Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 305
Filter
1.
J Mater Chem B ; 10(4): 537-548, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34985094

ABSTRACT

The antimicrobial resistance crisis requires novel approaches for the therapy of infections especially with Gram-negative pathogens. Pseudomonas aeruginosa is defined as priority 1 pathogen by the WHO and thus of particular interest. Its drug resistance is primarily associated with biofilm formation and essential constituents of its extracellular biofilm matrix are the two lectins, LecA and LecB. Here, we report microbial lectin-specific targeted nanovehicles based on liposomes. LecA- and LecB-targeted phospholipids were synthesized and used for the preparation of liposomes. These liposomes with varying surface ligand density were then analyzed for their competitive and direct lectin binding activity. We have further developed a microfluidic device that allowed the optical detection of the targeting process to the bacterial lectins. Our data showed that the targeted liposomes are specifically binding to their respective lectin and remain firmly attached to surfaces containing these lectins. This synthetic and biophysical study provides the basis for future application in targeted antibiotic delivery to overcome antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Lectins/antagonists & inhibitors , Liposomes/chemistry , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Lectins/metabolism , Materials Testing , Microbial Sensitivity Tests , Pseudomonas aeruginosa/metabolism
2.
Angew Chem Int Ed Engl ; 61(1): e202109339, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34713573

ABSTRACT

Carbohydrate-binding proteins (lectins) are auspicious targets in drug discovery to combat antimicrobial resistance; however, their non-carbohydrate drug-like inhibitors are still unavailable. Here, we present a druggable pocket in a ß-propeller lectin BambL from Burkholderia ambifaria as a potential target for allosteric inhibitors. This site was identified employing 19 F NMR fragment screening and a computational pocket prediction algorithm SiteMap. The structure-activity relationship study revealed the most promising fragment with a dissociation constant of 0.3±0.1 mM and a ligand efficiency of 0.3 kcal mol-1 HA-1 that affected the orthosteric site. This effect was substantiated by site-directed mutagenesis in the orthosteric and secondary pockets. Future drug-discovery campaigns that aim to develop small molecule inhibitors can benefit from allosteric sites in lectins as a new therapeutic approach against antibiotic-resistant pathogens.


Subject(s)
Lectins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Allosteric Site/drug effects , Burkholderia/chemistry , Humans , Lectins/metabolism , Molecular Structure , Small Molecule Libraries/chemistry
3.
J Chem Inf Model ; 61(12): 6085-6093, 2021 12 27.
Article in English | MEDLINE | ID: mdl-34905361

ABSTRACT

Selectins interact with cell-surface glycans to promote the initial tethering and rolling of leukocytes, and these interactions are targets for designs of inhibitors to neutralize diseases related to excessive inflammatory responses in many cardiovascular and immune dysfunctions, as well as tumor markers in different cancers. The isomeric endogenous tetrasaccharides, sialyl Lewis X (sLex) and sialyl Lewis A (sLea), are minimal sugar structures required for selectin binding. Understanding their subtle structural variances and significant advanced binding strengths of sLea over sLex could benefit the rational designs for selectin inhibitors. Modeling based on the E-selectin-sLex crystal structure in the present study demonstrated that the N-acetyl group of GlcNAc in sLex could form steric hindrances in the E-selectin-sLex complex, but the hydroxy methylene group of GlcNAc in sLea at the same position allows for stronger binding interactions. The subsequent designed inhibitor with a synthetic accessible linker molecule that has no exo-cyclic moieties replacing GlcNAc displayed comparable dynamic and energetic binding features to sLea. The present study deciphered the clues from endogenous isomeric sLea and sLex and provided insights into designing selectin inhibitors with simplified synthesis.


Subject(s)
Lectins , Oligosaccharides , Selectins , Sialyl Lewis X Antigen , Lectins/antagonists & inhibitors , Ligands , Oligosaccharides/chemistry , Sialyl Lewis X Antigen/chemistry
4.
Org Biomol Chem ; 19(14): 3234-3240, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33885578

ABSTRACT

Aspergillus fumigatus is a pathogenic fungus infecting the respiratory system and responsible for a variety of life-threatening lung diseases. A fucose-binding lectin named FleA which has a controversial role in A. fumigatus pathogenesis was recently identified. New chemical probes with high affinity and enzymatic stability are needed to explore the role of FleA in the infection process. In this study, we developed potent FleA antagonists based on optimized and non-hydrolysable thiofucoside ligands. We first synthesized a set of monovalent sugars showing micromolar affinity for FleA by isothermal titration calorimetry. The most potent derivative was co-crystallized with FleA to gain insights into the binding mode in operation. Its chemical multimerization on a cyclodextrin scaffold led to an hexavalent compound with a significantly enhanced binding affinity (Kd = 223 ± 21 nM) thanks to a chelate binding mode. The compound could probe the role of bronchial epithelial cells in a FleA-mediated response to tissue invasion.


Subject(s)
Aspergillus fumigatus/chemistry , Fucose/pharmacology , Lectins/antagonists & inhibitors , Sulfhydryl Compounds/pharmacology , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/pathogenicity , Dose-Response Relationship, Drug , Drug Design , Fucose/chemical synthesis , Fucose/chemistry , Lectins/metabolism , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Sulfhydryl Compounds/chemical synthesis , Sulfhydryl Compounds/chemistry
5.
Front Immunol ; 12: 650331, 2021.
Article in English | MEDLINE | ID: mdl-33777047

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection represents a global health crisis. Immune cell activation via pattern recognition receptors has been implicated as a driver of the hyperinflammatory response seen in COVID-19. However, our understanding of the specific immune responses to SARS-CoV-2 remains limited. Mast cells (MCs) and eosinophils are innate immune cells that play pathogenic roles in many inflammatory responses. Here we report MC-derived proteases and eosinophil-associated mediators are elevated in COVID-19 patient sera and lung tissues. Stimulation of viral-sensing toll-like receptors in vitro and administration of synthetic viral RNA in vivo induced features of hyperinflammation, including cytokine elevation, immune cell airway infiltration, and MC-protease production-effects suppressed by an anti-Siglec-8 monoclonal antibody which selectively inhibits MCs and depletes eosinophils. Similarly, anti-Siglec-8 treatment reduced disease severity and airway inflammation in a respiratory viral infection model. These results suggest that MC and eosinophil activation are associated with COVID-19 inflammation and anti-Siglec-8 antibodies are a potential therapeutic approach for attenuating excessive inflammation during viral infections.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , COVID-19/immunology , Eosinophils/immunology , Lectins/immunology , Mast Cells/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , SARS-CoV-2/immunology , Toll-Like Receptors/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , COVID-19/metabolism , COVID-19/prevention & control , COVID-19/virology , Case-Control Studies , Cytokines/metabolism , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/metabolism , Eosinophils/virology , Host-Pathogen Interactions , Humans , Lectins/antagonists & inhibitors , Lectins/genetics , Lectins/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/virology , Mice, Transgenic , Peptide Hydrolases/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Toll-Like Receptors/metabolism
6.
Leukemia ; 35(9): 2581-2591, 2021 09.
Article in English | MEDLINE | ID: mdl-33633313

ABSTRACT

The only current curative treatment for chronic lymphocytic leukemia (CLL) is allogenic hematopoietic stem cell transplantation. Chimeric antigen receptor treatment targeting CD19 for CLL achieved some complete responses, suggesting the need for alternative or combinational therapies to achieve a more robust response. In this work, we evaluated CAR-T cells specific for Siglec-6, an antigen expressed in CLL, as a novel CAR-T cell treatment for CLL. We found that detection of SIGLEC6 mRNA and Siglec-6 protein is highly restricted to placenta and immune cells in other tissues and it is not expressed in hematopoietic stem cells. We generated CAR-T cells specific for Siglec-6 based on the sequence of the fully human anti-Siglec-6 antibody (JML1), which was identified in a CLL patient that was cured after allo-hematopoietic stem cell transplantation (alloHSCT), and observed that it specifically targeted CLL cells in vitro and in a xenograft mouse model. Interestingly, a short hinge region increased the activity of CAR-T cells to target cells expressing higher Siglec-6 levels but similarly targeted CLL cells expressing lower Siglec-6 levels in vitro and in vivo. Our results identify a novel CAR-T cell therapy for CLL and establish Siglec-6 as a possible target for immunotherapy.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunotherapy, Adoptive/methods , Lectins/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, Chimeric Antigen/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Cell Proliferation , Combined Modality Therapy , Humans , Lectins/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Expert Opin Drug Discov ; 16(5): 513-536, 2021 05.
Article in English | MEDLINE | ID: mdl-33337918

ABSTRACT

Introduction: The direct binding of carbohydrates or those presented on glycoproteins or glycolipids to proteins is the primary effector of many biological responses. One class of carbohydrate-binding proteins, lectins are important in all forms of life. Their functions in animals include regulating cell adhesion, glycoprotein synthesis, metabolism, and mediating immune system response while in bacteria and viruses a lectin-mediated carbohydrate-protein interaction between host cells and the pathogen initiates pathogenesis of the infection.Areas covered: In this review, the authors outline the structural and functional pathogenesis of lectins from bacteria, amoeba, and humans. Mimics of a carbohydrate are referred to as glycomimetics, which are much smaller in molecular weight and are devised to mimic the key binding interactions of the carbohydrate while also allowing additional contacts with the lectin. This article emphasizes the various approaches used over the past 10-15 years in the rational design of glycomimetic ligands.Expert opinion: Medicinal chemistry efforts enabled by X-ray structural biology have identified small-molecule glycomimetic lectin antagonists that have entered or are nearing clinical trials. A common theme in these strategies is the use of biaryl ring systems to emulate the carbohydrate interactions with the lectin.


Subject(s)
Drug Design , Lectins/metabolism , Animals , Carbohydrate Metabolism , Chemistry, Pharmaceutical/methods , Drug Development , Humans , Lectins/antagonists & inhibitors , Ligands , Molecular Weight
8.
N Engl J Med ; 383(17): 1624-1634, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33085861

ABSTRACT

BACKGROUND: Eosinophilic gastritis and duodenitis are characterized by gastrointestinal mucosal eosinophilia, chronic symptoms, impaired quality of life, and a lack of adequate treatments. Mast-cell activity may contribute to the pathogenesis of the conditions. AK002 (lirentelimab) is an anti-Siglec-8 antibody that depletes eosinophils and inhibits mast cells and that has shown potential in animal models as a treatment for eosinophilic gastritis and duodenitis. METHODS: In this phase 2 trial, we randomly assigned adults who had symptomatic eosinophilic gastritis, eosinophilic duodenitis, or both conditions in a 1:1:1 ratio to receive four monthly infusions of low-dose AK002, high-dose AK002, or placebo. The primary end point was the change in gastrointestinal eosinophil count from baseline to 2 weeks after the final dose; to maximize statistical power, we evaluated this end point in the placebo group as compared with the combined AK002 group. Secondary end points were treatment response (>30% reduction in total symptom score and >75% reduction in gastrointestinal eosinophil count) and the change in total symptom score. RESULTS: Of the 65 patients who underwent randomization, 43 were assigned to receive AK002 and 22 were assigned to receive placebo. The mean percentage change in gastrointestinal eosinophil count was -86% in the combined AK002 group, as compared with 9% in the placebo group (least-squares mean difference, -98 percentage points; 95% confidence interval [CI], -121 to -76; P<0.001). Treatment response occurred in 63% of the patients who received AK002 and in 5% of the patients who received placebo (difference, 58 percentage points; 95% CI, 36 to 74; P<0.001). The mean change in total symptom score was -48% with AK002 and -22% with placebo (least-squares mean difference, -26 percentage points; 95% CI, -44 to -9; P = 0.004). Adverse events associated with AK002 were similar to those with placebo, with the exception of higher percentages of patients having mild-to-moderate infusion-related reactions with AK002 (60% in the combined AK002 group and 23% in the placebo group). CONCLUSIONS: In patients with eosinophilic gastritis or duodenitis, AK002 reduced gastrointestinal eosinophils and symptoms. Infusion-related reactions were more common with AK002 than with placebo. (Funded by Allakos; ENIGMA ClinicalTrials.gov number, NCT03496571.).


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Duodenitis/drug therapy , Enteritis/drug therapy , Eosinophilia/drug therapy , Eosinophils , Gastritis/drug therapy , Lectins/antagonists & inhibitors , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Duodenitis/complications , Enteritis/complications , Eosinophilia/complications , Female , Gastritis/complications , Gastrointestinal Tract/immunology , Humans , Infusions, Intravenous/adverse effects , Lectins/immunology , Leukocyte Count , Male , Middle Aged , Young Adult
9.
J Med Chem ; 63(20): 11707-11724, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32924479

ABSTRACT

Chronic infections by Pseudomonas aeruginosa are characterized by biofilm formation, which effectively enhances resistance toward antibiotics. Biofilm-specific antibiotic delivery could locally increase drug concentration to break antimicrobial resistance and reduce the drug's peripheral side effects. Two extracellular P. aeruginosa lectins, LecA and LecB, are essential structural components for biofilm formation and thus render a possible anchor for biofilm-targeted drug delivery. The standard-of-care drug ciprofloxacin suffers from severe systemic side effects and was therefore chosen for this approach. We synthesized several ciprofloxacin-carbohydrate conjugates and established a structure-activity relationship. Conjugation of ciprofloxacin to lectin probes enabled biofilm accumulation in vitro, reduced the antibiotic's cytotoxicity, but also reduced its antibiotic activity against planktonic cells due to a reduced cell permeability and on target activity. This work defines the starting point for new biofilm/lectin-targeted drugs to modulate antibiotic properties and ultimately break antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Carbohydrates/pharmacology , Ciprofloxacin/pharmacology , Lectins/antagonists & inhibitors , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemistry , Carbohydrates/chemistry , Cell Line, Tumor , Ciprofloxacin/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Lectins/metabolism , Molecular Structure , Pseudomonas aeruginosa/metabolism , Structure-Activity Relationship
10.
ChemMedChem ; 15(18): 1706-1719, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32744401

ABSTRACT

Siglecs are members of the immunoglobulin gene family containing sialic acid binding N-terminal domains. Among them, Siglec-8 is expressed on various cell types of the immune system such as eosinophils, mast cells and weakly on basophils. Cross-linking of Siglec-8 with monoclonal antibodies triggers apoptosis in eosinophils and inhibits degranulation of mast cells, making Siglec-8 a promising target for the treatment of eosinophil- and mast cell-associated diseases such as asthma. The tetrasaccharide 6'-sulfo-sialyl Lewisx has been identified as a specific Siglec-8 ligand in glycan array screening. Here, we describe an extended study enlightening the pharmacophores of 6'-sulfo-sialyl Lewisx and the successful development of a high-affinity mimetic. Retaining the neuraminic acid core, the introduction of a carbocyclic mimetic of the Gal moiety and a sulfonamide substituent in the 9-position gave a 20-fold improved binding affinity. Finally, the residence time, which usually is the Achilles tendon of carbohydrate/lectin interactions, could be improved.


Subject(s)
Lectins/antagonists & inhibitors , Oligosaccharides/pharmacology , Sialyl Lewis X Antigen/analogs & derivatives , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Humans , Lectins/metabolism , Ligands , Molecular Structure , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry , Sialyl Lewis X Antigen/chemistry , Sialyl Lewis X Antigen/pharmacology , Thermodynamics
11.
Chembiochem ; 21(23): 3433-3448, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32701213

ABSTRACT

Galacto- and fuco-clusters conjugated with one to three catechol or hydroxamate motifs were synthesised to target LecA and LecB lectins of Pseudomonas aeruginosa (PA) localised in the outer membrane and inside the bacterium. The resulting glycocluster-pseudosiderophore conjugates were evaluated as Trojan horses to cross the outer membrane of PA by iron transport. The data suggest that glycoclusters with catechol moieties are able to hijack the iron transport, whereas those with hydroxamates showed strong nonspecific interactions. Mono- and tricatechol galactoclusters (G1C and G3C) were evaluated as inhibitors of infection by PA in comparison with the free galactocluster (G0). All of them exhibited an inhibitory effect between 46 to 75 % at 100 µM, with a higher potency than G0. This result shows that LecA localised in the outer membrane of PA is involved in the infection mechanism.


Subject(s)
Adhesins, Bacterial/metabolism , Anti-Bacterial Agents/pharmacology , Lectins/antagonists & inhibitors , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Fucose/chemical synthesis , Fucose/chemistry , Fucose/pharmacology , Galactose/chemical synthesis , Galactose/chemistry , Galactose/pharmacology , Lectins/metabolism , Microbial Sensitivity Tests , Molecular Structure , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Siderophores/chemistry , Siderophores/pharmacology , Virulence
12.
Nephron ; 144(9): 453-458, 2020.
Article in English | MEDLINE | ID: mdl-32721954

ABSTRACT

IgA vasculitis can present as a glomerulonephritis histologically indistinguishable from IgA nephropathy (IgAN). In IgAN, the alternative and lectin pathways mediate glomerular injury and contribute to kidney function decline. Narsoplimab is a monoclonal antibody against mannan-binding lectin serine peptidase 2 (MASP-2), a key component of the lectin pathway. It is being evaluated in a phase III trial in IgAN (NCT03608033). Histopathological similarities with IgAN suggest lectin pathway activation also occurs in IgAV-associated nephritis (IgAVN). Here, we report the first ever case of narsoplimab use for the treatment of IgAVN.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Complement System Proteins/metabolism , Glomerulonephritis, IGA/therapy , Lectins/antagonists & inhibitors , Vasculitis/therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Female , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/physiopathology , Humans , Immunoglobulin A/blood , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Kidney Transplantation , Mannose-Binding Protein-Associated Serine Proteases/antagonists & inhibitors , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/immunology , Prednisolone/administration & dosage , Prednisolone/adverse effects , Vasculitis/immunology , Vasculitis/physiopathology , Young Adult
13.
Clin Exp Allergy ; 50(8): 904-914, 2020 08.
Article in English | MEDLINE | ID: mdl-32542913

ABSTRACT

BACKGROUND: Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is expressed on mast cells and eosinophils, but information about Siglec-8 expression and function in the lung is limited. A humanized antibody, AK002, targeting Siglec-8 is undergoing development for treatment of diseases associated with mast cell and eosinophil-driven inflammation. OBJECTIVE: To characterize Siglec-8 expression in the airway in asthma and determine whether antibodies that target Siglec-8 (S8mAbs) can decrease airway eosinophils in asthma or inhibit lung mast cell activation. METHODS: Gene expression profiling and flow cytometry were used to characterize Siglec-8 expression in sputum cells from stable asthma. An antibody-dependent cellular cytotoxicity (ADCC) assay was used to determine whether an S8mAb can decrease eosinophils in sputum from asthma patients ex vivo. A mast cell activation assay was used to determine whether an S8mAb can inhibit mast cell activation in human lung tissue ex vivo. RESULTS: Gene expression for Siglec-8 is increased in sputum cells in asthma and correlates with gene expression for eosinophils and mast cells. Gene expression for Siglec-8 is inversely and significantly correlated with measures of airflow obstruction in asthma patients. Siglec-8 is prominently expressed on the surface of eosinophils and mast cells in sputum. S8mAbs decrease eosinophils in sputum from patients with asthma and inhibit FcεR1-activated mast cells in lung tissues. CONCLUSIONS AND CLINICAL RELEVANCE: Siglec-8 is highly expressed on eosinophils and mast cells in asthmatic sputum and targeting Siglec-8 with an antibody is a plausible strategy to decrease sputum eosinophils and inhibit lung mast cells in asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Asthma/drug therapy , Eosinophils/drug effects , Lectins/antagonists & inhibitors , Lung/drug effects , Mast Cells/drug effects , Adult , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , Apoptosis/drug effects , Asthma/immunology , Asthma/metabolism , Case-Control Studies , Cells, Cultured , Eosinophils/immunology , Eosinophils/metabolism , Female , Humans , Lectins/genetics , Lectins/immunology , Lectins/metabolism , Lung/immunology , Lung/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Middle Aged , Receptors, IgE/genetics , Receptors, IgE/metabolism , Sputum/cytology , Young Adult
14.
Front Immunol ; 11: 732, 2020.
Article in English | MEDLINE | ID: mdl-32425936

ABSTRACT

It is well-known that heparin and other glycosaminoglycans (GAGs) inhibit complement activation. It is however not known whether fractionation and/or modification of GAGs might deliver pathway-specific inhibition of the complement system. Therefore, we evaluated a library of GAGs and their derivatives for their functional pathway specific complement inhibition, including the MASP-specific C4 deposition assay. Interaction of human MASP-2 with heparan sulfate/heparin was evaluated by surface plasmon resonance, ELISA and in renal tissue. In vitro pathway-specific complement assays showed that highly sulfated GAGs inhibited all three pathways of complement. Small heparin- and heparan sulfate-derived oligosaccharides were selective inhibitors of the lectin pathway (LP). These small oligosaccharides showed identical inhibition of the ficolin-3 mediated LP activation, failed to inhibit the binding of MBL to mannan, but inhibited C4 cleavage by MASPs. Hexa- and pentasulfated tetrasaccharides represent the smallest MASP inhibitors both in the functional LP assay as well in the MASP-mediated C4 assay. Surface plasmon resonance showed MASP-2 binding with heparin and heparan sulfate, revealing high Kon and Koff rates resulted in a Kd of ~2 µM and confirmed inhibition by heparin-derived tetrasaccharide. In renal tissue, MASP-2 partially colocalized with agrin and heparan sulfate, but not with activated C3, suggesting docking, storage, and potential inactivation of MASP-2 by heparan sulfate in basement membranes. Our data show that highly sulfated GAGs mediated inhibition of all three complement pathways, whereas short heparin- and heparan sulfate-derived oligosaccharides selectively blocked the lectin pathway via MASP-2 inhibition. Binding of MASP-2 to immobilized heparan sulfate/heparin and partial co-localization of agrin/heparan sulfate with MASP, but not C3b, might suggest that in vivo heparan sulfate proteoglycans act as a docking platform for MASP-2 and possibly prevent the lectin pathway from activation.


Subject(s)
Heparin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Oligosaccharides/antagonists & inhibitors , Animals , Cattle , Complement Activation/drug effects , Complement System Proteins/metabolism , Heparin/pharmacology , Heparitin Sulfate/metabolism , Humans , Intestinal Mucosa/metabolism , Kidney/metabolism , Lectins/antagonists & inhibitors , Lectins/metabolism , Lung/metabolism , Mannose-Binding Protein-Associated Serine Proteases/antagonists & inhibitors , Oligosaccharides/pharmacology , Protein Binding , Sheep , Swine , Tissue Donors
15.
Chem Biodivers ; 16(12): e1900401, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31654480

ABSTRACT

The complement system participates in host defense by eliminating microorganisms and triggering inflammation. However, insufficient control or exacerbated complement activation contributes to inflammatory diseases. Since promising antioxidant and anti-inflammatory activities have been identified in Arctium lappa L. extracts, this study aims to explore the effect of A. lappa extracts on the lectin pathway (LP) of complement activation. Four extracts were obtained by supercritical extraction using scCO2 with or without ethanol as co-solvent, at different temperatures and pressures (E1: 2.2 mg/mL, E2: 2.6 mg/mL and E3: 2.0 mg/mL, E4: 1.5 mg/mL). To evaluate the effect of A. lappa extracts on the LP activation, an ELISA assay using mannose binding lectin pathway of complement was carried out with C4 detection. All extracts showed a concentration-dependent inhibitory effect on the activation of complement by the LP. The following IC50 were observed for E1, E2, E3 and E4: 179.4 µg/mL, 74.69 µg/mL, 119.1 µg/mL and 72.19 µg/mL, respectively. Our results suggest that A. lappa extracts are potential candidates for the treatment of inflammatory disorders that are complement-related.


Subject(s)
Arctium/chemistry , Chromatography, Supercritical Fluid/methods , Complement System Proteins/metabolism , Lectins/metabolism , Plant Extracts/chemistry , Arctium/metabolism , Carbon Dioxide/chemistry , Complement System Proteins/agonists , Lectins/antagonists & inhibitors , Plant Leaves/chemistry , Plant Leaves/metabolism , Temperature
16.
Sci Rep ; 9(1): 14904, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624296

ABSTRACT

A recently described bangle lectin (PHL) from the bacterium Photorhabdus asymbiotica was identified as a mainly fucose-binding protein that could play an important role in the host-pathogen interaction and in the modulation of host immune response. Structural studies showed that PHL is a homo-dimer that contains up to seven L-fucose-specific binding sites per monomer. For these reasons, potential ligands of the PHL lectin: α-L-fucopyranosyl-containing mono-, di-, tetra-, hexa- and dodecavalent ligands were tested. Two types of polyvalent structures were investigated - calix[4]arenes and dendrimers. The shared feature of all these structures was a C-glycosidic bond instead of the more common but physiologically unstable O-glycosidic bond. The inhibition potential of the tested structures was assessed using different techniques - hemagglutination, surface plasmon resonance, isothermal titration calorimetry, and cell cross-linking. All the ligands proved to be better than free L-fucose. The most active hexavalent dendrimer exhibited affinity three orders of magnitude higher than that of standard L-fucose. To determine the binding mode of some ligands, crystal complex PHL/fucosides 2 - 4 were prepared and studied using X-ray crystallography. The electron density in complexes proved the presence of the compounds in 6 out of 7 fucose-binding sites.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Bacterial Proteins/antagonists & inhibitors , Lectins/antagonists & inhibitors , Photorhabdus/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/microbiology , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Dendrimers/chemistry , Dendrimers/pharmacology , Dendrimers/therapeutic use , Erythrocytes , Fucose/analogs & derivatives , Fucose/pharmacology , Fucose/therapeutic use , Hemagglutination/drug effects , Host-Pathogen Interactions/drug effects , Humans , Lectins/chemistry , Lectins/isolation & purification , Lectins/metabolism , Ligands , Models, Molecular , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Surface Plasmon Resonance
17.
J Med Chem ; 62(20): 9201-9216, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31553873

ABSTRACT

Biofilm formation is a key mechanism of antimicrobial resistance. We have recently reported two classes of orally bioavailable C-glycosidic inhibitors of the Pseudomonas aeruginosa lectin LecB with antibiofilm activity. They proved efficient in target binding, were metabolically stable, nontoxic, selective, and potent in inhibiting formation of bacterial biofilm. Here, we designed and synthesized six new carboxamides and 24 new sulfonamides for a detailed structure-activity relationship for two clinically representative LecB variants. Sulfonamides generally showed higher inhibition compared to carboxamides, which was rationalized based on crystal structure analyses. Substitutions at the thiophenesulfonamide increased binding through extensive contacts with a lipophilic protein patch. These metabolically stable compounds showed a further increase in potency toward the target and in biofilm inhibition assays. In general, we established the structure-activity relationship for these promising antibiofilm agents and showed that modification of the sulfonamide residue bears future optimization potential.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Glycosides/chemistry , Lectins/antagonists & inhibitors , Pseudomonas aeruginosa/physiology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Binding Sites , Cell Line , Cell Survival/drug effects , Crystallography, X-Ray , Drug Design , Humans , Lectins/metabolism , Mice , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology
18.
J Am Chem Soc ; 141(36): 14032-14037, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31460762

ABSTRACT

The Siglec family of cell surface receptors have emerged as attractive targets for cell-directed therapies due to their restricted expression on immune cells, endocytic properties, and ability to modulate receptor signaling. Human Siglec-8, for instance, has been identified as a therapeutic target for the treatment of eosinophil and mast cell disorders. A promising strategy to target Siglecs involves the use of liposomal nanoparticles with a multivalent display of Siglec ligands. A key challenge for this approach is the identification of a high affinity ligand for the target Siglec. Here, we report the development of a ligand of Siglec-8 and its closest murine functional orthologue Siglec-F that is capable of targeting liposomes to cells expressing Siglec-8 or -F. A glycan microarray library of synthetic 9-N-sulfonyl sialoside analogues was screened to identify potential lead compounds. The best ligand, 9-N-(2-naphthyl-sulfonyl)-Neu5Acα2-3-[6-O-sulfo]-Galß1-4GlcNAc (6'-O-sulfo NSANeu5Ac) combined the lead 2-naphthyl sulfonyl C-9 substituent with the preferred sulfated scaffold. The ligand 6'-O-sulfo NSANeu5Ac was conjugated to lipids for display on liposomes to evaluate targeted delivery to cells. Targeted liposomes showed strong in vitro binding/uptake and selectivity to cells expressing Siglec-8 or -F and, when administered to mice, exhibit in vivo targeting to Siglec-F+ eosinophils.


Subject(s)
Antigens, Differentiation, Myelomonocytic/metabolism , B-Lymphocytes/drug effects , Lectins/antagonists & inhibitors , Sialic Acids/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , Animals , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/metabolism , CHO Cells , Cricetulus , Humans , Lectins/metabolism , Liposomes/chemistry , Liposomes/metabolism , Mice , Molecular Conformation , Sialic Acid Binding Immunoglobulin-like Lectins , Sialic Acids/chemistry , Sulfonamides/chemistry , T-Lymphocytes/metabolism
19.
Curr Opin Chem Biol ; 53: 51-67, 2019 12.
Article in English | MEDLINE | ID: mdl-31470348

ABSTRACT

Lectins are proteins found in all domains of life with a plethora of biological functions, especially in the infection process, immune response, and inflammation. Targeting these carbohydrate-binding proteins is challenged by the fact that usually low affinity interactions between lectin and glycoconjugate are observed. Nature often circumvents this process through multivalent display of ligand and lectin. Consequently, the vast majority of synthetic antagonists are multivalently displayed native carbohydrates. At the cost of disadvantageous pharmacokinetic properties and possibly a reduced selectivity for the target lectin, the molecules usually possess very high affinities to the respective lectin through ligand epitope avidity. Recent developments include the advent of glycomimetic or allosteric small molecule inhibitors for this important protein class and their use in chemical biology and drug research. This evolution has culminated in the transition of the small molecule GMI-1070 into clinical phase III. In this opinion article, an overview of the most important developments of lectin antagonists in the last two decades with a focus on the last five years is given.


Subject(s)
Drug Discovery , Immunity , Infections/metabolism , Lectins/antagonists & inhibitors , Animals , Humans , Immunity/drug effects , Infections/drug therapy , Inflammation/drug therapy , Inflammation/metabolism
20.
Nature ; 572(7769): 392-396, 2019 08.
Article in English | MEDLINE | ID: mdl-31367043

ABSTRACT

Ovarian cancer and triple-negative breast cancer are among the most lethal diseases affecting women, with few targeted therapies and high rates of metastasis. Cancer cells are capable of evading clearance by macrophages through the overexpression of anti-phagocytic surface proteins called 'don't eat me' signals-including CD471, programmed cell death ligand 1 (PD-L1)2 and the beta-2 microglobulin subunit of the major histocompatibility class I complex (B2M)3. Monoclonal antibodies that antagonize the interaction of 'don't eat me' signals with their macrophage-expressed receptors have demonstrated therapeutic potential in several cancers4,5. However, variability in the magnitude and durability of the response to these agents has suggested the presence of additional, as yet unknown 'don't eat me' signals. Here we show that CD24 can be the dominant innate immune checkpoint in ovarian cancer and breast cancer, and is a promising target for cancer immunotherapy. We demonstrate a role for tumour-expressed CD24 in promoting immune evasion through its interaction with the inhibitory receptor sialic-acid-binding Ig-like lectin 10 (Siglec-10), which is expressed by tumour-associated macrophages. We find that many tumours overexpress CD24 and that tumour-associated macrophages express high levels of Siglec-10. Genetic ablation of either CD24 or Siglec-10, as well as blockade of the CD24-Siglec-10 interaction using monoclonal antibodies, robustly augment the phagocytosis of all CD24-expressing human tumours that we tested. Genetic ablation and therapeutic blockade of CD24 resulted in a macrophage-dependent reduction of tumour growth in vivo and an increase in survival time. These data reveal CD24 as a highly expressed, anti-phagocytic signal in several cancers and demonstrate the therapeutic potential for CD24 blockade in cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , CD24 Antigen/antagonists & inhibitors , Immunotherapy/methods , Lectins/metabolism , Macrophages/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , CD24 Antigen/deficiency , CD24 Antigen/genetics , CD24 Antigen/immunology , Cell Line, Tumor , Humans , Lectins/antagonists & inhibitors , Lectins/genetics , Macrophages/drug effects , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis/drug effects , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Survival Analysis , Tumor Escape/drug effects , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...