role in Aβ aggregation. We show that removal of neuronal HS by conditional deletion of the Ext1 gene, which encodes an essential glycosyltransferase for HS biosynthesis, in postnatal neurons of amyloid model APP/PS1 mice led to a reduction in both Aβ oligomerization and the deposition of amyloid plaques. In vivo microdialysis experiments also detected an accelerated rate of Aβ clearance in the brain interstitial fluid, suggesting that neuronal HS either inhibited or represented an inefficient pathway for Aβ clearance. We found that the amounts of various HS proteoglycans (HSPGs) were increased in postmortem human brain tissues from AD patients, suggesting that this pathway may contribute directly to amyloid pathogenesis. Our findings have implications for AD pathogenesis and provide insight into therapeutic interventions targeting Aβ-HSPG interactions. Topics: Aged, 80 and over; Alzheimer Disease; Amyloid; Amyloid beta-Peptides; Animals; Disease Models, Animal; Heparitin Sulfate; Hippocampus; Humans; Inflammation; Mice; Neurons; Protein Aggregation, Pathological | 2016 |
Heparanase Overexpression Reduces Hepcidin Expression, Affects Iron Homeostasis and Alters the Response to Inflammation.
Hepcidin is the key regulator of systemic iron availability that acts by controlling the degradation of the iron exporter ferroportin. It is expressed mainly in the liver and regulated by iron, inflammation, erythropoiesis and hypoxia. The various agents that control its expression act mainly via the BMP6/SMAD signaling pathway. Among them are exogenous heparins, which are strong hepcidin repressors with a mechanism of action not fully understood but that may involve the competition with the structurally similar endogenous Heparan Sulfates (HS). To verify this hypothesis, we analyzed how the overexpression of heparanase, the HS degrading enzyme, modified hepcidin expression and iron homeostasis in hepatic cell lines and in transgenic mice. The results showed that transient and stable overexpression of heparanase in HepG2 cells caused a reduction of hepcidin expression and of SMAD5 phosphorylation. Interestingly, the clones showed also altered level of TfR1 and ferritin, indices of a modified iron homeostasis. The heparanase transgenic mice showed a low level of liver hepcidin, an increase of serum and liver iron with a decrease in spleen iron content. The hepcidin expression remained surprisingly low even after treatment with the inflammatory LPS. The finding that modification of HS structure mediated by heparanase overexpression affects hepcidin expression and iron homeostasis supports the hypothesis that HS participate in the mechanisms controlling hepcidin expression. Topics: Animals; Bone Morphogenetic Protein 6; Gene Expression; Gene Expression Regulation; Glucuronidase; Hep G2 Cells; Heparin; Heparitin Sulfate; Hepcidins; Homeostasis; Humans; Inflammation; Interleukin-6; Iron; Lipopolysaccharides; Liver; Mice; Signal Transduction; Smad Proteins | 2016 |
Effects of restoring normoglycemia in type 1 diabetes on inflammatory profile and renal extracellular matrix structure after simultaneous pancreas and kidney transplantation.
Patients with type 1 diabetes and end-stage renal disease with simultaneous pancreas and kidney (SPK) or kidney transplants alone (KA) were recruited 9-12 years post transplantation. We investigated differences between these groups with regard to inflammatory parameters and long-term structural changes in kidneys.. Blood samples were analyzed by ELISA and multiplex for chemokines, cytokines, growth factors, cell adhesion molecules and matrix metalloproteinases. Kidney graft biopsies were analyzed by electron microscopy for glomerular basement membrane thickness. Heparan- and chondroitin sulfate disaccharide structures were determined by size exclusion chromatography mass-spectrometry.. The SPK and the KA group had average glycated hemoglobin A1c (HbA1c) of 5.8% (40 mmol/mol) and 8.6% (70 mmol/mol) respectively. SPK recipients also had 16.2% lower body mass index (BMI) and 46.4% lower triglyceride levels compared with KA recipients, compatible with an improved metabolic profile in the SPK group. Plasminogen activator inhibitor (PAI-1), C-reactive protein (CRP) and vascular endothelial growth factor (VEGF) were lower in the SPK group. In kidney graft biopsies of the KA-patients an 81.2% increase in average glomerular basement membrane thickness was observed, accompanied by alterations in heparan sulfate proteoglycan structure. In addition to a decrease in 6-O-sulfated disaccharides, an increase in non-N-sulfated disaccharides with a corresponding slight decrease in N-sulfation was found in kidney biopsies from hyperglycemic patients.. Patients with end stage renal disease subjected to KA transplantation showed impaired inflammatory profile, increased thickness of basement membranes and distinct changes in heparan sulfate structures compared with SPK recipients. Topics: Adult; Cross-Sectional Studies; Diabetes Mellitus, Type 1; Female; Heparitin Sulfate; Humans; Hyperglycemia; Inflammation; Kidney; Kidney Failure, Chronic; Kidney Transplantation; Male; Middle Aged; Pancreas Transplantation; Proteoglycans | 2015 |
S-maltoheptaose targets syndecan-bound effectors to reduce smoking-related neutrophilic inflammation.
Cigarette smoke induces injury and neutrophilic inflammation in the airways of smokers. The stability and activity of inflammatory effectors, IL8 and neutrophil elastase (NE), can be prolonged by binding to airway heparan sulfate (HS)/syndecan-1, posing risk for developing chronic obstructive pulmonary disease(COPD). We hypothesize that antagonizing HS/syndecan-1 binding of the inflammatory effectors could reduce smoking-related neutrophil-mediated airway inflammation. Analysis of bronchoalveolar lavage fluid(BALF) of COPD patients found both total and unopposed NE levels to be significantly higher among smokers with COPD than non-COPD subjects. Similar NE burden was observed in smoke-exposed rats compared to sham air controls. We chose sulfated-maltoheptaose(SM), a heparin-mimetic, to antagonize HS/sydecan-1 binding of the inflammatory mediators in airway fluids and lung tissues of the smoke-exposed rat model. Airway treatment with SM resulted in displacement of CINC-1 and NE from complexation with bronchio-epithelial HS/syndecan-1, dissipating the chemokine gradient for neutrophil flux across to the bronchial lumen. Following SM displacement of NE from shed HS/syndecan-1 in bronchial fluids, NE became accessible to inhibition by α1-antitrypsin endogenous in test samples. The antagonistic actions of SM against syndecan-1 binding of NE and CINC-1 in smoke-exposed airways suggest new therapeutic opportunities for modulating airway inflammation in smokers with SM delivery. Topics: Aged; alpha 1-Antitrypsin; Animals; Bronchi; Bronchoalveolar Lavage Fluid; Case-Control Studies; Chemokine CXCL1; Chitosan; Disease Models, Animal; Enzyme-Linked Immunosorbent Assay; Female; Glucans; Heparitin Sulfate; Humans; Inflammation; Inflammation Mediators; Leukocyte Elastase; Male; Middle Aged; Neutrophils; Peroxidase; Pulmonary Disease, Chronic Obstructive; Rats; Rats, Sprague-Dawley; Smoking; Syndecan-1 | 2015 |
Small molecule inhibitors of protein interaction with glycosaminoglycans (SMIGs), a novel class of bioactive agents with anti-inflammatory properties.
Small molecule inhibitors of biologically important protein-glycosaminoglycan (GAG) interactions have yet to be identified.. Compound libraries were screened in an assay of L-selectin-IgG binding to heparin (a species of heparan sulfate [HS-GAG]). Hits were validated, IC-50s established and direct binding of hits to HS-GAGs was investigated by incubating compounds alone with heparin. Selectivity of inhibitors was assessed in 11 different protein-GAG binding assays. Anti-inflammatory activity of selected compounds was evaluated in animal models.. Screening identified a number of structurally-diverse planar aromatic cationic amines. Scaffolds similar to known GAG binders, chloroquine and tilorone, were also identified. Inhibitors displayed activity also against bovine kidney heparan sulfate. Direct binding of compounds to GAGs was verified by incubating compounds with heparin alone. Selectivity of inhibitors was demonstrated in a panel of 11 heparin binding proteins, including selectins, chemokines (IL-8, IP-10), Beta Amyloid and cytokines (VEGF, IL-6). A number of selected lead compounds showed dose-dependent efficacy in peritonitis, paw edema and delayed type hypersensitivity.. A new class of compounds, SMIGs, inhibits protein-GAG interaction by direct binding to GAGs. Although their IC-50s were in the low micro-molar range, SMIGs binding to HS-GAGs appeared to be stable in physiological conditions, indicating high avidity binding. SMIGs may interfere with major checkpoints for inflammatory and autoimmune events.. SMIGs are a class of structurally-diverse planar aromatic cationic amines that have an unusual mode of action - inhibiting protein-GAG interactions via direct and stable binding to GAGs. SMIGs may have therapeutic potential in inflammatory and autoimmune disorders. Topics: Animals; Anti-Inflammatory Agents; Carrageenan; Cattle; Chemokines; Cytokines; Edema; Glycosaminoglycans; Heparin; Heparitin Sulfate; High-Throughput Screening Assays; Humans; Hypersensitivity, Delayed; Immunoglobulin G; Inflammation; L-Selectin; Leukocytes; Mice; Mice, Inbred BALB C; Mice, Inbred ICR; Molecular Structure; Neutrophils; Protein Binding; Protein Interaction Domains and Motifs; Small Molecule Libraries | 2014 |
Fibrinogen, an endogenous ligand of Toll-like receptor 4, activates monocytes in pre-eclamptic patients.
Pre-eclampsia (PE) remains the leading cause of pregnancy-associated mortality and morbidity, urging the need for a better understanding of its aetiology and pathophysiological progression. A key characteristic of PE is a systemic, exaggerated, inflammatory condition involving abnormal cytokine levels in serum, altered immune cell phenotype and Th1/Th2-type immunological imbalance. However, it is unknown how this heightened inflammatory condition manifests. We previously reported increased expression of the lipopolysaccharide receptor, Toll-like receptor 4 (TLR4), on monocytes from PE patients compared with normotensive, pregnant patients (NP). This upregulation of TLR4 on PE monocytes was accompanied by a hyper-responsiveness to bacterial TLR4 ligands. To determine whether non-microbial, endogenous TLR4 ligands also activate monocytes from PE patients, we investigated the expression of host-derived TLR4 ligands and the response of monocytes to these endogenous ligands. Plasma levels of fibrinogen - but not fibronectin or heparan sulphate - were higher in PE patients than in NP. Exposure to fibrinogen was associated with significantly increased production of inflammatory cytokines by monocytes from PE patients. Interestingly, this effect was not observed with NP monocytes. Our findings suggest that the fibrinogen-TLR4 axis might play an important role in the atypical activation of monocytes observed in PE patients that may contribute to the exaggerated inflammatory condition. Topics: Adult; Cytokines; Female; Fibrinogen; Fibronectins; Heparitin Sulfate; Humans; Hypertension; Inflammation; Monocytes; Pre-Eclampsia; Pregnancy; Proteinuria; Toll-Like Receptor 4 | 2014 |
Endothelial and leukocyte heparan sulfates regulate the development of allergen-induced airway remodeling in a mouse model.
Heparan sulfate (HS) proteoglycans (HSPGs) participate in several aspects of inflammation because of their ability to bind to growth factors, chemokines, interleukins and extracellular matrix proteins as well as promote inflammatory cell trafficking and migration. We investigated whether HSPGs play a role in the development of airway remodeling during chronic allergic asthma using mice deficient in endothelial- and leukocyte-expressed N-deacetylase/N-sulfotransferase-1 (Ndst1), an enzyme involved in modification reactions during HS biosynthesis. Ndst1-deficient and wild-type (WT) mice exposed to repetitive allergen (ovalbumin [OVA]) challenge were evaluated for the development of airway remodeling. Chronic OVA-challenged WT mice exhibited increased HS expression in the lungs along with airway eosinophilia, mucus hypersecretion, peribronchial fibrosis, increased airway epithelial thickness and smooth muscle mass. In OVA-challenged Ndst1-deficient mice, lung eosinophil and macrophage infiltration as well as airway mucus accumulation, peribronchial fibrosis and airway epithelial thickness were significantly lower than in allergen-challenged WT mice along with a trend toward decreased airway smooth muscle mass. Leukocyte and endothelial Ndst 1 deficiency also resulted in significantly decreased expression of IL-13 as well as remodeling-associated mediators such as VEGF, FGF-2 and TGF-β1 in the lung tissue. At a cellular level, exposure to eotaxin-1 failed to induce TGF-β1 expression by Ndst1-deficient eosinophils relative to WT eosinophils. These studies suggest that leukocyte and endothelial Ndst1-modified HS contribute to the development of allergen-induced airway remodeling by promoting recruitment of inflammatory cells as well as regulating expression of pro-remodeling factors such as IL-13, VEGF, TGF-β1 and FGF-2 in the lung. Topics: Airway Remodeling; Allergens; Animals; Endothelial Cells; Heparitin Sulfate; Inflammation; Leukocytes; Mice; Mice, Inbred C57BL; Models, Animal; Proteoglycans | 2014 |
Interference with glycosaminoglycan-chemokine interactions with a probe to alter leukocyte recruitment and inflammation in vivo.
In vivo leukocyte recruitment is not fully understood and may result from interactions of chemokines with glycosaminoglycans/GAGs. We previously showed that chlorite-oxidized oxyamylose/COAM binds the neutrophil chemokine GCP-2/CXCL6. Here, mouse chemokine binding by COAM was studied systematically and binding affinities of chemokines to COAM versus GAGs were compared. COAM and heparan sulphate bound the mouse CXC chemokines KC/CXCL1, MIP-2/CXCL2, IP-10/CXCL10 and I-TAC/CXCL11 and the CC chemokine RANTES/CCL5 with affinities in the nanomolar range, whereas no binding interactions were observed for mouse MCP-1/CCL2, MIP-1α/CCL3 and MIP-1β/CCL4. The affinities of COAM-interacting chemokines were similar to or higher than those observed for heparan sulphate. Although COAM did not display chemotactic activity by itself, its co-administration with mouse GCP-2/CXCL6 and MIP-2/CXCL2 or its binding of endogenous chemokines resulted in fast and cooperative peritoneal neutrophil recruitment and in extravasation into the cremaster muscle in vivo. These local GAG mimetic features by COAM within tissues superseded systemic effects and were sufficient and applicable to reduce LPS-induced liver-specific neutrophil recruitment and activation. COAM mimics glycosaminoglycans and is a nontoxic probe for the study of leukocyte recruitment and inflammation in vivo. Topics: Amino Acid Sequence; Amylose; Animals; Cell Adhesion; Cell Communication; Cell Movement; Chemokines; Endothelial Cells; Extracellular Traps; Female; Glycosaminoglycans; Heparitin Sulfate; Inflammation; Injections, Intraperitoneal; Isoelectric Point; Kinetics; Lipopolysaccharides; Liver; Male; Mice, Inbred C57BL; Molecular Sequence Data; Muscles; Neutrophil Infiltration; Peritoneal Cavity; Surface Plasmon Resonance | 2014 |
Individualized early goal-directed therapy in systemic inflammation: is full utilization of preload reserve the optimal strategy?
In severe acute pancreatitis, the administration of fluids in the presence of positive fluid responsiveness is associated with better outcome when compared to guiding therapy on central venous pressure. We compared the effects of such consequent maximization of stroke volume index with a regime using individual values of stroke volume index assessed prior to severe acute pancreatitis induction as therapeutic hemodynamic goals.. Prospective, randomized animal study.. University animal research laboratory.. Thirty domestic pigs.. After randomization, fluid resuscitation was started 2 hours after severe acute pancreatitis induction and continued for 6 hours according to the respective treatment algorithms. In the control group, fluid therapy was directed by maximizing stroke volume index, and in the study group, stroke volume index assessed prior to severe acute pancreatitis served as primary hemodynamic goal.. Within the first 6 hours of severe acute pancreatitis, the study group received a total of 1,935.8 ± 540.7 mL of fluids compared with 3,462.8 ± 828.2 mL in the control group (p < 0.001). Pancreatic tissue oxygenation did not differ significantly between both groups. Vascular endothelial function, measured by flow-mediated vasodilation before and 6 hours after severe acute pancreatitis induction, revealed less impairment in the study group after treatment interval (-90.76% [study group] vs -130.89% [control group]; p = 0.046). Further, lower levels of heparan sulfate (3.41 ± 5.6 pg/mL [study group] vs 43.67 ± 46.61 pg/mL [control group]; p = 0.032) and interleukin 6 (32.18 ± 8.81 pg/mL [study group] vs 77.76 ± 56.86 pg/mL [control group]; p = 0.021) were found in the study group compared with control group. Histopathological examination of the pancreatic head and corpus at day 7 revealed less edema for the study group compared with the control group (1.82 ± 0.87 [study group] vs 2.89 ± 0.33 [control group, pancreatic head]; p = 0.03; 2.2 ± 0.92 [study group] vs 2.91 ± 0.3 [control group, pancreatic corpus]; p = 0.025).. Individualized optimization of intravascular fluid status during the early course of severe acute pancreatitis, compared with a treatment strategy of maximizing stroke volume by fluid loading, leads to less vascular endothelial damage, pancreatic edema, and inflammatory response. Topics: Acute Disease; Animals; Disease Models, Animal; Endothelium, Vascular; Enzyme-Linked Immunosorbent Assay; Fluid Therapy; Glycocalyx; Hemodynamics; Heparitin Sulfate; Inflammation; Pancreatitis; Prospective Studies; Random Allocation; Severity of Illness Index; Stroke Volume; Swine; Syndecan-1 | 2014 |
Development of cerebellar pathology in the canine model of mucopolysaccharidosis type IIIA (MPS IIIA).
The temporal relationship between the onset of clinical signs in the mucopolysaccharidosis type IIIA (MPS IIIA) Huntaway dog model and cerebellar pathology has not been described. Here we sought to characterize the accumulation of primary (heparan sulfate) and secondary (G(M3)) substrates and onset of other changes in cerebellar tissues, and investigate the relationship to the onset of motor dysfunction in these animals. We observed that Purkinje cells were present in dogs aged up to and including 30.9 months, however by 40.9 months of age only ~12% remained, coincident with the onset of clinical signs. Primary and secondary substrate accumulation and inflammation were detected as early as 2.2 months and axonal spheroids were observed from 4.3 months in the deep cerebellar nuclei and later (11.6 months) in cerebellar white matter tracts. Degenerating neurons and apoptotic cells were not observed at any time. Our findings suggest that cell autonomous mechanisms may contribute to Purkinje cell death in the MPS IIIA dog. Topics: Animals; Asymptomatic Diseases; Cell Death; Cerebellar Nuclei; Cerebellum; Disease Models, Animal; Dogs; Heparitin Sulfate; Humans; Inflammation; Motor Activity; Mucopolysaccharidosis III; Purkinje Cells; White Matter | 2014 |
Molecular imaging of heparan sulfate expression with radiolabeled recombinant eosinophil cationic protein predicts allergic lung inflammation in a mouse model for asthma.
Heparan sulfate proteoglycans (HSPGs) are glycoproteins consisting of a core protein to which linear heparan sulfate (HS) side chains are covalently attached. These HS side chains mediate a variety of biologic functions involved in inflammation. Radionuclide imaging of HS side chains in tissues with inflammation may be used for the stratification of patients who would most likely benefit from HSPG-targeting therapy. The goal of this study was to evaluate the feasibility of in vivo radionuclide imaging of HS side chain expression in a mouse model of asthma using the recombinant eosinophil cationic protein (rECP).. rECP was radioiodinated with (125)I or (123)I using the Chloramine-T method. The 50% inhibitory concentration value for (125)I-labeled rECP was determined in a competitive cell-binding assay using Beas-2B cells. The binding of radiolabeled rECP to HS side chains was evaluated both in vitro and in vivo. The biodistribution of radiolabeled rECP was assessed in asthma mice or in control mice using SPECT imaging, ex vivo biodistribution measurements, and microautoradiography.. The 50% inhibitory concentration value for (125)I-rECP was 7.4 ± 0.1 nM. The loss of HS side chains substantially inhibited the cellular and tissue uptake of (125)I- or (123)I-rECP, indicating that HS side chains of HSPGs are required for (125)I- or (123)I-eosinophil cationic protein binding and uptake both in vitro and in vivo. SPECT imaging demonstrated an appreciably higher accumulation of radioactivity in the lungs of asthma mice than in those of control mice. Ex vivo biodistribution studies also confirmed that there was at least a 4-fold increase in the lung-to-muscle ratio of asthma mice, compared with control mice. The accumulation of radiolabeled rECP was linearly correlated with leukocyte infiltration.. This study illustrates the feasibility of using radiolabeled rECP for the visualization of HS side chains of HSPGs and the evaluation of allergic lung inflammation in living subjects. Our data indicate that radiolabeled rECP is a novel imaging agent for HS side chains of HSPGs in predicting allergic lung inflammation in living mice. Topics: Allergens; Animals; Asthma; Biological Transport; Cell Line, Tumor; Disease Models, Animal; Drug Stability; Eosinophil Cationic Protein; Female; Gene Expression Regulation; Heparitin Sulfate; Humans; Hypersensitivity; Inflammation; Iodine Radioisotopes; Isotope Labeling; Leukocytes; Mice; Molecular Imaging; Multimodal Imaging; Positron-Emission Tomography; Radiochemistry; Recombinant Proteins; Tomography, X-Ray Computed | 2013 |
Expression of heparan sulfate proteoglycans in murine models of experimental colitis.
Heparan sulfate proteoglycans (HSPGs) are considered important in maintaining physiological homeostasis in many systems. Their expression is altered greatly in several pathophysiological conditions. Herein, we assess the expression and cellular localization of HSPGs in two murine models of human inflammatory bowel disease (IBD).. Expression and localization of HSPGs, syndecans, and HS epitopes were examined in the colon of 129SvEv interleukin 10 knockout (IL10(-/-)), C3Bir IL10(-/-), and their genetic control (IL10(+/+)) counterparts (129SvEv; C3H/HeJ). mRNA expression of syndecans and heparan sulfate biosynthesis enzymes were evaluated by real-time polymerase chain reaction (PCR). Localization of HSPGs was determined by immunofluorescence.. mRNA for all syndecans was detected and expression in colonic tissues altered in IL10(-/-) mice. Syndecan-1 protein was expressed in the intestinal epithelium and on lamina propria cells of IL10(-/-) and control mice but was significantly reduced on the intestinal epithelial cells of IL10(-/-), mice particularly with severe colitis. Syndecan-2 was not detected, whereas syndecan-3 immunoreactivity was localized in the lamina propria but did not differ between control and IL10(-/-) mice. Syndecan-4 was present on epithelial cells of all mice but was significantly reduced in IL10(-/-) mice. Differences in the expression of HS epitopes between control and IL10(-/-) mice were also confirmed.. The study has revealed altered expression of syndecan-1 and -4 and HS epitopes in the gut of mice with an IBD-like gut disorder. The IL10(-/-) mouse is a useful model for further study of the functional role of HSPGs in chronic inflammation and in maintaining healthy gut barrier. Topics: Animals; Blotting, Western; Cells, Cultured; Colitis; Colon; Disease Models, Animal; Female; Fluorescent Antibody Technique; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Inflammation; Interleukin-10; Intestinal Mucosa; Mice; Mice, Inbred C3H; Mice, Knockout; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Syndecan-1; Syndecan-2; Syndecan-3; Syndecan-4; Syndecans | 2012 |
Inactivation of heparan sulfate 2-O-sulfotransferase accentuates neutrophil infiltration during acute inflammation in mice.
Neutrophil recruitment and extravasation at sites of inflammation provide a mechanism for host defense. We showed previously that heparan sulfate, a type of sulfated glycosaminoglycan, facilitates neutrophil recruitment based on the reduction of neutrophil infiltration in mice in which the overall sulfation of the chains was reduced by selective inactivation of N-acetylglucosamine N-deacetylase-N-sulfotransferase (Ndst1) in endothelial cells. Here we show that inactivation of uronyl 2-O-sulfotransferase in endothelial cells (Hs2st), an enzyme that acts downstream from Ndst1, results in enhanced neutrophil recruitment in several models of acute inflammation. Enhanced neutrophil infiltration resulted in part from reduced rolling velocity under flow both in vivo and in vitro, which correlated with stronger binding of neutrophil L-selectin to mutant endothelial cells. Hs2st-deficient endothelial cells also displayed a striking increase in binding of IL-8 and macrophage inflammatory protein-2. The enhanced binding of these mediators of neutrophil recruitment resulted from a change in heparan sulfate structure caused by increased N-sulfation and 6-O-sulfation of glucosamine units in response to the decrease in 2-O-sulfation of uronic acid residues. This gain-of-function phenotype provides formidable evidence demonstrating the importance of endothelial heparan sulfate in inflammation and suggests a novel enzyme target for enhancing the innate immune response. Topics: Animals; Cells, Cultured; Chemokines; Endothelial Cells; Gene Silencing; Heparitin Sulfate; Inflammation; L-Selectin; Male; Mice; Mice, Inbred C57BL; Neutrophil Infiltration; Neutrophils; Peritonitis; Sulfotransferases; Thioglycolates | 2012 |
Accelerated resolution of AA amyloid in heparanase knockout mice is associated with matrix metalloproteases.
AA-amyloidosis is a disease characterized by abnormal deposition of serum A amyloid (SAA) peptide along with other components in various organs. The disease is a complication of inflammatory conditions that cause persistent high levels of the acute phase reactant SAA in plasma. In experimental animal models, the deposited amyloid is resolved when the inflammation is stopped, suggesting that there is an efficient clearance mechanism for the amyloid. As heparan sulfate (HS) is one of the major components in the amyloid, its metabolism is expected to affect the pathology of AA amyloidosis. In this study, we investigated the effect of heparanase, a HS degradation enzyme, in resolution of the AA amyloid. The transgenic mice deficient in heparanase (Hpa-KO) produced a similar level of SAA in plasma as the wildtype control (Ctr) mice upon induction by injection of AEF (amyloid enhancing factor) and inflammatory stimuli. The induction resulted in formation of SAA amyloid 7-days post treatment in the spleen that displayed a comparable degree of amyloid load in both groups. The amyloid became significantly less in the Hpa-KO spleen than in the Ctr spleen 10-days post treatment, and was completely resolved in the Hpa-KO spleen on day 21 post induction, while a substantial amount was still detected in the Ctr spleen. The rapid clearance of the amyloid in the Hpa-KO mice can be ascribed to upregulated matrix metalloproteases (MMPs) that are believed to contribute to degradation of the protein components in the AA amyloid. The results indicate that both heparanase and MMPs play important parts in the pathological process of AA amyloidosis. Topics: Amyloidosis; Animals; Glucuronidase; Glycoproteins; Heparitin Sulfate; Inflammation; Matrix Metalloproteinases; Mice; Mice, Knockout; Serum Amyloid A Protein; Signal Transduction; Silver Nitrate; Spleen; Up-Regulation | 2012 |
Danaparoid sodium attenuates the effects of heat stress.
Heat stroke is a condition characterized by high body temperature that can lead to hemorrhage and necrosis in multiple organs. Anticoagulants, such as danaparoid sodium (DA), inhibit various types of inflammation; however, the anti-inflammatory mechanism of action is not well understood. Given that heat stroke is a severe inflammatory response disease, we hypothesized that DA could inhibit inflammation from heat stress and prevent acute heat stroke.. Male Wistar rats were given a bolus injection of saline or DA (50 U/kg body weight) into the tail vein just prior to heat stress (42 °C for 30 min). Markers of inflammation were then determined in serum and tissue samples.. In rats pretreated with DA, induction of cytokines (interleukin [IL]-1β, IL-6, and tumor necrosis factor [TNF]-α), nitric oxide (NO), and high mobility group box 1 (HMGB1) protein were reduced compared with saline-treated rats. Histologic changes observed in lung, liver, and small intestine tissue samples of saline-treated rats were attenuated in DA-treated rats. Moreover, DA pretreatment improved survival in our rat model of heat stress-induced acute inflammation.. Collectively, our findings demonstrate that DA pretreatment may have value as a new therapeutic tool for heat stroke. Topics: Acute Disease; Animals; Anticoagulants; Antithrombin III; Chondroitin Sulfates; Cytokines; Dermatan Sulfate; Disease Models, Animal; Fibrin Fibrinogen Degradation Products; Heat Exhaustion; Heat Stroke; Heparitin Sulfate; HMGB1 Protein; Inflammation; Intestine, Small; Liver; Lung; Male; Nitrates; Nitric Oxide; Nitrites; Peptide Hydrolases; Rats; Rats, Wistar; Survival Rate | 2011 |
Adenosine reduces cell surface expression of toll-like receptor 4 and inflammation in response to lipopolysaccharide and matrix products.
Recent evidence suggests that Toll-like receptor 4 (TLR4) is not only involved in innate immunity but is also an important mediator of adverse left ventricular remodeling and heart failure following acute myocardial infarction (MI). TLR4 is activated by lipopolysaccharide (LPS) but also by products of matrix degradation such as hyaluronic acid and heparan sulfate. Although cardioprotective properties of adenosine (Ado) have been extensively studied, its potential to interfere with TLR4 activation is unknown. We observed that TLR4 pathway is activated in white blood cells from MI patients. TLR4 mRNA expression correlated with troponin T levels (R (2) = 0.75; P = 0.01) but not with levels of white blood cells and C-reactive protein. Ado downregulated TLR4 expression at the surface of human macrophages (-50%, P < 0.05). Tumor necrosis factor-α production induced by the TLR4 ligands LPS, hyaluronic acid, and heparan sulfate was potently inhibited by Ado (-75% for LPS, P < 0.005). This effect was reproduced by the A2A Ado receptor agonist CGS21680 and the non-selective agonist NECA and was inhibited by the A2A antagonist SCH58261 and the A2A/A2B antagonist ZM241,385. In contrast, Ado induced a 3-fold increase of TLR4 mRNA expression (P = 0.008), revealing the existence of a feedback mechanism to compensate for the loss of TLR4 expression at the cell surface. In conclusion, the TLR4 pathway is activated after MI and correlates with infarct severity but not with the extent of inflammation. Reduction of TLR4 expression by Ado may therefore represent an important strategy to limit remodeling post-MI. Topics: Adenosine; Adenosine A2 Receptor Agonists; Adenosine A2 Receptor Antagonists; Adult; Aged; Case-Control Studies; Cell Membrane; Cells, Cultured; Dose-Response Relationship, Drug; Down-Regulation; Female; Heparitin Sulfate; Humans; Hyaluronic Acid; Inflammation; Ligands; Lipopolysaccharides; Luxembourg; Macrophages; Male; Middle Aged; Myocardial Infarction; Receptor, Adenosine A2A; Receptor, Adenosine A2B; Registries; RNA, Messenger; Time Factors; Toll-Like Receptor 4; Tumor Necrosis Factor-alpha | 2011 |
Nebulized anticoagulants limit coagulopathy but not inflammation in pseudomonas aeruginosa-induced pneumonia in rats.
Disturbed alveolar fibrin turnover is a characteristic feature of pneumonia. Inhibitors of coagulation could exert lung-protective effects via anticoagulant (inhibiting fibrin deposition) and possibly anti-inflammatory pathways, but could also affect host defense. In this randomized controlled in vivo laboratory study, rats were challenged intratracheally with Pseudomonas aeruginosa, inducing pneumonia, and randomized to local treatment with normal saline (placebo), recombinant human activated protein C (rh-APC), plasma-derived antithrombin (AT), heparin, or danaparoid. Induction of P. aeruginosa pneumonia resulted in activation of pulmonary coagulation and inhibition of pulmonary fibrinolysis, as reflected by increased pulmonary levels of thrombin-AT complexes and fibrin degradation products and decreased pulmonary levels plasminogen activator activity. Pseudomonas aeruginosa pneumonia was accompanied by systemic coagulopathy, since systemic levels of thrombin-AT complexes increased, and systemic levels of plasminogen activator activity decreased. Although rh-APC and plasma-derived AT potently limited pulmonary coagulopathy, neither heparin nor danaparoid affected net pulmonary fibrin turnover. Recombinant human APC also displayed systemic anticoagulant effects. Neither bacterial clearance nor pulmonary inflammation was affected by anticoagulant therapy. Nebulization of rh-APC or plasma-derived AT attenuated pulmonary coagulopathy, but not bacterial clearance or inflammation, in a rat model of P. aeruginosa pneumonia. Topics: Animals; Anticoagulants; Antithrombins; Blood Coagulation; Chondroitin Sulfates; Dermatan Sulfate; Fibrinolysis; Heparin; Heparitin Sulfate; Inflammation; Male; Pneumonia; Pseudomonas aeruginosa; Rats; Rats, Sprague-Dawley | 2011 |
TLR4 dependent heparan sulphate-induced pancreatic inflammatory response is IRF3-mediated.
Degraded extracellular matrix can stimulate the innate immune system via the Toll-Like Receptor-4 (TLR4). In the pancreas, syndecan-anchored heparan sulphate (HS) on the ductal epithelium can be cleaved off its protein cores by the proteases (trypsin and elastase) and potentially activate TLR4 signalling.. To investigate this signalling event, a low sulphated HS (500 μg/ml) was infused into the biliary-pancreatic duct of C57BL/6J wild-type mice. Phosphate buffered saline (PBS) and lipopolysaccharide (LPS) were used as negative and positive controls, respectively. Mice were sacrificed after 1, 3, 6, 9, and 48 hours and tissues were analysed for neutrophil and cytokine contents. In order to study the TLR4 signalling pathway of HS in the pancreas, genetically engineered mice lacking TLR4, Myeloid Differentiation primary response gene (88) (MyD88) or Interferon Regulatory Factor 3 (IRF3) were subjected to pancreatic infusion of HS.. Neutrophil sequestration and corresponding myeloperoxidase (MPO) activity in the pancreas were increased 9 hours following HS challenge. In wild-type mice, the monocyte chemoattractant protein-1(MCP-1) increased at 3 hours after infusion, while RANTES increased after 9 hours.TLR4, MyD88, and IRF3 knockout mice showed an abrogated neutrophil recruitment and myeloperoxidase activity in the HS group, while the LPS response was only abolished in TLR4 and MyD88 knockouts.. The results of this study show that HS is capable of initiating a TLR4-dependent innate immune response in the pancreas which is distinctly different from that induced by LPS. This inflammatory response was mediated predominantly through IRF3- dependent pathway. Release of HS into the pancreatic duct may be one important mediator in the pancreatic ductal defence. Topics: Animals; Chemokine CCL5; Chemotactic Factors; Cytokines; Disaccharides; Heparitin Sulfate; Inflammation; Interferon Regulatory Factor-3; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Neutrophil Infiltration; Pancreas; Peroxidase; Sugar Phosphates; Time Factors; Toll-Like Receptor 4 | 2011 |
Composition of the endothelial glycocalyx and its relation to its thickness and diffusion of small solutes.
The endothelial glycocalyx is well endowed with the glycosaminoglycans (GAGs) heparan sulfate, chondroitin sulfate and hyaluronan. The current studies aimed to assess the relative contributions of each of these GAGs to the thickness and permeability of the glycocalyx layer by direct enzymatic removal of each using micropipettes to infuse heparinase, chondroitinase and hyaluronidase into post-capillary venules of the intestinal mesentery of the rat. The relative losses of GAGs due to enzymatic removal were compared with stimulated shedding of glycans induced by superfusing the mesentery with 10(-)(7)M fMLP. Thickness of the glycocalyx was assessed by infiltration of the glycocalyx with circulating FITC labeled 70kDa dextran (Dx70) and measuring the distance from the dye front to the surface of the endothelium (EC), which averaged 463nm under control conditions. Reductions in thickness were 43.3%, 34.1% and 26.1% following heparinase, chondroitinase and hyaluronidase, respectively, and 89.7% with a mixture of all three enzymes. Diffusion coefficients of FITC in the glycocalyx were determined using a 1-D diffusion model. By comparison of measured transients in radial intensity of a bolus of FITC with that of a computational model a diffusion coefficient D was obtained. Values of D were obtained corresponding to the thickness of the layer demarcated by Dx70 (D(Dx70)), and a smaller sublayer 173nm above the EC surface (D(173)), prior to and following enzyme infusion and superfusion with fMLP. The magnitude of D(Dx70) was twice that of D(173) suggesting that the glycocalyx is more compact near the EC surface. Chondroitinase and hyaluronidase significantly increased both D(Dx70) and D(173). However, heparinase decreased D(Dx70), and did not induce any significant change for the D(173). These observations suggest that the three GAGs are not evenly distributed throughout the glycocalyx and that they each contribute to permeability of the glycocalyx to a differing extent. The fMLP-induced shedding caused a reduction in glycocalyx thickness (which may increase permeability) and as with heparinase, decreased the diffusion coefficient of solutes (which may decrease permeability). This behavior suggests that the removal of heparan sulfate may cause a collapse of the glycocalyx which counters decreases in thickness by compacting the layer to maintain a constant resistance to filtration. Topics: Animals; Cell Membrane Permeability; Chondroitin Sulfates; Chondroitinases and Chondroitin Lyases; Computer Simulation; Dextrans; Diffusion; Endothelial Cells; Fluorescein-5-isothiocyanate; Fluorescent Dyes; Glycocalyx; Glycosaminoglycans; Heparin Lyase; Heparitin Sulfate; Hyaluronic Acid; Hyaluronoglucosaminidase; Inflammation; Infusions, Intravenous; Male; Mesentery; Models, Cardiovascular; N-Formylmethionine Leucyl-Phenylalanine; Rats; Rats, Wistar; Venules | 2010 |
Deficiency of endothelial heparan sulfates attenuates allergic airway inflammation.
The effect of targeted inactivation of the gene encoding N-deacetylase/N-sulfotransferase-1 (Ndst1), a key enzyme involved in the biosynthesis of heparan sulfate (HS) chains, on the inflammatory response associated with allergic inflammation in a murine model of OVA-induced acute airway inflammation was investigated. OVA-exposed Ndst1(f/f)TekCre(+) (mutant) mice deficient in endothelial and leukocyte Ndst1 demonstrated significantly decreased allergen-induced airway hyperresponsiveness and inflammation characterized by a significant reduction in airway recruitment of inflammatory cells (eosinophils, macrophages, neutrophils, and lymphocytes), diminished IL-5, IL-2, TGF-beta1, and eotaxin levels, as well as decreased expression of TGF-beta1 and the angiogenic protein FIZZ1 (found in inflammatory zone 1) in lung tissue compared with OVA-exposed Ndst1(f/f)TekCre(-) wild-type littermates. Furthermore, murine eosinophils demonstrated significantly decreased rolling on lung endothelial cells (ECs) from mutant mice compared with wild-type ECs under conditions of flow in vitro. Treatment of wild-type ECs, but not eosinophils, with anti-HS Abs significantly inhibited eosinophil rolling, mimicking that observed with Ndst1-deficient ECs. In vivo, trafficking of circulating leukocytes in lung microvessels of allergen-challenged Ndst1-deficient mice was significantly lower than that observed in corresponding WT littermates. Endothelial-expressed HS plays an important role in allergic airway inflammation through the regulation of recruitment of inflammatory cells to the airways by mediating interaction of leukocytes with the vascular endothelium. Furthermore, HS may also participate by sequestering and modulating the activity of allergic asthma-relevant mediators such as IL-5, IL-2, and TGF-beta1. Topics: Animals; Chemotaxis; Endothelial Cells; Heparitin Sulfate; Inflammation; Interleukin-2; Interleukin-5; Leukocytes; Mice; Respiratory Hypersensitivity; Sulfotransferases; Transforming Growth Factor beta1 | 2009 |
Heparan sulfate in perlecan promotes mouse atherosclerosis: roles in lipid permeability, lipid retention, and smooth muscle cell proliferation.
Heparan sulfate (HS) has been proposed to be antiatherogenic through inhibition of lipoprotein retention, inflammation, and smooth muscle cell proliferation. Perlecan is the predominant HS proteoglycan in the artery wall. Here, we investigated the role of perlecan HS chains using apoE null (ApoE0) mice that were cross-bred with mice expressing HS-deficient perlecan (Hspg2(Delta3/Delta3)). Morphometry of cross-sections from aortic roots and en face preparations of whole aortas revealed a significant decrease in lesion formation in ApoE0/Hspg2(Delta3/Delta3) mice at both 15 and 33 weeks. In vitro, binding of labeled mouse triglyceride-rich lipoproteins and human LDL to total extracellular matrix, as well as to purified proteoglycans, prepared from ApoE0/Hspg2(Delta3/Delta3) smooth muscle cells was reduced. In vivo, at 20 minutes influx of human (125)I-LDL or mouse triglyceride-rich lipoproteins into the aortic wall was increased in ApoE0/Hspg2(Delta3/Delta3) mice compared to ApoE0 mice. However, at 72 hours accumulation of (125)I-LDL was similar in ApoE0/Hspg2(Delta3/Delta3) and ApoE0 mice. Immunohistochemistry of lesions from ApoE0/Hspg2(Delta3/Delta3) mice showed decreased staining for apoB and increased smooth muscle alpha-actin content, whereas accumulation of CD68-positive inflammatory cells was unchanged. We conclude that the perlecan HS chains are proatherogenic in mice, possibly through increased lipoprotein retention, altered vascular permeability, or other mechanisms. The ability of HS to inhibit smooth muscle cell growth may also influence development as well as instability of lesions. Topics: Actins; Animals; Antigens, CD; Antigens, Differentiation, Myelomonocytic; Aorta; Apolipoproteins B; Apolipoproteins E; Atherosclerosis; Capillary Permeability; Cell Proliferation; Crosses, Genetic; Disease Models, Animal; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Inflammation; Lipoproteins, LDL; Mice; Mice, Knockout; Myocytes, Smooth Muscle; Protein Binding; Triglycerides | 2008 |
Human follicular fluid heparan sulfate contains abundant 3-O-sulfated chains with anticoagulant activity.
Anticoagulant heparan sulfate proteoglycans bind and activate antithrombin by virtue of a specific 3-O-sulfated pentasaccharide. They not only occur in the vascular wall but also in extravascular tissues, such as the ovary, where their functions remain unknown. The rupture of the ovarian follicle at ovulation is one of the most striking examples of tissue remodeling in adult mammals. It involves tightly controlled inflammation, proteolysis, and fibrin deposition. We hypothesized that ovarian heparan sulfates may modulate these processes through interactions with effector proteins. Our previous work has shown that anticoagulant heparan sulfates are synthesized by rodent ovarian granulosa cells, and we now have set out to characterize heparan sulfates from human follicular fluid. Here we report the first anticoagulant heparan sulfate purified from a natural human extravascular source. Heparan sulfate chains were fractionated according to their affinity for antithrombin, and their structure was analyzed by 1H NMR and MS/MS. We find that human follicular fluid is a rich source of anticoagulant heparan sulfate, comprising 50.4% of total heparan sulfate. These antithrombin-binding chains contain more than 6% 3-O-sulfated glucosamine residues, convey an anticoagulant activity of 2.5 IU/ml to human follicular fluid, and have an anti-Factor Xa specific activity of 167 IU/mg. The heparan sulfate chains that do not bind antithrombin surprisingly exhibit an extremely high content in 3-O-sulfated glucosamine residues, which suggest that they may exhibit biological activities through interactions with other proteins. Topics: Anticoagulants; Chromatography, Gel; Chromatography, Ion Exchange; Female; Follicular Fluid; Heparitin Sulfate; Humans; Inflammation; Magnetic Resonance Spectroscopy; Mass Spectrometry; Models, Biological; Models, Chemical; Ovary; Sulfur; Sulfuric Acid Esters | 2008 |
Anti-inflammatory properties of a heparin-like glycosaminoglycan with reduced anti-coagulant activity isolated from a marine shrimp.
The anti-inflammatory properties of a heparin-like compound from the shrimp Litopenaeus vannamei are related. Besides reducing significantly (p<0.001) the influx of inflammatory cells to injury site in a model of acute inflammation, shrimp heparin-like compound was able to reduce the matrix metalloproteinase (MMPs) activity in the peritoneal lavage of inflamed animals. Moreover, this compound also reduced almost 90% the activity of MMP-9 secreted by human activated leukocytes. Negligible anti-coagulant activities in aPPT assay and a poor bleeding potential make this compound a better alternative than mammalian heparin as a possible anti-inflammatory drug. Topics: Animals; Anti-Inflammatory Agents; Anticoagulants; Endothelium, Vascular; Glycosaminoglycans; Hemorrhage; Heparin; Heparitin Sulfate; Inflammation; Leukocytes; Matrix Metalloproteinase 9; Neutrophils; Penaeidae; Peritoneal Cavity; Rabbits; Rats; Swine | 2008 |
Danaparoid sodium reduces ischemia/reperfusion-induced liver injury in rats by attenuating inflammatory responses.
This study was undertaken to examine the mechanism by which danaparoid sodium (DS), a heparinoid that contains mainly heparan sulfate, prevents reperfusion-induced hepatic damage in a rat model of ischemia/reperfusion (I/R)-induced liver injury. Administration of DS significantly reduced liver injury and inhibited the decrease in hepatic tissue blood flow in rats. DS attenuated hepatic I/R-induced increases in hepatic tissue levels of tumor necrosis factor (TNF) and myeloperoxidase (MPO) in vivo. In contrast, neither monocytic TNF production nor neutrophil activation was inhibited by DS in vitro. DS enhanced I/R-induced increases in levels of calcitonin-gene related peptide (CGRP), a neuropeptide released from sensory neurons, and of 6-ketoprostaglandin (PG) F (1a) , a stable metabolite of PGI (2) , in liver tissues. The therapeutic effects of DS were not seen in animals pretreated with capsazepine, an inhibitor of sensory neuron activation. The distribution of heparan sulfate in the perivascular area was significantly increased by DS administration in this rat model. DS significantly increased CGRP release from isolated rat dorsal root ganglion neurons (DRG) in vitro, while DX-9065a, a selective inhibitor of activated factor X, did not. DS enhanced anandamide-induced CGRP release from DRG in vitro. These observations strongly suggested that DS might reduce I/R-induced liver injury in rats by attenuating inflammatory responses. These therapeutic effects of DS might be at least partly explained by its enhancement of sensory neuron activation, leading to the increase the endothelial production of PGI (2) . Topics: Animals; Calcitonin Gene-Related Peptide; Chondroitin Sulfates; Dermatan Sulfate; Epoprostenol; Ganglia, Spinal; Heparitin Sulfate; Inflammation; Liver; Male; Neurons, Afferent; Rats; Rats, Wistar; Regional Blood Flow; Reperfusion Injury | 2007 |
Eotaxin selectively binds heparin. An interaction that protects eotaxin from proteolysis and potentiates chemotactic activity in vivo.
An important feature of chemokines is their ability to bind to the glycosaminoglycan (GAG) side chains of proteoglycans, predominately heparin and heparan sulfate. To date, all chemokines tested bind to immobilized heparin in vitro, as well as cell surface heparan sulfate in vitro and in vivo. These interactions play an important role in modulating the action of chemokines by facilitating the formation of stable chemokine gradients within the vascular endothelium and directing leukocyte migration, by protecting chemokines from proteolysis, by inducing chemokine oligomerization, and by facilitating transcytosis. Despite the importance of eotaxin in eosinophil differentiation and recruitment being well established, little is known about the interaction between eotaxin and GAGs and the functional consequences of such an interaction. Here we report that eotaxin binds selectively to immobilized heparin with high affinity (K(d) = 1.23 x 10(-8) M), but not to heparan sulfate or a range of other GAGs. The interaction of eotaxin with heparin does not promote eotaxin oligomerization but protects eotaxin from proteolysis directly by plasmin and indirectly by cathepsin G and elastase. In vivo, co-administration of eotaxin and heparin is able to significantly enhance eotaxin-mediated eosinophil recruitment in a mouse air-pouch model. Furthermore, when heparin is co-administered with eotaxin at a concentration that does not normally result in eosinophil infiltration, eosinophil recruitment occurs. In contrast, heparin does not enhance eotaxin-mediated eosinophil chemotaxis in vitro, suggesting protease protection or haptotactic gradient formation as the mechanism by which heparin enhances eotaxin action in vivo. These results suggest a role for mast cell-derived heparin in the recruitment of eosinophils, reinforcing Th2 polarization of inflammatory responses. Topics: Animals; Anticoagulants; Cathepsin G; Cathepsins; Chemokine CCL11; Chemokines, CC; Chemotaxis, Leukocyte; Endothelium, Vascular; Eosinophils; Fibrinolysin; Heparin; Heparitin Sulfate; Inflammation; Male; Mast Cells; Mice; Mice, Inbred BALB C; Mice, Transgenic; Models, Biological; Protein Binding; Protein Processing, Post-Translational; Serine Endopeptidases; Th2 Cells | 2007 |
Adiponectin binds to chemokines via the globular head and modulates interactions between chemokines and heparan sulfates.
Adiponectin, a fat cell-derived protein, has been attracting considerable attention because of its antidiabetic and antiatherogenic activities. The aim of the present study is to identify molecules physiologically associating with adiponectin and to understand how the protein displays diverse biological activities.. We used an expression cloning method combined with enzyme-linked immunosorbent assay to clone adiponectin-binding proteins from the MS-5 complementary DNA library.. We successfully isolated two chemokines, stromal cell-derived factor-1 (SDF-1) and CCF18, and verified that adiponectin bound to them via its globular head. Adiponectin bound with various chemokines in vitro, such as macrophage-inflammatory protein-1alpha (MIP-1alpha), RANTES, and monocyte chemoattractant protein-1 (MCP-1), suggesting that the protein had a feature commonly to bind to the chemokine family. The middle part of chemokines, dispensable for interacting with their receptors, was found to be important for the adiponectin binding. Although the interaction of adiponectin to SDF-1 affected neither the SDF-1-CXCR4 binding nor the SDF-1 signaling in Jurkat cells, adiponectin and heparin mutually interfered in their association to SDF-1 and MCP-1 in vitro, implying that their association might influence the distribution of adiponectin and SDF-1 in inflammatory sites. Indeed, both adiponectin and SDF-1 was positively immunostained in vascular walls in guts from acute graft-vs-host disease patients. In addition, peripheral blood of adiponectin-deficient mice contained more hematopoietic progenitors than that of wild-type mice.. Adiponectin may be involved in regulation of inflammation via binding to specific chemokines. Additionally, the interaction possibly enables adiponectin to gather and play its role in inflammatory sites. Topics: Acute Disease; Adiponectin; Animals; Chemokines; Cloning, Molecular; Graft vs Host Disease; HeLa Cells; Hematopoietic Stem Cells; Heparitin Sulfate; Humans; Inflammation; Intestinal Diseases; Intestinal Mucosa; Intestines; Jurkat Cells; Mice; Mice, Knockout; Protein Binding; Signal Transduction | 2007 |
Heparan sulfate plays a central role in a dynamic in vitro model of protein-losing enteropathy.
Protein-losing enteropathy (PLE), the loss of plasma proteins through the intestine, is a symptom in ostensibly unrelated diseases. Emerging commonalities indicate that genetic insufficiencies predispose for PLE and environmental insults, e.g. viral infections and inflammation, trigger PLE onset. The specific loss of heparan sulfate (HS) from the basolateral surface of intestinal epithelial cells only during episodes of PLE suggests a possible mechanistic link. In the first tissue culture model of PLE using a monolayer of intestinal epithelial HT29 cells, we proved that HS loss directly causes protein leakage and amplifies the effects of the proinflammatory cytokine tumor necrosis factor alpha (TNFalpha). Here, we extend our in vitro model to assess the individual and combined effects of HS loss, interferon gamma (IFNgamma), TNFalpha, and increased pressure, and find that HS plays a central role in the patho-mechanisms underlying PLE. Increased pressure, mimicking venous hypertension seen in post-Fontan PLE patients, substantially increased protein leakage, but HS loss, IFNgamma, or TNFalpha alone had only minor effects. However, IFNgamma up-regulated TNFR1 expression and amplified TNFalpha-induced protein leakage. IFNgamma and TNFalpha compromised the integrity of the HT29 monolayer and made it more susceptible to increased pressure. HS loss itself compromises the integrity of the monolayer, amplifying the effects of pressure, but also amplifies the effects of both cytokines. In the absence of HS a combination of increased pressure, IFNgamma, and TNFalpha caused maximum protein leakage. Soluble heparin fully compensated for HS loss, providing a reasonable explanation for patient favorable response to heparin therapy. Topics: Albumins; Cell Line; Cytokines; Dose-Response Relationship, Drug; Epithelial Cells; Flow Cytometry; Genetic Predisposition to Disease; Heparin; Heparitin Sulfate; Humans; Hydrogen Peroxide; Inflammation; Interferon-gamma; Intestinal Mucosa; Microscopy, Confocal; Models, Statistical; Pressure; Protein Binding; Protein Conformation; Protein-Losing Enteropathies; Time Factors; Tumor Necrosis Factor-alpha; Up-Regulation | 2006 |
Endothelial heparan sulfate deficiency impairs L-selectin- and chemokine-mediated neutrophil trafficking during inflammatory responses.
Here we have studied the involvement of endothelial heparan sulfate in inflammation by inactivating the enzyme N-acetyl glucosamine N-deacetylase-N-sulfotransferase-1 in endothelial cells and leukocytes, which is required for the addition of sulfate to the heparin sulfate chains. Mutant mice developed normally but showed impaired neutrophil infiltration in various inflammation models. These effects were due to changes in heparan sulfate specifically in endothelial cells. Decreased neutrophil infiltration was partially due to altered rolling velocity correlated with weaker binding of L-selectin to endothelial cells. Chemokine transcytosis across endothelial cells and presentation on the cell surface were also reduced, resulting in decreased neutrophil firm adhesion and migration. Thus, endothelial heparan sulfate has three functions in inflammation: by acting as a ligand for L-selectin during neutrophil rolling; in chemokine transcytosis; and by binding and presenting chemokines at the lumenal surface of the endothelium. Topics: Amidohydrolases; Animals; Chemokine CXCL1; Chemokine CXCL2; Chemokines; Chemokines, CXC; Cytokines; Endothelial Cells; Heparitin Sulfate; Inflammation; L-Selectin; Leukocyte Rolling; Lung; Mice; Mice, Inbred C57BL; Mice, Knockout; Neutrophils; P-Selectin; Sulfotransferases | 2005 |
Heparan sulfate and inflammation.
Topics: Animals; Chemokines; Endothelial Cells; Endothelium, Vascular; Heparitin Sulfate; Humans; Inflammation; L-Selectin; Leukocytes; Ligands; P-Selectin | 2005 |
Enzymatic redesigning of biologically active heparan sulfate.
Heparan sulfate carries a wide range of biological activities, regulating blood coagulation, cell differentiation, and inflammatory responses. The sulfation patterns of the polysaccharide are essential for the biological activities. In this study, we report an enzymatic method for the sulfation of multimilligram amounts of heparan sulfate with specific functions using immobilized sulfotransferases combined with a 3'-phosphoadenosine 5'-phosphosulfate regeneration system. By selecting appropriate enzymatic modification steps, an inactive precursor has been converted to the heparan sulfate having three distinct biological activities, associated with binding to antithrombin, fibroblast growth factor-2, and herpes simplex virus envelope glycoprotein D. Because the recombinant sulfotransferases are expressed in bacteria, and the method uses a low cost sulfo donor, it can be readily utilized to synthesize large quantities of anticoagulant heparin drug or other biologically active heparan sulfates. Topics: Animals; Anticoagulants; Antithrombins; Carbohydrate Sequence; Cattle; CHO Cells; Chromatography, High Pressure Liquid; Cricetinae; Disaccharides; Dose-Response Relationship, Drug; Escherichia coli; Factor Xa; Fibroblast Growth Factor 2; Heparin; Heparitin Sulfate; Inflammation; Inhibitory Concentration 50; Kinetics; Magnetic Resonance Spectroscopy; Mice; Models, Chemical; Molecular Sequence Data; Phosphoadenosine Phosphosulfate; Polysaccharides; Protein Binding; Recombinant Proteins; Simplexvirus; Sulfotransferases; Surface Plasmon Resonance; Thrombin; Time Factors; Viral Envelope Proteins | 2005 |
Interstitial heparan sulfate in granulomatous inflammatory skin diseases.
Heparan sulfate (HS) is a glycosaminoglycan that is anchored to the outside of cell membranes. Under ordinary circumstances, it is not present in the interstitium, but under certain circumstances, mainly in the setting of inflammation and tissue repair, HS can be shed from the cell surface into the interstitium in a regulated fashion. Under these circumstances, interstitial HS seems to have an immunomodulatory function because of its binding of many cytokines. However, it is not known which cell types present at an inflammatory site are responsible for this shedding.. We have investigated the presence of interstitial HS by immunohistochemistry in various inflammatory skin diseases characterized by different compositions of the inflammatory infiltrate.. Strong interstitial HS immunoreactivity was present only in diseases with a predominantly histiocytic infiltrate but not in diseases with a predominantly lymphocytic or neutrophilic infiltrate.. This indicates that histiocytes have a direct or indirect role in the HS shedding process. In the well-formed granulomas of sarcoidosis, interstitial HS immunoreactivity was spatially associated with the fibrotic ring at the periphery of the granulomas, but not with the center harboring the histiocytes. This suggests that histiocytes can stimulate fibroblasts to shed HS into the interstitium. Topics: Cell Communication; Fibroblasts; Granuloma; Heparitin Sulfate; Histiocytes; Humans; Immunohistochemistry; Inflammation; Retrospective Studies; Skin Diseases | 2004 |
Histidine-rich glycoprotein binds to cell-surface heparan sulfate via its N-terminal domain following Zn2+ chelation.
Histidine-rich glycoprotein (HRG) is an alpha2-glycoprotein found in mammalian plasma at high concentrations (approximately 150 microg/ml) and is distinguished by its high content of histidine and proline. Structurally, HRG is a modular protein consisting of an N-terminal cystatin-like domain (N1N2), a central histidine-rich region (HRR) flanked by proline-rich sequences, and a C-terminal domain. HRG binds to cell surfaces and numerous ligands such as plasminogen, fibrinogen, thrombospondin, C1q, heparin, and IgG, suggesting that it may act as an adaptor protein either by targeting ligands to cell surfaces or by cross-linking soluble ligands. Despite the suggested functional importance of HRG, the cell-binding characteristics of the molecule are poorly defined. In this study, HRG was shown to bind to most cell lines in a Zn(2+)-dependent manner, but failed to interact with the Chinese hamster ovary cell line pgsA-745, which lacks cell-surface glycosaminoglycans (GAGs). Subsequent treatment of GAG-positive Chinese hamster ovary cells with mammalian heparanase or bacterial heparinase III, but not chondroitinase ABC, abolished HRG binding. Furthermore, blocking studies with various GAG species indicated that only heparin was a potent inhibitor of HRG binding. These data suggest that heparan sulfate is the predominate cell-surface ligand for HRG and that mammalian heparanase is a potential regulator of HRG binding. Using recombinant forms of full-length HRG and the N-terminal N1N2 domain, it was shown that the N1N2 domain bound specifically to immobilized heparin and cell-surface heparan sulfate. In contrast, synthetic peptides corresponding to the Zn(2+)-binding HRR of HRG did not interact with cells. Furthermore, the binding of full-length HRG, but not the N1N2 domain, was greatly potentiated by physiological concentrations of Zn2+. Based on these data, we propose that the N1N2 domain binds to cell-surface heparan sulfate and that the interaction of Zn2+ with the HRR can indirectly enhance cell-surface binding. Topics: Animals; Baculoviridae; Blotting, Western; Cell Membrane; Chelating Agents; CHO Cells; Chondroitin Sulfates; Complement C1q; COS Cells; Cricetinae; Dose-Response Relationship, Drug; Enzyme-Linked Immunosorbent Assay; Fibrinogen; Flow Cytometry; Glucuronidase; Glycosaminoglycans; Heparitin Sulfate; Histidine; Humans; Immunoglobulin G; Inflammation; Jurkat Cells; Ligands; Microscopy, Fluorescence; Neoplasm Metastasis; Peptides; Plasmids; Plasminogen; Proline; Protein Binding; Protein Structure, Tertiary; Proteins; Recombinant Proteins; Thrombospondins; Transfection; Zinc | 2004 |
Different affinities of glycosaminoglycan oligosaccharides for monomeric and dimeric interleukin-8: a model for chemokine regulation at inflammatory sites.
The binding of interleukin-8 (IL-8) to heparan sulfate (HS) proteoglycans on the surface of endothelial cells is crucial for the recruitment of neutrophils to an inflammatory site. Fluorescence anisotropy measurements yielded an IL-8 dimerization constant of 120 nM. The binding affinities, obtained by isothermal fluorescence titration, of size-defined heparin and HS oligosaccharides to the chemokine were found to depend on the oligomerization state of IL-8: high affinity was detected for monomeric and low affinity was detected for dimeric IL-8, referring to a self-regulatory mechanism for its chemoattractant effect. The highest affinity for monomeric IL-8 was detected for the HS octamer with a K(d) < 5 nM whereas the dissociation constants of dimeric IL-8 were found in the medium micromolar range. No indication for increasing affinities for monomeric IL-8 with increasing oligosaccharide chain length was found. Instead, a periodic pattern was obtained for the dissociation constants of the GAG oligosaccharides with respect to chain length, referring to optimum and least optimum chain lengths for IL-8 binding. GAG disaccharides were identified to be the minimum length for chemokine binding. Conformational changes of the dimeric chemokine, determined using CD spectroscopy, were detected only for the IL-8/HS complexes and not for heparin, pointing to an HS-induced activation of the chemokine with respect to receptor binding. Thermal unfolding of IL-8 yielded a single transition at 56 degrees C which was completely prevented by the presence of undigested HS or heparin, indicating structural stabilization, thereby prolonging the biological effect of the chemokine. Topics: Animals; Binding Sites; Cell Line; Circular Dichroism; Dimerization; Glycosaminoglycans; Heparin; Heparitin Sulfate; Hot Temperature; Humans; Inflammation; Interleukin-8; Kinetics; Models, Chemical; Oligosaccharides; Protein Binding; Protein Denaturation; Protein Folding; Protein Structure, Quaternary; Spectrometry, Fluorescence; Swine | 2002 |
Heparanase expression in invasive trophoblasts and acute vascular damage.
Heparan sulfate proteoglycans play a pivotal role in tissue function, development, inflammation, and immunity. We have identified a novel cDNA encoding human heparanase, an enzyme thought to cleave heparan sulfate in physiology and disease, and have located the HEP gene on human chromosome 4q21. Monoclonal antibodies against human heparanase located the enzyme along invasive extravillous trophoblasts of human placenta and along endothelial cells in organ xenografts targeted by hyperacute rejection, both sites of heparan sulfate digestion. Heparanase deposition was evident in arterial walls in normal tissues; however, vascular heparan sulfate cleavage was coincident with heparanase enzyme during inflammatory episodes. These findings suggest that heparanase elaboration and control of catalytic activity may contribute to the development and pathogenesis of vascular disease and suggest that heparanase intervention might be a useful therapeutic target. Topics: Amino Acid Sequence; Animals; Antibodies, Monoclonal; Cell Line; Chromosomes, Human, Pair 4; DNA, Complementary; Endothelium, Vascular; Gene Expression; Glucuronidase; Glycoside Hydrolases; Graft Rejection; Heparitin Sulfate; Humans; Inflammation; Molecular Sequence Data; Peptide Fragments; Swine; Trophoblasts; Vascular Diseases | 2000 |
Inflammatory mediators regulate cathepsin S in macrophages and microglia: A role in attenuating heparan sulfate interactions.
Cathepsin S is a member of the family of cysteine lysosomal proteases. The distribution of cathepsin S is restricted to cells from the mononuclear lineage both in the brain and in the periphery. Also, its protease activity is uniquely stable at neutral pH.. We compared the expression of cathepsin S, B, and L mRNAs in various undifferentiated and differentiated cells of mononuclear origin, and examined the modulation of these mRNAs by inflammatory mediators (lipopolysaccharide and various cytokines). In addition, the effect of these agents on cathepsin S protein levels and protease activity was also determined. Lastly, the ability of cathepsin S to process basement membrane components such as heparan sulfate proteoglycans in vitro and in vivo was assessed.. Cathepsin S, B, and L mRNAs are expressed in mature macrophages and microglial cells and not in undifferentiated monocytes. Activators of macrophages negatively regulate all three transcripts. Consistent with this, treatment with these agents leads to a decrease in intracellular cathepsin S protein levels and activity. However, the same treatments result in stimulation of secreted cathepsin S activity. Cathepsin S is capable of degrading heparan sulfate proteoglycans in vitro. Also, when expressed in endothelial cells, cathepsin S autocrinely attenuates the basic fibroblast growth factor (bFGF)-mediated binding of FGF receptor containing cells to endothelial cells, by acting on basement membrane proteoglycans.. Taken together, these data imply that cathepsin S is a regulatable cysteine protease that plays a role in the degradation of extracellular proteins, whose secretion from macrophages and microglia is increased by signals that lead to activation of these cells, and may be important in regulating extracellular matrix interactions. http://link.springer-ny. com/link/service/journals/00020/bibs/5n5p320.html Topics: Animals; Blotting, Northern; Blotting, Western; Cathepsin B; Cathepsin L; Cathepsins; Cell Adhesion; Cell Line; Cysteine Endopeptidases; Cysteine Proteinase Inhibitors; Endopeptidases; Fibroblast Growth Factor 2; Granulocyte-Macrophage Colony-Stimulating Factor; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Inflammation; Interferon-alpha; Interferon-gamma; Interleukin-1; Leucine; Lipopolysaccharides; Macrophages; Mice; Microglia; Proteoglycans; RNA, Messenger; Tumor Necrosis Factor-alpha | 1999 |
Regulation of platelet heparanase during inflammation: role of pH and proteinases.
Heparan sulfate is rapidly degraded by an endoglycosidase (heparanase) secreted by activated platelets. Since the cleavage and release of heparan sulfate would profoundly alter the local physiology of the endothelium, platelet heparanase activity should be tightly regulated. Consistent with this hypothesis, platelet heparanase was found to degrade endothelial cell heparan sulfate at pH 6.0 but not at pH 7.4, even though 25% of maximum activity was detected at pH 7.4. Loss of heparanase activity occurred rapidly (t1/2 is approximately equal to 20 min) and reversibly at physiologic pH but did not occur at acidic pH (<7.0). Inactivation of heparanase at pH 7.4 did not affect heparin binding and was reversed by 0.5 M NaCl or by heparan sulfate but not by chondroitin sulfate, suggesting inactive heparanase could be tethered on cell surfaces and the function regulated by heparan sulfate. Heparanase was gradually inactivated by trypsin and urokinase (t1/2 = 5 h) but resisted cleavage by leukocyte cathepsin G, leukocyte elastase, plasmin, and thrombin. These findings are consistent with a model in which platelet heparanase is active at the low pH of inflammation but inactive under physiologic conditions preventing inadvertent cleavage of heparan sulfate and loss of physiologic functions of endothelial cells. Topics: Animals; Blood Coagulation Factors; Blood Platelets; Endothelium; Enzyme Activation; Enzyme Stability; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Humans; Hydrogen-Ion Concentration; Inflammation; Molecular Weight; Peptides; Serine Endopeptidases; Sodium Chloride; Swine | 1998 |
Vascular heparan sulfates may limit the ability of leukocytes to penetrate the endothelial barrier--implications for use of glucosamine in inflammatory disorders.
Oral glucosamine has anti-inflammatory activity in rodents, and anecdotal evidence suggests that it may be clinically useful in inflammatory bowel disorders. A possible explanation is that supplemental glucosamine increases production of heparan sulfate (HS) proteoglycans by the vascular endothelium, thereby improving the endothelium's barrier function. Extravasation of leukocytes and metastatic cancer cells requires degradation of HS. Heparin can inhibit neutrophil activation, adhesion, and chemotaxis, and--like glucosamine--has been reported effective for managing inflammatory bowel syndromes. Cytokine-mediated loss of endothelial HS may be a key factor in the coordinated inflammatory response. These considerations suggest that glucosamine may have clinical utility in a range of inflammatory disorders, and should be assessed with regard to its impact on cancer metastasis and peripheral ischemic disease. In inflammatory bowel disease, fish oil, ginkgolides, and enteric-coated 5-aminosalicylic acid may safely complement the efficacy of glucosamine. Topics: Animals; Endothelium, Vascular; Glucosamine; Heparitin Sulfate; Humans; Inflammation; Inflammatory Bowel Diseases; Leukocytes; Models, Biological | 1998 |
Aging is associated with reduced deposition of specific extracellular matrix components, an upregulation of angiogenesis, and an altered inflammatory response in a murine incisional wound healing model.
The concept that aging impairs wound healing is largely unsubstantiated, the literature being contradictory because of poor experimental design and a failure to adequately characterize animal models. This study tested the hypothesis that aging retards the rate of wound repair using standardized cutaneous incisional wounds in a well-characterized aging mouse colony. Against the background of age-related changes in normal dermal composition, marked differences in healing were observed. Immunostaining for fibronectin was decreased in the wounds of the old mice, with a delay in the inflammatory response, re-epithelialization, and the appearance of extracellular matrix components. Heparan sulfate and blood vessel staining were both unexpectedly increased in the wounds of the old animals at late time points. Despite an overall decrease in collagen I and III deposition in the wounds of old mice, the dermal organization was surprisingly similar to that of normal dermal basket-weave collagen architecture. By contrast, young animals developed abnormal, dense scars. Intriguingly, some of these age-related changes in scar quality and inflammatory cell profile are similar to those seen in fetal wound healing. The rate of healing in young animals appears to be increased at the expense of the scar quality, perhaps resulting from an altered inflammatory response. Topics: Aging; Animals; Chondroitin Sulfates; Collagen; Elastin; Endothelium; Extracellular Matrix; Heparitin Sulfate; Immunohistochemistry; Inflammation; Male; Mice; Mice, Inbred C57BL; Neovascularization, Physiologic; Skin; Up-Regulation; Wound Healing | 1997 |
Epicutaneous application of leukotriene B4 induces patterns of tenascin and a heparan sulfate proteoglycan epitope that are typical for psoriatic lesions.
Application of leukotriene B4 (LTB4) to normal human skin induces changes similar to those found in psoriatic skin, and it has proved to be a useful model for studying the pathogenesis and treatment of psoriasis. We studied the expression patterns of molecules that have recently been shown to be altered in lesional psoriatic skin, including the extracellular matrix protein tenascin (TN) and the basement membrane and cell surface-associated heparan sulfate proteoglycans (HSPGs). During 72-h the expression of these markers was studied immunohistochemically and the expression of TN was correlated with epidermal proliferation and influx of inflammatory cells. Following the peak influx of polymorphonuclear leukocytes, a marked increase in TN expression was noted in the papillary dermis 72 h after LTB4 application. The expression patterns of basal membrane-associated epitopes of HSPG remained unaltered, whereas the expression of cell surface-associated HSPG disappeared 72 h after LTB4 application. A significant correlation was found between dermal TN expression and epidermal hyperproliferation, and between TN expression and the presence of dermal T cells. These findings indicate that the model of LTB4-induced acute cutaneous inflammation displays many characteristics of early psoriatic lesions and could serve as a model to study some of the cell biological changes in this disease. Topics: Administration, Cutaneous; Adult; Cell Division; Dermatitis, Irritant; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Immunohistochemistry; Inflammation; Leukotriene B4; Male; Models, Biological; Proteoglycans; Psoriasis; Skin; Tenascin | 1997 |
Comparative analysis of the ability of leucocytes, endothelial cells and platelets to degrade the subendothelial basement membrane: evidence for cytokine dependence and detection of a novel sulfatase.
The subendothelial basement membrane (BM) is regarded as an important barrier to the entry of leucocytes into inflammatory sites. This study compares the ability of leucocytes, platelets and endothelial cells (EC) to degrade a [35SO4]-labelled subendothelial extracellular matrix (ECM) and assesses the effect of PMA and various pro-inflammatory cytokines on this degradative activity. The different products of degradation, identified by fast protein liquid chromatography (FPLC) gel filtration chromatography, were indicative of protease, endoglycosidase (heparanase) and exoglycosidase and/or sulfatase activity. In terms of ECM degradation, EC and platelets were the most active, with PMA stimulation further enhancing the degradative activity of these two cell types. Platelets exhibited predominantly heparanase activity whereas the EC degradation products suggested a range of enzymic activities, namely proteases, heparanases and sulfatases. Interestingly, EC in suspension expressed these three enzymic activities whereas confluent EC monolayers only exhibited sulfatase activity, suggesting that the former situation might represent an angiogenic response. In the case of leucocytes, neutrophils and lymphocytes degraded the ECM to a much greater extent than monocytes. Each cell type also differed in the predominant enzymic activities it expressed, for example, heparanase activity by lymphocytes, protease activity by neutrophils and sulfatase activity by monocytes. Furthermore, PMA stimulation was shown to have differential effects on these enzymic activities. Some pro-inflammatory cytokines were found to be cell-type specific in their effects on ECM degradation. Thus, IL-1 + TNF enhanced neutrophil and EC degradation of the ECM but inhibited lymphocyte ECM degradation. In contrast, the chemokine IL-8 enhanced ECM degradation by neutrophils, lymphocytes and EC. Of particular interest was the unique sulfatase activity expressed by EC and monocytes which was induced in EC by TNF + IL-1 and IL-8, whereas in monocytes the sulfatase activity was exclusively induced by the chemokine monocyte chemotactic and activating factor (MCAF). Collectively, the results of this study show that leucocytes differ markedly in the enzymes they express to degrade the BM during extravasation and that PMA and cytokines are cell-type specific in their induction of hydrolytic enzyme activity. These results also indicate that EC may play an important role, not only in the recruitment of leuco Topics: Animals; Basement Membrane; Blood Platelets; Cattle; Cell Movement; Cells, Cultured; Chromatography, Liquid; Cytokines; Endopeptidases; Endothelium; Endothelium, Corneal; Endothelium, Vascular; Extracellular Matrix; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Inflammation; Leukocytes; Rats; Sulfatases; Tumor Cells, Cultured | 1995 |
Neurite growth modulation associated with astrocyte proteoglycans: influence of activators of inflammation.
Of the three classes of sulphated proteoglycans produced by type 1 astrocytes in vitro and released into conditioned medium, only heparan sulphate (HS) was associated with enhanced neurite growth by sensory neurons following pretreatment of a laminin substratum. Astrocyte-conditioned medium (ACM) produced in the presence of certain inflammatory mediators had reduced titers of neurite-promoting activity. The low activity ACM contained inhibitors of neurite growth. Heparan sulphate proteoglycans may modulate neurite growth when complexed to constituents of the extracellular milieu either directly or by interacting with other growth-promoting or growth-inhibitory factors. Topics: Animals; Animals, Newborn; Astrocytes; Cells, Cultured; Chick Embryo; Culture Media; Female; Growth Inhibitors; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Inflammation; Interleukin-1; Lipopolysaccharides; Macrophages; Mice; Nerve Growth Factors; Proteoglycans; Rats | 1992 |
Thrombin enhances degradation of heparan sulfate in the extracellular matrix by tumor cell heparanase.
The ability of normal and malignant blood-borne cells to extravasate correlates with the activity of an endo-beta-D-glucuronidase (heparanase) which degrades heparan sulfate (HS) in the subendothelial extracellular matrix (ECM). The association of malignancy with different types of coagulopathies prompted us to study the effect of thrombin (EC 3.4.21.5), a serine protease elaborated during activation of the clotting cascade, on the ability of heparanase to degrade the ECM-HS. The circulating zymogen form of thrombin, prothrombin, was converted to proteolytically active thrombin during incubation with ECM. Thrombin generation by the ECM was time and dose dependent, reaching maximal conversion by 6 h incubation at 3 U/ml of prothrombin. Heparanase-mediated release of low Mr HS cleavage products from sulfate-labeled ECM was stimulated four- to sixfold in the presence of alpha-thrombin, but there was no effect on degradation of soluble HS. Similar results were obtained with heparanase preparations derived from mouse lymphoma and human hepatoma cell lines and from human placenta. Incubation of ECM with alpha-thrombin alone resulted in release of nearly intact high-Mr labeled proteoglycans. Thrombin stimulation of heparanase action was dose and time dependent, reaching a maximal value at 24 h incubation with 1 microM alpha-thrombin. The effect of modified thrombin preparations correlated with their proteolytic activity. Catalytically blocked preparations of thrombin (e.g., DIP-alpha-thrombin, MeSO2-alpha-thrombin) failed to facilitate heparanase action, while catalytically modified preparations (e.g., gamma-thrombin, NO2-alpha-thrombin) exerted only a slight enhancement. Antithrombin III (ATIII) and hirudin both inhibited thrombin-stimulated heparanase degradation of ECM-bound HS. Heparanase action was also facilitated by ECM-immobilized thrombin to an extent which was similar to that induced by soluble thrombin. This result implies that thrombin sequestered by the subendothelial ECM and protected from interaction with its natural inhibitor ATIII (Bar-Shavit et al., 1989, J. Clin. Invest. 84, 1096-1104) may participate locally in cellular invasion during tumor metastasis, inflammation, and autoimmunity. Topics: Animals; Autoimmunity; Cattle; Cells, Cultured; Cornea; Dose-Response Relationship, Drug; Enzyme Activation; Epithelial Cells; Extracellular Matrix; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Inflammation; Neoplasm Metastasis; Thrombin | 1992 |
Human neutrophil elastase: degradation of basement membrane components and immunolocalization in the tissue.
Human neutrophil elastase was purified to homogeneity as two isozymes named E1 and E2. The isozymes degraded Type IV collagen, laminin, fibronectin, and heparan sulfate proteoglycan similarly to each other. The degradation of such basement membrane components by elastase may assist the extravasation of neutrophils in the process of inflammation. Among the substrates tested, only type V collagen, which is susceptible to neutrophil gelatinase, was resistant to elastase. This broad substrate specificity of the enzyme may also contribute to tissue destruction at the sites of inflammation. We produced a monoclonal antibody against the purified enzyme and applied it to immunohistochemical studies. In bronchopneumonia and polyarteritis nodosa, elastase was associated with the cleaved elastic fibers, indicating that the enzyme really destroys tissue in vivo. In the exudates of rheumatoid joint, elastase was stained as diffuse fine granules. Immunohistochemical studies with the monoclonal antibody will provide a complementary way to disclose the mechanism of diseases related to neutrophil infiltration. Topics: Antibodies, Monoclonal; Arthritis, Rheumatoid; Basement Membrane; Bronchopneumonia; Chondroitin Sulfate Proteoglycans; Collagen; Extracellular Matrix; Fibronectins; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Hydrolysis; Inflammation; Isoenzymes; Laminin; Neutrophils; Pancreatic Elastase; Polyarteritis Nodosa; Substrate Specificity | 1990 |
Inflammation-induced synthesis of proteoheparan sulfate: a novel acute-phase reactant in rat hepatocytes.
The synthesis of proteoheparan sulfate in hepatocytes is positively regulated under acute-phase conditions produced either by turpentine or deep back incision. In both cases the incorporation of [35S]sulfate and [14C]glucosamine is doubled during a 4-h incubation period if compared with control rat hepatocytes. Neither the fractional secretion rate of heparan sulfate into the medium (less than 0.1 of cell-associated glycosaminoglycans) nor the composition of newly formed proteoglycans in hepatocytes are affected during acute phase reaction. Topics: Acute-Phase Proteins; Animals; Carbon Radioisotopes; Chondroitin Sulfate Proteoglycans; Glucosamine; Glycosaminoglycans; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Inflammation; Liver; Male; Proteoglycans; Rats; Rats, Inbred Strains; Sulfur Radioisotopes; Turpentine | 1987 |