Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(2): 101507, 2022 02.
Article in English | MEDLINE | ID: mdl-34929169

ABSTRACT

Heparin, a naturally occurring glycosaminoglycan, has been found to have antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus of COVID-19. To elucidate the mechanistic basis for the antiviral activity of heparin, we investigated the binding of heparin to the SARS-CoV-2 spike glycoprotein by means of sliding window docking, molecular dynamics simulations, and biochemical assays. Our simulations show that heparin binds at long, positively charged patches on the spike glycoprotein, thereby masking basic residues of both the receptor-binding domain (RBD) and the multifunctional S1/S2 site. Biochemical experiments corroborated the simulation results, showing that heparin inhibits the furin-mediated cleavage of spike by binding to the S1/S2 site. Our simulations showed that heparin can act on the hinge region responsible for motion of the RBD between the inactive closed and active open conformations of the spike glycoprotein. In simulations of the closed spike homotrimer, heparin binds the RBD and the N-terminal domain of two adjacent spike subunits and hinders opening. In simulations of open spike conformations, heparin induces stabilization of the hinge region and a change in RBD motion. Our results indicate that heparin can inhibit SARS-CoV-2 infection by three mechanisms: by allosterically hindering binding to the host cell receptor, by directly competing with binding to host heparan sulfate proteoglycan coreceptors, and by preventing spike cleavage by furin. Furthermore, these simulations provide insights into how host heparan sulfate proteoglycans can facilitate viral infection. Our results will aid the rational optimization of heparin derivatives for SARS-CoV-2 antiviral therapy.


Subject(s)
COVID-19/metabolism , Heparin/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding Sites , Heparin/chemistry , Heparin/pharmacology , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry , COVID-19 Drug Treatment
2.
J Virol ; 90(20): 9237-50, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27489270

ABSTRACT

UNLABELLED: Human metapneumovirus (HMPV), a recently discovered paramyxovirus, infects nearly 100% of the world population and causes severe respiratory disease in infants, the elderly, and immunocompromised patients. We previously showed that HMPV binds heparan sulfate proteoglycans (HSPGs) and that HMPV binding requires only the viral fusion (F) protein. To characterize the features of this interaction critical for HMPV binding and the role of this interaction in infection in relevant models, we utilized sulfated polysaccharides, heparan sulfate mimetics, and occluding compounds. Iota-carrageenan demonstrated potent anti-HMPV activity by inhibiting binding to lung cells mediated by the F protein. Furthermore, analysis of a minilibrary of variably sulfated derivatives of Escherichia coli K5 polysaccharide mimicking the HS structure revealed that the highly O-sulfated K5 polysaccharides inhibited HMPV infection, identifying a potential feature of HS critical for HMPV binding. The peptide dendrimer SB105-A10, which binds HS, reduced binding and infection in an F-dependent manner, suggesting that occlusion of HS at the target cell surface is sufficient to prevent infection. HMPV infection was also inhibited by these compounds during apical infection of polarized airway tissues, suggesting that these interactions take place during HMPV infection in a physiologically relevant model. These results reveal key features of the interaction between HMPV and HS, supporting the hypothesis that apical HS in the airway serves as a binding factor during infection, and HS modulating compounds may serve as a platform for potential antiviral development. IMPORTANCE: Human metapneumovirus (HMPV) is a paramyxovirus that causes respiratory disease worldwide. It has been previously shown that HMPV requires binding to heparan sulfate on the surfaces of target cells for attachment and infection. In this study, we characterize the key features of this binding interaction using heparan sulfate mimetics, identify an important sulfate modification, and demonstrate that these interactions occur at the apical surface of polarized airway tissues. These findings provide insights into the initial binding step of HMPV infection that has potential for antiviral development.


Subject(s)
Antiviral Agents/pharmacology , Heparitin Sulfate/metabolism , Metapneumovirus/drug effects , Paramyxoviridae Infections/drug therapy , Respiratory System/metabolism , Respiratory System/virology , A549 Cells , Bacterial Capsules/metabolism , Cell Line , Cell Line, Tumor , Dendrimers/metabolism , Dendrimers/pharmacology , Escherichia coli/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Peptides/pharmacology , Viral Fusion Proteins/metabolism
3.
Oncotarget ; 7(23): 35353-68, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27174917

ABSTRACT

Angiogenesis plays a key role in various physiological and pathological conditions, including inflammation and tumor growth. The bone morphogenetic protein (BMP) antagonist gremlin has been identified as a novel pro-angiogenic factor. Gremlin promotes neovascular responses via a BMP-independent activation of the vascular endothelial growth factor (VEGF) receptor-2 (VEGFR2). BMP antagonists may act as covalent or non-covalent homodimers or in a monomeric form, while VEGFRs ligands are usually dimeric. However, the oligomeric state of gremlin and its role in modulating the biological activity of the protein remain to be elucidated.Here we show that gremlin is expressed in vitro and in vivo both as a monomer and as a covalently linked homodimer. Mutagenesis of amino acid residue Cys141 prevents gremlin dimerization leading to the formation of gremlinC141A monomers. GremlinC141A monomer retains a BMP antagonist activity similar to the wild-type dimer, but is devoid of a significant angiogenic capacity. Notably, we found that gremlinC141A mutant engages VEGFR2 in a non-productive manner, thus acting as receptor antagonist. Accordingly, both gremlinC141A and wild-type monomers inhibit angiogenesis driven by dimeric gremlin or VEGF-A165. Moreover, by acting as a VEGFR2 antagonist, gremlinC141A inhibits the angiogenic and tumorigenic potential of murine breast and prostate cancer cells in vivo.In conclusion, our data show that gremlin exists in multiple forms endowed with specific bioactivities and provide new insights into the molecular bases of gremlin dimerization. Furthermore, we propose gremlin monomer as a new inhibitor of VEGFR2 signalling during tumor growth.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Heterografts , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Mice , Mice, Inbred C57BL
4.
Diabetes ; 64(7): 2581-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25695948

ABSTRACT

Vascular endothelial growth factor (VEGF) blockers have been developed for the treatment of proliferative diabetic retinopathy (PDR), the leading cause of visual impairments in the working-age population in the Western world. However, limitations to anti-VEGF therapies may exist because of the local production of other proangiogenic factors that may cause resistance to anti-VEGF interventions. Thus, novel therapeutic approaches targeting additional pathways are required. Here, we identified a sulfated derivative of the Escherichia coli polysaccharide K5 [K5-N,OS(H)] as a multitarget molecule highly effective in inhibiting VEGF-driven angiogenic responses in different in vitro, ex vivo, and in vivo assays, including a murine model of oxygen-induced retinopathy. Furthermore, K5-N,OS(H) binds a variety of heparin-binding angiogenic factors upregulated in PDR vitreous humor besides VEGF, thus inhibiting their biological activity. Finally, K5-N,OS(H) hampers the angiogenic activity exerted in vitro and in vivo by human vitreous fluid samples collected from patients with PDR. Together, the data provide compelling experimental evidence that K5-N,OS(H) represents an antiangiogenic multitarget molecule with potential implications for the therapy of pathologic neovessel formation in the retina of patients with PDR.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bacterial Capsules , Diabetic Retinopathy/drug therapy , Animals , CHO Cells , Chick Embryo , Cricetulus , Heparin/metabolism , Humans , Mice , Mice, Inbred C57BL , Retinal Neovascularization/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vitreous Body/drug effects
5.
Antimicrob Agents Chemother ; 58(8): 4782-94, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24914125

ABSTRACT

Respiratory syncytial virus (RSV) exploits cell surface heparan sulfate proteoglycans (HSPGs) as attachment receptors. The interaction between RSV and HSPGs thus presents an attractive target for the development of novel inhibitors of RSV infection. In this study, selective chemical modification of the Escherichia coli K5 capsular polysaccharide was used to generate a collection of sulfated K5 derivatives with a backbone structure that mimics the heparin/heparan sulfate biosynthetic precursor. The screening of a series of N-sulfated (K5-NS), O-sulfated (K5-OS), and N,O-sulfated (K5-N,OS) derivatives with different degrees of sulfation revealed the highly sulfated K5 derivatives K5-N,OS(H) and K5-OS(H) to be inhibitors of RSV. Their 50% inhibitory concentrations were between 1.07 nM and 3.81 nM in two different cell lines, and no evidence of cytotoxicity was observed. Inhibition of RSV infection was maintained in binding and attachment assays but not in preattachment assays. Moreover, antiviral activity was also evident when the K5 derivatives were added postinfection, both in cell-to-cell spread and viral yield reduction assays. Finally, both K5-N,OS(H) and K5-OS(H) prevented RSV infection in human-derived tracheal/bronchial epithelial cells cultured to form a pseudostratified, highly differentiated model of the epithelial tissue of the human respiratory tract. Together, these features put K5-N,OS(H) and K5-OS(H) forward as attractive candidates for further development as RSV inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Bacterial Capsules/chemistry , Polysaccharides, Bacterial/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Virus Attachment/drug effects , Antiviral Agents/isolation & purification , Bronchi/drug effects , Bronchi/virology , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/virology , Escherichia coli/chemistry , Giant Cells/drug effects , Giant Cells/ultrastructure , Heparin/pharmacology , Humans , Polysaccharides, Bacterial/isolation & purification , Respiratory Syncytial Virus, Human/physiology , Tissue Culture Techniques , Trachea/drug effects , Trachea/virology , Viral Load/drug effects , Viral Plaque Assay , Virus Replication/drug effects
6.
PLoS One ; 8(8): e74035, 2013.
Article in English | MEDLINE | ID: mdl-24015314

ABSTRACT

Dengue virus (DENV) is an emerging mosquito-borne pathogen that causes cytokine-mediated alterations in the barrier function of the microvascular endothelium, leading to dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). We observed that DENV (serotype 2) productively infects primary (HMVEC-d) and immortalized (HMEC-1) human dermal microvascular endothelial cells, despite the absence of well-described DENV receptors, such as dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) or the mannose receptor on the cell surface. However, heparan sulfate proteoglycans (HSPGs) were highly expressed on these cells and pre-treatment of HMEC-1 cells with heparinase II or with glycosaminoglycans reduced DENV infectivity up to 90%, suggesting that DENV uses HSPGs as attachment receptor on microvascular endothelial cells. Sulfated Escherichia coli K5 derivatives, which are structurally similar to heparin/heparan sulfate but lack anticoagulant activity, were able to block DENV infection of HMEC-1 and HMVEC-d cells in the nanomolar range. The highly sulfated K5-OS(H) and K5-N,OS(H) inhibited virus attachment and subsequent entry into microvascular endothelial cells by interacting with the viral envelope (E) protein, as shown by surface plasmon resonance (SPR) analysis using the receptor-binding domain III of the E protein.


Subject(s)
Dengue Virus/metabolism , Escherichia coli/chemistry , Polysaccharides, Bacterial/pharmacology , Viral Envelope Proteins/metabolism , Aedes , Animals , Cell Adhesion Molecules/metabolism , Cell Line, Transformed , Dengue/metabolism , Dengue Virus/chemistry , Endothelial Cells/metabolism , Endothelial Cells/virology , Humans , Lectins, C-Type/metabolism , Polysaccharide-Lyases/chemistry , Polysaccharides, Bacterial/chemistry , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism
7.
J Biol Chem ; 288(2): 1150-61, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23166320

ABSTRACT

Once released by HIV(+) cells, p17 binds heparan sulfate proteoglycans (HSPGs) and CXCR1 on leukocytes causing their dysfunction. By exploiting an approach integrating computational modeling, site-directed mutagenesis of p17, chemical desulfation of heparin, and surface plasmon resonance, we characterized the interaction of p17 with heparin, a HSPG structural analog, and CXCR1. p17 binds to heparin with an affinity (K(d) = 190 nm) that is similar to those of other heparin-binding viral proteins. Two stretches of basic amino acids (basic motifs) are present in p17 N and C termini. Neutralization (Arg→Ala substitution) of the N-terminal, but not of the C-terminal basic motif, causes the loss of p17 heparin-binding capacity. The N-terminal heparin-binding motif of p17 partially overlaps the CXCR1-binding domain. Accordingly, its neutralization prevents also p17 binding to the chemochine receptor. Competition experiments demonstrated that free heparin and heparan sulfate (HS), but not selectively 2-O-, 6-O-, and N-O desulfated heparins, prevent p17 binding to substrate-immobilized heparin, indicating that the sulfate groups of the glycosaminoglycan mediate p17 interaction. Evaluation of the p17 antagonist activity of a panel of biotechnological heparins derived by chemical sulfation of the Escherichia coli K5 polysaccharide revealed that the highly N,O-sulfated derivative prevents the binding of p17 to both heparin and CXCR1, thus inhibiting p17-driven chemotactic migration of human monocytes with an efficiency that is higher than those of heparin and HS. Here, we characterized at a molecular level the interaction of p17 with its cellular receptors, laying the basis for the development of heparin-mimicking p17 antagonists.


Subject(s)
HIV Antigens/metabolism , Heparin/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Chemotaxis, Leukocyte , HIV Antigens/chemistry , Humans , Models, Molecular , Molecular Docking Simulation , Protein Binding , Surface Plasmon Resonance , gag Gene Products, Human Immunodeficiency Virus/chemistry
8.
Arterioscler Thromb Vasc Biol ; 31(12): e116-27, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21921258

ABSTRACT

OBJECTIVE: Heparan sulfate proteoglycans (HSPGs) modulate the interaction of proangiogenic heparin-binding vascular endothelial growth factors (VEGFs) with signaling VEGF receptor-2 (VEGFR2) and neuropilin coreceptors in endothelial cells (ECs). The bone morphogenic protein antagonist gremlin is a proangiogenic ligand of VEGFR2, distinct from canonical VEGFs. Here we investigated the role of HSPGs in VEGFR2 interaction, signaling, and proangiogenic capacity of gremlin in ECs. METHODS AND RESULTS: Surface plasmon resonance demonstrated that gremlin binds heparin and heparan sulfate, but not other glycosaminoglycans, via N-, 2-O, and 6-O-sulfated groups of the polysaccharide. Accordingly, gremlin binds HSPGs of the EC surface and extracellular matrix. Gremlin/HSPG interaction is prevented by free heparin and heparan sulfate digestion or undersulfation following EC treatment with heparinase II or sodium chlorate. However, at variance with canonical heparin-binding VEGFs, gremlin does not interact with neuropilin-1 coreceptor. On the other hand, HSPGs mediate VEGFR2 engagement and autophosphorylation, extracellular signaling-regulated kinase(1/2) and p38 mitogen-activated protein kinase activation, and consequent proangiogenic responses of ECs to gremlin. On this basis, we evaluated the gremlin-antagonist activity of a panel of chemically sulfated derivatives of the Escherichia coli K5 polysaccharide. The results demonstrate that the highly N,O-sulfated derivative K5-N,OS(H) binds gremlin with high potency, thus inhibiting VEGFR2 interaction and angiogenic activity in vitro and in vivo. CONCLUSIONS: HSPGs act as functional gremlin coreceptors in ECs, affecting its productive interaction with VEGFR2 and angiogenic activity. This has allowed the identification of the biotechnological K5-N,OS(H) as a novel angiostatic gremlin antagonist.


Subject(s)
Endothelium, Vascular/metabolism , Heparan Sulfate Proteoglycans/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor Receptor-2/agonists , Animals , Bacterial Capsules/pharmacology , Cattle , Cell Line , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Heparin/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Ligands , Models, Animal , Neovascularization, Physiologic/drug effects , Neuropilin-1/metabolism , Signal Transduction/physiology
9.
Antimicrob Agents Chemother ; 54(11): 4561-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713657

ABSTRACT

To date, there are few drugs licensed for the treatment of human cytomegalovirus (HCMV) infections, most of which target the viral DNA polymerase and suffer from many drawbacks. Thus, there is still a strong need for new anti-HCMV compounds with novel mechanisms of action. In this study, we investigated the anti-HCMV activity of chemically sulfated derivatives of Escherichia coli K5 capsular polysaccharide. These compounds are structurally related to cellular heparan sulfate and have been previously shown to be effective against some enveloped and nonenveloped viruses. We demonstrated that two derivatives, i.e., K5-N,OS(H) and K5-N,OS(L), are able to prevent cell infection by different strains of HCMV at concentrations in the nanomolar range while having no significant cytotoxicity. Studies performed to elucidate the mechanism of action of their anti-HCMV activity revealed that these compounds do not interact with either the host cell or the viral particle but need a virus-cell interaction to exert antiviral effects. Furthermore, these K5 derivatives were able to inhibit the attachment step of HCMV infection, as well as the viral cell-to-cell spread. Since the mode of inhibition of these compounds appears to differ from that of the available anti-HCMV drugs, sulfated K5 derivatives could represent the basis for the development of a novel class of potent anti-HCMV compounds. Interestingly, our studies highlight that small variations of the K5 derivatives structure can modulate the selectivity and potency of their activities against different viruses, including viruses belonging to the same family.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Bacterial Capsules/chemistry , Bacterial Capsules/pharmacology , Cytomegalovirus/drug effects , Escherichia coli/chemistry , Sulfates/chemistry , Animals , Cell Line , Humans , Mice , NIH 3T3 Cells , Virion/drug effects , Virus Replication/drug effects
10.
Pharmacol Ther ; 123(3): 310-22, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19447134

ABSTRACT

Antiviral microbicides, topical agents that prevent sexually transmitted infections, mainly work by blocking the interaction between viral proteins and cell surface components. In many instances, virus-cell interaction is mediated by cell surface heparan sulfate proteoglycans (HSPGs). HSPGs are exploited as attachment receptors by three sexually transmitted viruses: Human Immunodeficiency Virus (HIV), Herpes Simplex Virus (HSV) and Human Papilloma Virus (HPV). Since these viruses can either infect or co-infect humans, virus/HSPGs interaction is a preferential target for the development of wide-spectrum antiviral microbicides. Several polyanionic compounds prevent HIV, HSV and HPV infections in cell culture models by acting as heparan sulfate (HS)-antagonists. However, three promising polyanionic compounds recently failed to pass phase III clinical trials designed to establish their efficacy in preventing HIV acquisition. In this scenario, new polyanionic compounds must be added to the pipeline of candidate microbicides and their development as effective drugs reconsidered. The capsular K5 polysaccharide from Escherichia coli has the same structure as the heparin/HS biosynthetic precursor. Chemical and enzymatic modifications have led to the synthesis of K5 derivatives with different degrees of sulfation and charge distribution and devoid of anticoagulant activity and cell toxicity. Recently attracting attention as candidate microbicides, they potently inhibit a broad spectrum of HIV-1 strains and genital types of HPV and HSV-1 and 2 in vitro. With a focus on the K5 derivatives, this article reviews the literature on polyanions as antiviral microbicides and discusses the possible therapeutic implications of this novel class of compounds.


Subject(s)
Antiviral Agents/pharmacology , Bacterial Capsules/pharmacology , Escherichia coli/chemistry , Antiviral Agents/chemistry , Bacterial Capsules/chemistry , Clinical Trials as Topic , HIV Infections/drug therapy , HIV Infections/physiopathology , Herpes Simplex/drug therapy , Herpes Simplex/physiopathology , Humans , Papillomavirus Infections/drug therapy , Papillomavirus Infections/physiopathology
11.
Antimicrob Agents Chemother ; 52(9): 3078-84, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18573926

ABSTRACT

Herpes simplex virus type 1 (HSV-1) and HSV-2 are neurotropic viruses and common human pathogens causing major public health problems such as genital herpes, a sexually transmitted disease also correlated with increased transmission and replication of human immunodeficiency virus type 1 (HIV-1). Therefore, compounds capable of blocking HIV-1, HSV-1, and HSV-2 transmission represent candidate microbicides with a potential added value over that of molecules acting selectively against either infection. We report here that sulfated derivatives of the Escherichia coli K5 polysaccharide, structurally highly similar to heparin and previously shown to inhibit HIV-1 entry and replication in vitro, also exert suppressive activities against both HSV-1 and HSV-2 infections. In particular, the N,O-sulfated [K5-N,OS(H)] and O-sulfated epimerized [Epi-K5-OS(H)] forms inhibited the infection of Vero cells by HSV-1 and -2, with 50% inhibitory concentrations (IC(50)) between 3 +/- 0.05 and 48 +/- 27 nM, and were not toxic to the cells at concentrations as high as 5 muM. These compounds impaired the early steps of HSV-1 and HSV-2 virion attachment and entry into host cells and reduced the cell-to-cell spread of HSV-2. Since K5-N,OS(H) and Epi-K5-OS(H) also inhibit HIV-1 infection, they may represent valid candidates for development as topical microbicides preventing sexual transmission of HIV-1, HSV-1, and HSV-2.


Subject(s)
Bacterial Capsules/pharmacology , Epithelial Cells/virology , Escherichia coli/chemistry , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Sulfates/pharmacology , Animals , Bacterial Capsules/chemistry , Cell Line, Tumor , Chlorocebus aethiops , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/pathogenicity , Humans , Recombination, Genetic , Sulfates/metabolism , Vero Cells
12.
Antimicrob Agents Chemother ; 52(4): 1374-81, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18250186

ABSTRACT

Genital human papillomaviruses (HPV) represent the most common sexually transmitted agents and are classified into low or high risk by their propensity to cause genital warts or cervical cancer, respectively. Topical microbicides against HPV may be a useful adjunct to the newly licensed HPV vaccine. A main objective in the development of novel microbicides is to block HPV entry into epithelial cells through cell surface heparan sulfate proteoglycans. In this study, selective chemical modification of the Escherichia coli K5 capsular polysaccharide was integrated with innovative biochemical and biological assays to prepare a collection of sulfated K5 derivatives with a backbone structure resembling the heparin/heparan biosynthetic precursor and to test them for their anti-HPV activity. Surface plasmon resonance assays revealed that O-sulfated K5 with a high degree of sulfation [K5-OS(H)] and N,O-sulfated K5 with a high [K5-N,OS(H)] or low [K5-N,OS(L)] sulfation degree, but not unmodified K5, N-sulfated K5, and O-sulfated K5 with low levels of sulfation, prevented the interaction between HPV-16 pseudovirions and immobilized heparin. In cell-based assays, K5-OS(H), K5-N,OS(H), and K5-N,OS(L) inhibited HPV-16, HPV-18, and HPV-6 pseudovirion infection. Their 50% inhibitory concentration was between 0.1 and 0.9 mug/ml, without evidence of cytotoxicity. These findings provide insights into the design of novel, safe, and broad-spectrum microbicides against genital HPV infections.


Subject(s)
Antiviral Agents/pharmacology , Bacterial Capsules/chemistry , Bacterial Capsules/pharmacology , Escherichia coli/metabolism , Human papillomavirus 16/drug effects , Human papillomavirus 18/drug effects , Human papillomavirus 6/drug effects , Sulfates/chemistry , Antiviral Agents/chemistry , Cell Line, Tumor , Heparin/metabolism , Human papillomavirus 16/pathogenicity , Human papillomavirus 18/pathogenicity , Human papillomavirus 6/pathogenicity , Humans , Surface Plasmon Resonance , Virion/drug effects , Virion/metabolism , Virion/pathogenicity
13.
Curr Pharm Des ; 11(19): 2489-99, 2005.
Article in English | MEDLINE | ID: mdl-16026302

ABSTRACT

Heparin is a sulphated glycosaminoglycan currently used as an anticoagulant and antithrombotic drug. It consists largely of 2-O-sulphated IdoA not l&r arrow N, 6-O-disulphated GlcN disaccharide units. Other disaccharides containing unsulphated IdoA or GlcA and N-sulphated or N-acetylated GlcN are also present as minor components. This heterogeneity is more pronounced in heparan sulphate (HS), where the low-sulphated disaccharides are the most abundant. Heparin/HS bind to a variety of biologically active polypeptides, including enzymes, growth factors and cytokines, and viral proteins. This capacity can be exploited to design multi-target heparin/HS-derived drugs for pharmacological interventions in a variety of pathologic conditions besides coagulation and thrombosis, including neoplasia and viral infection. The capsular K5 polysaccharide from Escherichia coli has the same structure as the heparin precursor N-acetyl heparosan. The possibility of producing K5 polysaccharide derivatives by chemical and enzymatic modifications, thus generating heparin/HS-like compounds, has been demonstrated. These K5 polysaccharide derivatives are endowed with different biological properties, including anticoagulant/antithrombotic, antineoplastic, and anti-AIDS activities. Here, the literature data are discussed and the possible therapeutic implications for this novel class of multi-target "biotechnological heparin/HS" molecules are outlined.


Subject(s)
Heparin/biosynthesis , Heparitin Sulfate/biosynthesis , Technology, Pharmaceutical/methods , Animals , Anticoagulants/therapeutic use , Bacterial Capsules , Heparin/therapeutic use , Heparitin Sulfate/therapeutic use , Humans , Neoplasms/complications , Neoplasms/drug therapy , Polysaccharides, Bacterial/biosynthesis , Polysaccharides, Bacterial/therapeutic use , Technology, Pharmaceutical/trends , Venous Thrombosis/etiology , Venous Thrombosis/prevention & control
14.
J Med Chem ; 48(2): 349-52, 2005 Jan 27.
Article in English | MEDLINE | ID: mdl-15658847

ABSTRACT

Heparin remains a major drug in prevention of thromboembolic disease. Concerns related to its animal source have prompted search for heparin analogues. The anticoagulant activity of heparin depends on a specific pentasaccharide sequence that binds antithrombin. We report the generation of a product with antithrombin-binding, anticoagulant, and antithrombotic properties similar to those of heparin, through combined chemical and enzymatic modification of a bacterial (E. coli K5) polysaccharide. The process is readily applicable to large-scale production.


Subject(s)
Anticoagulants/chemical synthesis , Escherichia coli/chemistry , Fibrinolytic Agents/chemical synthesis , Polysaccharides, Bacterial/chemistry , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology , Antithrombins/metabolism , Bacterial Capsules , Carbohydrate Sequence , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Molecular Sequence Data , Polysaccharides, Bacterial/pharmacology , Protein Binding , Rats , Venous Thrombosis/drug therapy
15.
Arterioscler Thromb Vasc Biol ; 25(1): 71-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15514208

ABSTRACT

OBJECTIVE: Low-molecular-weight heparin (LMWH) exerts antitumor activity in clinical trials. The K5 polysaccharide from Escherichia coli has the same structure as the heparin precursor. Chemical and enzymatic modifications of K5 polysaccharide lead to the production of biotechnological heparin-like compounds. We investigated the fibroblast growth factor-2 (FGF2) antagonist and antiangiogenic activity of a series of LMW N,O-sulfated K5 derivatives. METHODS AND RESULTS: Surface plasmon resonance analysis showed that LMW-K5 derivatives bind FGF2, thus inhibiting its interaction with heparin immobilized to a BIAcore sensor chip. Interaction of FGF2 with tyrosine-kinase receptors (FGFRs), heparan sulfate proteoglycans (HSPGs), and alpha(v)beta3 integrin is required for biological response in endothelial cells. Similar to LMWH, LMW-K5 derivatives abrogate the formation of HSPG/FGF2/FGFR ternary complexes by preventing FGF2-mediated attachment of FGFR1-overexpressing cells to HSPG-bearing cells and inhibit FGF2-mediated endothelial cell proliferation. However, LMW-K5 derivatives, but not LMWH, also inhibit FGF2/alpha(v)beta3 integrin interaction and consequent FGF2-mediated endothelial cell sprouting in vitro and angiogenesis in vivo in the chick embryo chorioallantoic membrane. CONCLUSIONS: LMW N,O-sulfated K5 derivatives affect both HSPG/FGF2/FGFR and FGF2/alpha(v)beta3 interactions and are endowed with FGF2 antagonist and antiangiogenic activity. These compounds may provide the basis for the design of novel LMW heparin-like angiostatic compounds.


Subject(s)
Angiogenesis Inhibitors/biosynthesis , Escherichia coli/chemistry , Fibroblast Growth Factor 2/antagonists & inhibitors , Genetic Engineering/methods , Heparin, Low-Molecular-Weight/biosynthesis , Polysaccharides, Bacterial/biosynthesis , Angiogenesis Inhibitors/genetics , Animals , Bacterial Capsules , CHO Cells/chemistry , CHO Cells/metabolism , Cattle , Cell Adhesion/physiology , Cell Line , Cell Proliferation/drug effects , Chick Embryo , Chorioallantoic Membrane/drug effects , Cricetinae , Cricetulus , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Escherichia coli/genetics , Fibroblast Growth Factor 2/analogs & derivatives , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factors/analogs & derivatives , Fibroblast Growth Factors/metabolism , Heparan Sulfate Proteoglycans/analogs & derivatives , Heparan Sulfate Proteoglycans/deficiency , Heparan Sulfate Proteoglycans/metabolism , Heparin, Low-Molecular-Weight/chemical synthesis , Heparin, Low-Molecular-Weight/genetics , Integrin alphaVbeta3/metabolism , Mice , Neovascularization, Physiologic/drug effects , Polysaccharides, Bacterial/genetics
16.
FEBS Lett ; 568(1-3): 171-7, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15196942

ABSTRACT

The HIV-1 transactivating factor (Tat) acts as an extracellular cytokine on target cells, including endothelium. Here, we report about the Tat-antagonist capacity of chemically sulfated derivatives of the Escherichia coli K5 polysaccharide. O-sulfated K5 with high sulfation degree (K5-OS(H)) and N,O-sulfated K5 with high (K5-N,OS(H)) or low (K5-N,OS(L)) sulfation degree, but not unmodified K5, N-sulfated K5, and O-sulfated K5 with low sulfation degree, bind to Tat preventing its interaction with cell surface heparan sulfate proteoglycans, cell internalization, and consequent HIV-LTR-transactivation. Also, K5-OS(H) and K5-N,OS(H) prevent the interaction of Tat to the vascular endothelial growth factor receptor-2 on endothelial cell (EC) surface. Finally, K5-OS(H) inhibits alphav beta3 integrin/Tat interaction and EC adhesion to immobilized Tat. Consequently, K5-OS(H) and K5-N,OS(H) inhibit the angiogenic activity of Tat in vivo. In conclusion, K5 derivatives with distinct sulfation patterns bind extracellular Tat and modulate its interaction with cell surface receptors and affect its biological activities. These findings provide the basis for the design of novel extracellular Tat antagonists with possible implications in anti-AIDS therapies.


Subject(s)
Escherichia coli/chemistry , Gene Products, tat/antagonists & inhibitors , HIV-1/drug effects , Polysaccharides, Bacterial/pharmacology , Animals , Bacterial Capsules , Chick Embryo , HIV Long Terminal Repeat , Polysaccharides, Bacterial/chemistry , Sulfates/chemistry , tat Gene Products, Human Immunodeficiency Virus
17.
AIDS ; 17(2): 177-81, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12545076

ABSTRACT

OBJECTIVE: HIV-1 entry into CD4 cells represents a main target for developing novel antiretroviral agents and microbicides. DESIGN: Sulfated derivatives of the K5 polysaccharide have a backbone structure resembling the heparin precursor, but are devoid of the anticoagulant activity. The derivatives were chemically sulfated in the N position after N-deacetylation, in the O position, or in both sites. METHODS: HIV replication in human T cell blasts, monocyte-derived macrophages and cell lines was studied in the presence of sulfated K5 derivatives. RESULTS: O-sulfated [K5-OS(H)] and N,O-sulfated [K5-N,OS(H)] K5 derivatives with high degree of sulfation inhibited the replication of an HIV strain using CXCR4 as entry co-receptor (X4 virus) in both cell lines and T-cell blasts. K5 derivatives also strongly inhibited the multiplication of CCR5-dependent HIV (R5 virus) in cell lines, T-cell blasts and primary monocyte-derived macrophages. Their 50% inhibitory concentration was between 0.07 and 0.46 microM, without evidence of cytotoxicity even at the maximal concentration tested (9 microM). In addition, both K5-N,OS(H) and K5-OS(H) potently inhibited the replication of several primary HIV-1 isolates in T-cell blasts, with K5-N,OS(H) being more active than K5-OS(H) on dual tropic R5X4 strains. K5 derivatives inhibited the early steps of virion attachment and/or entry. CONCLUSIONS: Because K5 derivatives are unlikely to penetrate into cells they may represent potential topical microbicides for the prevention of sexual HIV-1 transmission.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiviral Agents/therapeutic use , HIV Infections/prevention & control , HIV-1/drug effects , Polysaccharides, Bacterial/therapeutic use , Bacterial Capsules , Cells, Cultured , Escherichia coli , Giant Cells/drug effects , HIV-1/physiology , Humans , Macrophages/virology , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Receptors, Virus/metabolism , T-Lymphocytes/virology , Tumor Cells, Cultured , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...