Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
2.
Access Microbiol ; 5(9)2023.
Article in English | MEDLINE | ID: mdl-37841103

ABSTRACT

Dormant bacterial cells do not divide and are not immediately culturable, but they persist in a state of low metabolic activity, a physiological state having clinical relevance, for instance in latent tuberculosis. Resuscitation-promoting factors (Rpfs) are proteins that act as signalling molecules mediating growth and replication. In this study we aimed to test the effect of Rpfs from Micrococcus luteus on the number and diversity of cultured bacteria using insect and soil samples, and to examine if the increase in culturability could be reproduced with the putative reaction product of Rpf, 1,6-anhydro-N-acetylmuramic acid (1,6-anhydro-MurNAc). The rpf gene from Micrococcus luteus was amplified and cloned into a pET21b expression vector and the protein was expressed in Escherichia coli BL21(DE3) cells and purified by affinity chromatography using a hexa-histidine tag. 1,6-Anhydro-MurNAc was prepared using reported chemical synthesis methods. Recombinant Rpf protein or 1,6-anhydro-MurNAc were added to R2A cultivation media, and their effect on the culturability of bacteria from eight environmental samples including four cockroach guts and four soils was examined. Colony-forming units, 16S rRNA gene copies and Illumina amplicon sequencing of the 16S rRNA gene were measured for all eight samples subjected to three different treatments: Rpf, 1,6-anhydro-MurNAc or blank control. Both Rpf and 1,6-anhydro-MurNAc increased the number of colony-forming units and of 16S rRNA gene copies across the samples although the protein was more effective. The Rpf and 1,6-anhydro-MurNAc promoted the cultivation of a diverse set of bacteria and in particular certain clades of the phyla Actinomycetota and Bacillota . This study opens the path for improved cultivation strategies aiming to isolate and study yet undescribed living bacterial organisms.

3.
Chem Commun (Camb) ; 59(54): 8384-8387, 2023 Jul 04.
Article in English | MEDLINE | ID: mdl-37318770

ABSTRACT

Interactions of lectins with glycoconjugate-terbium(III) self-assembly complexes lead to sensing through enhanced lanthanide luminescence. This glycan-directed sensing paradigm detects an unlabelled lectin (LecA) associated with pathogen P. aeruginosa in solution, without any bactericidal activity. Further development of these probes could have potential as a diagnostic tool.


Subject(s)
Bacteria , Lectins/chemistry , Luminescence , Glycoconjugates/chemistry , Glycosides/chemistry , Ligands , Bacteria/chemistry , Bacterial Proteins/chemistry , Terbium/chemistry
4.
Chem Soc Rev ; 52(11): 3663-3740, 2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37232696

ABSTRACT

Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.


Subject(s)
Carbohydrates , Lectins , Animals , Lectins/chemistry , Carbohydrates/chemistry , Mammals/metabolism
5.
EMBO Rep ; 24(4): e55971, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36856136

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative bacterium causing morbidity and mortality in immuno-compromised humans. It produces a lectin, LecB, that is considered a major virulence factor, however, its impact on the immune system remains incompletely understood. Here we show that LecB binds to endothelial cells in human skin and mice and disrupts the transendothelial passage of leukocytes in vitro. It impairs the migration of dendritic cells into the paracortex of lymph nodes leading to a reduced antigen-specific T cell response. Under the effect of the lectin, endothelial cells undergo profound cellular changes resulting in endocytosis and degradation of the junctional protein VE-cadherin, formation of an actin rim, and arrested cell motility. This likely negatively impacts the capacity of endothelial cells to respond to extracellular stimuli and to generate the intercellular gaps for allowing leukocyte diapedesis. A LecB inhibitor can restore dendritic cell migration and T cell activation, underlining the importance of LecB antagonism to reactivate the immune response against P. aeruginosa infection.


Subject(s)
Pseudomonas aeruginosa , Transendothelial and Transepithelial Migration , Humans , Animals , Mice , Endothelial Cells/metabolism , Lectins/metabolism , Lectins/pharmacology , Immunity
6.
Chembiochem ; 24(3): e202200463, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36420784

ABSTRACT

The highly glycosylated spike protein of SARS-CoV-2 is essential for infection and constitutes a prime target for antiviral agents and vaccines. The pineapple-derived jacalin-related lectin AcmJRL is present in the medication bromelain in significant quantities and has previously been described to bind mannosides. Here, we performed a large ligand screening of AcmJRL by glycan array analysis, quantified the interaction with carbohydrates and validated high-mannose glycans as preferred ligands. Because the SARS-CoV-2 spike protein was previously reported to carry a high proportion of high-mannose N-glycans, we tested the binding of AcmJRL to the recombinantly produced extraviral domain of spike protein. We could demonstrate that AcmJRL binds the spike protein with a low-micromolar KD in a carbohydrate-dependent fashion.


Subject(s)
Ananas , Lectins , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Ananas/chemistry , Carbohydrates , Lectins/chemistry , Mannose/chemistry , Polysaccharides/chemistry , Protein Binding , Spike Glycoprotein, Coronavirus/chemistry
7.
Angew Chem Int Ed Engl ; 62(7): e202215535, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36398566

ABSTRACT

Bacterial adhesion, biofilm formation and host cell invasion of the ESKAPE pathogen Pseudomonas aeruginosa require the tetravalent lectins LecA and LecB, which are therefore drug targets to fight these infections. Recently, we have reported highly potent divalent galactosides as specific LecA inhibitors. However, they suffered from very low solubility and an intrinsic chemical instability due to two acylhydrazone motifs, which precluded further biological evaluation. Here, we isosterically substituted the acylhydrazones and systematically varied linker identity and length between the two galactosides necessary for LecA binding. The optimized divalent LecA ligands showed improved stability and were up to 1000-fold more soluble. Importantly, these properties now enabled their biological characterization. The lead compound L2 potently inhibited LecA binding to lung epithelial cells, restored wound closure in a scratch assay and reduced the invasiveness of P. aeruginosa into host cells.


Subject(s)
Adhesins, Bacterial , Pseudomonas aeruginosa , Humans , Adhesins, Bacterial/chemistry , Pseudomonas aeruginosa/metabolism , Virulence Factors/metabolism , Galactosides/chemistry , Galactosides/metabolism , Galactosides/pharmacology , Bacterial Adhesion
8.
J Med Chem ; 65(20): 13988-14014, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36201248

ABSTRACT

Chronic Pseudomonas aeruginosa infections are characterized by biofilm formation, a major virulence factor of P. aeruginosa and cause of extensive drug resistance. Fluoroquinolones are effective antibiotics but are linked to severe side effects. The two extracellular P. aeruginosa-specific lectins LecA and LecB are key structural biofilm components and can be exploited for targeted drug delivery. In this work, several fluoroquinolones were conjugated to lectin probes by cleavable peptide linkers to yield lectin-targeted prodrugs. Mechanistically, these conjugates therefore remain non-toxic in the systemic distribution and will be activated to kill only once they have accumulated at the infection site. The synthesized prodrugs proved stable in the presence of host blood plasma and liver metabolism but rapidly released the antibiotic cargo in the presence of P. aeruginosa in a self-destructive manner in vitro. Furthermore, the prodrugs showed good absorption, distribution, metabolism, and elimination (ADME) properties and reduced toxicity in vitro, thus establishing the first lectin-targeted antibiotic prodrugs against P. aeruginosa.


Subject(s)
Prodrugs , Pseudomonas Infections , Humans , Pseudomonas aeruginosa , Lectins/pharmacology , Lectins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Prodrugs/pharmacology , Prodrugs/metabolism , Biofilms , Pseudomonas Infections/drug therapy , Virulence Factors/metabolism , Fluoroquinolones/pharmacology
9.
J Med Chem ; 65(20): 14180-14200, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36256875

ABSTRACT

The Gram-negative pathogen Pseudomonas aeruginosa causes severe infections mainly in immunocompromised or cystic fibrosis patients and is able to resist antimicrobial treatments. The extracellular lectin LecB plays a key role in bacterial adhesion to the host and biofilm formation. For the inhibition of LecB, we designed and synthesized a set of fucosyl amides, sulfonamides, and thiourea derivatives. Then, we analyzed their binding to LecB in competitive and direct binding assays. We identified ß-fucosyl amides as unprecedented high-affinity ligands in the two-digit nanomolar range. X-ray crystallography of one α- and one ß-anomer of N-fucosyl amides in complex with LecB revealed the interactions responsible for the high affinity of the ß-anomer at atomic level. Further, the molecules showed good stability in murine and human blood plasma and hepatic metabolism, providing a basis for future development into antibacterial drugs.


Subject(s)
Lectins , Pseudomonas aeruginosa , Humans , Mice , Animals , Pseudomonas aeruginosa/metabolism , Lectins/metabolism , Ligands , Amides/pharmacology , Amides/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Sulfonamides/metabolism , Thiourea/metabolism , Biofilms
10.
Anal Chem ; 94(16): 6112-6119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35426308

ABSTRACT

Boronic acids are widely used for labeling catechols and carbohydrates in analytical (bio)chemistry due to their high binding affinities for diols. Here, we present two asymmetrically substituted Bodipy dyes with a boronic acid at the ß-position (BBB). We present a green-emitting BBB, gBBB, and, by expanding the conjugated system of the Bodipy core at α-position, a red-emitting rBBB. Especially, gBBB shows a bathochromic shift of the electronic spectra upon binding to saccharides and polyols, whereas the fluorescence lifetime of rBBB is more sensitive to hydroxy-ligand binding. Moreover, gBBB constantly shows higher binding affinities than rBBB, reaching Kb ≈ 103 M-1 at pH 8.5 for fructose. Finally, time-resolved fluorescence anisotropy allows to distinguish the number of saccharide units of oligosaccharides as the bond along the transition dipole moment ensures that the fluorescence anisotropy only decays due to the rotational diffusion of labeled carbohydrates. ß-substituted BODIPY dyes are, thus, foreseen as fluorescence anisotropy labels for macromolecule sizing, where conventional fluorophores fail to discriminate due to the chemical similarity of recognition sites.


Subject(s)
Boronic Acids , Fluorescent Dyes , Phosphotransferases/chemistry , Boron Compounds , Boronic Acids/chemistry , Carbohydrates , Fluorescence Polarization , Fluorescent Dyes/chemistry , Phosphotransferases/analysis
11.
J Bacteriol ; 204(3): e0059721, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35129368

ABSTRACT

The Gram-negative periodontal pathogen Tannerella forsythia is inherently auxotrophic for N-acetylmuramic acid (MurNAc), which is an essential carbohydrate constituent of the peptidoglycan (PGN) of the bacterial cell wall. Thus, to build up its cell wall, T. forsythia strictly depends on the salvage of exogenous MurNAc or sources of MurNAc, such as polymeric or fragmentary PGN, derived from cohabiting bacteria within the oral microbiome. In our effort to elucidate how T. forsythia satisfies its demand for MurNAc, we recognized that the organism possesses three putative orthologs of the exo-ß-N-acetylmuramidase BsNamZ from Bacillus subtilis, which cleaves nonreducing end, terminal MurNAc entities from the artificial substrate pNP-MurNAc and the naturally-occurring disaccharide substrate MurNAc-N-acetylglucosamine (MurNAc-GlcNAc). TfNamZ1 and TfNamZ2 were successfully purified as soluble, pure recombinant His6-fusions and characterized as exo-lytic ß-N-acetylmuramidases with distinct substrate specificities. The activity of TfNamZ1 was considerably lower compared to TfNamZ2 and BsNamZ, in the cleavage of MurNAc-GlcNAc. When peptide-free PGN glycans were used as substrates, we revealed striking differences in the specificity and mode of action of these enzymes, as analyzed by mass spectrometry. TfNamZ1, but not TfNamZ2 or BsNamZ, released GlcNAc-MurNAc disaccharides from these glycans. In addition, glucosamine (GlcN)-MurNAc disaccharides were generated when partially N-deacetylated PGN glycans from B. subtilis 168 were applied. This characterizes TfNamZ1 as a unique disaccharide-forming exo-lytic ß-N-acetylmuramidase (exo-disaccharidase), and, TfNamZ2 and BsNamZ as sole MurNAc monosaccharide-lytic exo-ß-N-acetylmuramidases. IMPORTANCE Two exo-N-acetylmuramidases from T. forsythia belonging to glycosidase family GH171 (www.cazy.org) were shown to differ in their activities, thus revealing a functional diversity within this family: NamZ1 releases disaccharides (GlcNAc-MurNAc/GlcN-MurNAc) from the nonreducing ends of PGN glycans, whereas NamZ2 releases terminal MurNAc monosaccharides. This work provides a better understanding of how T. forsythia may acquire the essential growth factor MurNAc by the salvage of PGN from cohabiting bacteria in the oral microbiome, which may pave avenues for the development of anti-periodontal drugs. On a broad scale, our study indicates that the utilization of PGN as a nutrient source, involving exo-lytic N-acetylmuramidases with different modes of action, appears to be a general feature of bacteria, particularly among the phylum Bacteroidetes.


Subject(s)
Peptidoglycan , Tannerella forsythia , Acetylglucosamine/metabolism , Bacillus subtilis/metabolism , Cell Wall/metabolism , Disaccharides/metabolism , Peptidoglycan/metabolism , Substrate Specificity , Tannerella forsythia/genetics
12.
Adv Healthc Mater ; 11(11): e2102117, 2022 06.
Article in English | MEDLINE | ID: mdl-35112802

ABSTRACT

Nontuberculous mycobacterial infections rapidly emerge and demand potent medications to cope with resistance. In this context, targeted loco-regional delivery of aerosol medicines to the lungs is an advantage. However, sufficient antibiotic delivery requires engineered aerosols for optimized deposition. Here, the effect of bedaquiline-encapsulating fucosylated versus nonfucosylated liposomes on cellular uptake and delivery is investigated. Notably, this comparison includes critical parameters for pulmonary delivery, i.e., aerosol deposition and the noncellular barriers of pulmonary surfactant (PS) and mucus. Targeting increases liposomal uptake into THP-1 cells as well as peripheral blood monocyte- and lung-tissue derived macrophages. Aerosol deposition in the presence of PS, however, masks the effect of active targeting. PS alters antibiotic release that depends on the drug's hydrophobicity, while mucus reduces the mobility of nontargeted more than fucosylated liposomes. Dry-powder microparticles of spray-dried bedaquiline-loaded liposomes display a high fine particle fraction of >70%, as well as preserved liposomal integrity and targeting function. The antibiotic effect is maintained when deposited as powder aerosol on cultured Mycobacterium abscessus. When treating M. abscessus infected THP-1 cells, the fucosylated variant enabled enhanced bacterial killing, thus opening up a clear perspective for the improved treatment of nontuberculous mycobacterial infections.


Subject(s)
Anti-Bacterial Agents , Liposomes , Administration, Inhalation , Aerosols , Anti-Bacterial Agents/pharmacology , Dry Powder Inhalers , Fucose , Lung , Macrophages , Particle Size , Powders
13.
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
14.
Chembiochem ; 23(3): e202100563, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34788491

ABSTRACT

Pseudomonas aeruginosa is an opportunistic ESKAPE pathogen that produces two lectins, LecA and LecB, as part of its large arsenal of virulence factors. Both carbohydrate-binding proteins are central to the initial and later persistent infection processes, i. e. bacterial adhesion and biofilm formation. The biofilm matrix is a major resistance determinant and protects the bacteria against external threats such as the host immune system or antibiotic treatment. Therefore, the development of drugs against the P. aeruginosa biofilm is of particular interest to restore efficacy of antimicrobials. Carbohydrate-based inhibitors for LecA and LecB were previously shown to efficiently reduce biofilm formations. Here, we report a new approach for inhibiting LecA with synthetic molecules bridging the established carbohydrate-binding site and a central cavity located between two LecA protomers of the lectin tetramer. Inspired by in silico design, we synthesized various galactosidic LecA inhibitors with aromatic moieties targeting this central pocket. These compounds reached low micromolar affinities, validated in different biophysical assays. Finally, X-ray diffraction analysis revealed the interactions of this compound class with LecA. This new mode of action paves the way to a novel route towards inhibition of P. aeruginosa biofilms.


Subject(s)
Adhesins, Bacterial/metabolism , Anti-Bacterial Agents/pharmacology , Carbohydrates/pharmacology , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Carbohydrates/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Pseudomonas aeruginosa/metabolism , Structure-Activity Relationship
15.
Commun Chem ; 5(1): 64, 2022 May 20.
Article in English | MEDLINE | ID: mdl-36697615

ABSTRACT

Carbohydrate-protein interactions are key for cell-cell and host-pathogen recognition and thus, emerged as viable therapeutic targets. However, their hydrophilic nature poses major limitations to the conventional development of drug-like inhibitors. To address this shortcoming, four fragment libraries were screened to identify metal-binding pharmacophores (MBPs) as novel scaffolds for inhibition of Ca2+-dependent carbohydrate-protein interactions. Here, we show the effect of MBPs on the clinically relevant lectins DC-SIGN, Langerin, LecA and LecB. Detailed structural and biochemical investigations revealed the specificity of MBPs for different Ca2+-dependent lectins. Exploring the structure-activity relationships of several fragments uncovered the functional groups in the MBPs suitable for modification to further improve lectin binding and selectivity. Selected inhibitors bound efficiently to DC-SIGN-expressing cells. Altogether, the discovery of MBPs as a promising class of Ca2+-dependent lectin inhibitors creates a foundation for fragment-based ligand design for future drug discovery campaigns.

16.
J Am Chem Soc ; 143(45): 18977-18988, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34748320

ABSTRACT

Dendritic cells (DC) are antigen-presenting cells coordinating the interplay of the innate and the adaptive immune response. The endocytic C-type lectin receptors DC-SIGN and Langerin display expression profiles restricted to distinct DC subtypes and have emerged as prime targets for next-generation immunotherapies and anti-infectives. Using heteromultivalent liposomes copresenting mannosides bearing aromatic aglycones with natural glycan ligands, we serendipitously discovered striking cooperativity effects for DC-SIGN+ but not for Langerin+ cell lines. Mechanistic investigations combining NMR spectroscopy with molecular docking and molecular dynamics simulations led to the identification of a secondary binding pocket for the glycomimetics. This pocket, located remotely of DC-SIGN's carbohydrate bindings site, can be leveraged by heteromultivalent avidity enhancement. We further present preliminary evidence that the aglycone allosterically activates glycan recognition and thereby contributes to DC-SIGN-specific cell targeting. Our findings have important implications for both translational and basic glycoscience, showcasing heteromultivalent targeting of DCs to improve specificity and supporting potential allosteric regulation of DC-SIGN and CLRs in general.


Subject(s)
Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , Antigens, CD/metabolism , Binding Sites , Cell Adhesion Molecules/chemistry , Cell Line, Tumor , Humans , Lectins, C-Type/chemistry , Ligands , Liposomes/chemistry , Liposomes/metabolism , Mannose-Binding Lectins/metabolism , Mannosides/chemistry , Mannosides/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Receptors, Cell Surface/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
17.
ACS Chem Biol ; 16(12): 2731-2745, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34779605

ABSTRACT

Antimicrobial resistance (AMR) has been increasing unrelentingly worldwide, thus negatively impacting human health. The discovery and development of novel antibiotics is an urgent unmet need of the hour. However, it has become more challenging, requiring increasingly time-consuming efforts with increased commercial risks. Hence, alternative strategies are urgently needed to potentiate the existing antibiotics. In this context, short cationic peptides or peptide-based antimicrobials that mimic the activity of naturally occurring antimicrobial peptides (AMPs) could overcome the disadvantages of AMPs having evolved as potent antibacterial agents. Besides their potent antibacterial efficacy, short peptide conjugates have also gained attention as potent adjuvants to conventional antibiotics. Such peptide antibiotic combinations have become an increasingly cost-effective therapeutic option to tackle AMR. This Review summarizes the recent progress for peptide-based small molecules as promising antimicrobials and as adjuvants for conventional antibiotics to counter multidrug resistant (MDR) pathogens.


Subject(s)
Anti-Infective Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Biomimetic Materials/chemistry , Adjuvants, Immunologic , Amino Acid Sequence , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Biomimetic Materials/pharmacology , Cell Membrane Permeability , Drug Development , Humans , Microbial Sensitivity Tests , Molecular Conformation , Peptides, Cyclic/chemistry , Polymyxins/chemistry , Structure-Activity Relationship
18.
Nat Rev Chem ; 5(10): 726-749, 2021.
Article in English | MEDLINE | ID: mdl-34426795

ABSTRACT

An ever-increasing demand for novel antimicrobials to treat life-threatening infections caused by the global spread of multidrug-resistant bacterial pathogens stands in stark contrast to the current level of investment in their development, particularly in the fields of natural-product-derived and synthetic small molecules. New agents displaying innovative chemistry and modes of action are desperately needed worldwide to tackle the public health menace posed by antimicrobial resistance. Here, our consortium presents a strategic blueprint to substantially improve our ability to discover and develop new antibiotics. We propose both short-term and long-term solutions to overcome the most urgent limitations in the various sectors of research and funding, aiming to bridge the gap between academic, industrial and political stakeholders, and to unite interdisciplinary expertise in order to efficiently fuel the translational pipeline for the benefit of future generations.

19.
J Biol Chem ; 296: 100519, 2021.
Article in English | MEDLINE | ID: mdl-33684445

ABSTRACT

Endo-ß-N-acetylmuramidases, commonly known as lysozymes, are well-characterized antimicrobial enzymes that catalyze an endo-lytic cleavage of peptidoglycan; i.e., they hydrolyze the ß-1,4-glycosidic bonds connecting N-acetylmuramic acid (MurNAc) and N-acetylglucosamine (GlcNAc). In contrast, little is known about exo-ß-N-acetylmuramidases, which catalyze an exo-lytic cleavage of ß-1,4-MurNAc entities from the non-reducing ends of peptidoglycan chains. Such an enzyme was identified earlier in the bacterium Bacillus subtilis, but the corresponding gene has remained unknown so far. We now report that ybbC of B. subtilis, renamed namZ, encodes the reported exo-ß-N-acetylmuramidase. A ΔnamZ mutant accumulated specific cell wall fragments and showed growth defects under starvation conditions, indicating a role of NamZ in cell wall turnover and recycling. Recombinant NamZ protein specifically hydrolyzed the artificial substrate para-nitrophenyl ß-MurNAc and the peptidoglycan-derived disaccharide MurNAc-ß-1,4-GlcNAc. Together with the exo-ß-N-acetylglucosaminidase NagZ and the exo-muramoyl-l-alanine amidase AmiE, NamZ degraded intact peptidoglycan by sequential hydrolysis from the non-reducing ends. A structure model of NamZ, built on the basis of two crystal structures of putative orthologs from Bacteroides fragilis, revealed a two-domain structure including a Rossmann-fold-like domain that constitutes a unique glycosidase fold. Thus, NamZ, a member of the DUF1343 protein family of unknown function, is now classified as the founding member of a new family of glycosidases (CAZy GH171; www.cazy.org/GH171.html). NamZ-like peptidoglycan hexosaminidases are mainly present in the phylum Bacteroidetes and less frequently found in individual genomes within Firmicutes (Bacilli, Clostridia), Actinobacteria, and γ-proteobacteria.


Subject(s)
Acetylglucosamine/metabolism , Bacillus subtilis/enzymology , Glycoside Hydrolases/metabolism , Muramic Acids/metabolism , Peptidoglycan/metabolism , Crystallography, X-Ray , Glycoside Hydrolases/chemistry , Hydrolysis , Protein Conformation
20.
Angew Chem Int Ed Engl ; 60(15): 8104-8114, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33314528

ABSTRACT

Because of the antimicrobial resistance crisis, lectins are considered novel drug targets. Pseudomonas aeruginosa utilizes LecA and LecB in the infection process. Inhibition of both lectins with carbohydrate-derived molecules can reduce biofilm formation to restore antimicrobial susceptibility. Here, we focused on non-carbohydrate inhibitors for LecA to explore new avenues for lectin inhibition. From a screening cascade we obtained one experimentally confirmed hit, a catechol, belonging to the well-known PAINS compounds. Rigorous analyses validated electron-deficient catechols as millimolar LecA inhibitors. The first co-crystal structure of a non-carbohydrate inhibitor in complex with a bacterial lectin clearly demonstrates the catechol mimicking the binding of natural glycosides with LecA. Importantly, catechol 3 is the first non-carbohydrate lectin ligand that binds bacterial and mammalian calcium(II)-binding lectins, giving rise to this fundamentally new class of glycomimetics.


Subject(s)
Adhesins, Bacterial/metabolism , Anti-Bacterial Agents/pharmacology , Calcium/metabolism , Glycosides/pharmacology , Pseudomonas aeruginosa/drug effects , Adhesins, Bacterial/chemistry , Anti-Bacterial Agents/chemistry , Catechols/chemistry , Glycosides/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Pseudomonas aeruginosa/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...