Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.019
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 584(7821): 479-483, 2020 08.
Article in English | MEDLINE | ID: mdl-32788728

ABSTRACT

Lipopolysaccharide (LPS) resides in the outer membrane of Gram-negative bacteria where it is responsible for barrier function1,2. LPS can cause death as a result of septic shock, and its lipid A core is the target of polymyxin antibiotics3,4. Despite the clinical importance of polymyxins and the emergence of multidrug resistant strains5, our understanding of the bacterial factors that regulate LPS biogenesis is incomplete. Here we characterize the inner membrane protein PbgA and report that its depletion attenuates the virulence of Escherichia coli by reducing levels of LPS and outer membrane integrity. In contrast to previous claims that PbgA functions as a cardiolipin transporter6-9, our structural analyses and physiological studies identify a lipid A-binding motif along the periplasmic leaflet of the inner membrane. Synthetic PbgA-derived peptides selectively bind to LPS in vitro and inhibit the growth of diverse Gram-negative bacteria, including polymyxin-resistant strains. Proteomic, genetic and pharmacological experiments uncover a model in which direct periplasmic sensing of LPS by PbgA coordinates the biosynthesis of lipid A by regulating the stability of LpxC, a key cytoplasmic biosynthetic enzyme10-12. In summary, we find that PbgA has an unexpected but essential role in the regulation of LPS biogenesis, presents a new structural basis for the selective recognition of lipids, and provides opportunities for future antibiotic discovery.


Subject(s)
Cell Membrane/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Escherichia coli/pathogenicity , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amino Acid Motifs , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/metabolism , Binding Sites , Cell Membrane/metabolism , Enzyme Stability , Escherichia coli/cytology , Escherichia coli/drug effects , Genes, Essential , Hydrolases/chemistry , Hydrolases/metabolism , Lipid A/chemistry , Lipid A/metabolism , Lipopolysaccharides/biosynthesis , Microbial Sensitivity Tests , Microbial Viability/drug effects , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Periplasm/chemistry , Periplasm/metabolism , Protein Binding , Virulence
2.
EMBO J ; 39(13): e104926, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32510692

ABSTRACT

In the outer membrane of gram-negative bacteria, O-antigen segments of lipopolysaccharide (LPS) form a chemomechanical barrier, whereas lipid A moieties anchor LPS molecules. Upon infection, human guanylate binding protein-1 (hGBP1) colocalizes with intracellular gram-negative bacterial pathogens, facilitates bacterial killing, promotes activation of the lipid A sensor caspase-4, and blocks actin-driven dissemination of the enteric pathogen Shigella. The underlying molecular mechanism for hGBP1's diverse antimicrobial functions is unknown. Here, we demonstrate that hGBP1 binds directly to LPS and induces "detergent-like" LPS clustering through protein polymerization. Binding of polymerizing hGBP1 to the bacterial surface disrupts the O-antigen barrier, thereby unmasking lipid A, eliciting caspase-4 recruitment, enhancing antibacterial activity of polymyxin B, and blocking the function of the Shigella outer membrane actin motility factor IcsA. These findings characterize hGBP1 as an LPS-binding surfactant that destabilizes the rigidity of the outer membrane to exert pleiotropic effects on the functionality of gram-negative bacterial cell envelopes.


Subject(s)
GTP-Binding Proteins/chemistry , Lipid A/chemistry , O Antigens/chemistry , Shigella/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Humans , Lipid A/metabolism , O Antigens/metabolism , Protein Binding , Shigella/metabolism
3.
Chemistry ; 30(32): e202400429, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38587187

ABSTRACT

Agonists of Toll like receptors (TLRs) have attracted interest as adjuvants and immune modulators. A crystal structure of TLR4/MD2 with E. coli LPS indicates that the fatty acid at C-2 of the lipid A component of LPS induces dimerization of two TLR4-MD2 complexes, which in turn initiates cell signaling leading to the production of (pro)inflammatory cytokines. To probe the importance of the (R)-3-hydroxymyristate at C-2 of lipid A, a range of bis- and mono-phosphoryl lipid A derivatives with different modifications at C-2 were prepared by a strategy in which 2-methylnaphthyl ethers were employed as permanent protecting group that could be readily removed by catalytic hydrogenation. The C-2 amine was protected as 9-fluorenylmethyloxycarbamate, which at a later stage could be removed to give a free amine that was modified by different fatty acids. LPS and the synthetic lipid As induced the same cytokines, however, large differences in activity were observed. A compound having a hexanoyl moiety at C-2 still showed agonistic properties, but further shortening to a butanoyl abolished activity. The modifications had a larger influence on monophosphoryl lipid As. The lipid As having a butanoyl moiety at C-2 could selectively antagonize TRIF associated cytokines induced by LPS or lipid A.


Subject(s)
Cytokines , Lipid A , Lipopolysaccharides , Lipid A/chemistry , Lipid A/pharmacology , Lipid A/analogs & derivatives , Lipid A/chemical synthesis , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Humans , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/chemistry , Drug Design , Structure-Activity Relationship , Signal Transduction/drug effects
4.
FASEB J ; 37(5): e22928, 2023 05.
Article in English | MEDLINE | ID: mdl-37071453

ABSTRACT

Colistin (polymyxin E) is a group of cationic antimicrobial cyclic peptides and is recognized as a last-resort defense against lethal infections with carbapenem-resistant pathogens. In addition to the plasmid-borne mobilized phosphoethanolamine (PEA) transferases, the functional expression of lipid A-modifying enzymes encoded on chromosomes has been attributed to intrinsic bacterial colistin resistance. However, the mechanisms of colistin resistance in Riemerella anatipestifer remain unknown. Herein, the GE296_RS09715 gene-encoded Lipid A PEA transferases (RaEptA) was identified in R. anatipestifer. Genetic and structural analyses revealed that the amino acid sequence of RaEptA shared 26.6%-33.1% similarities with the family of Lipid A PEA transferases (EptA) and MCR-like proteins and have defined 12 residues that contribute to the formation of phosphatidylethanolamine (PE)-recognizable cavities. Comparative analyses of colistin resistance in RA-LZ01 and RA-LZ01ΔRaEptA showed the level of colistin has fallen from 96 µg mL-1 down to 24 ~ 32 µg mL-1 . Site-directed mutagenesis assay of the PE-binding cavity and expression of the mutants reveals that K309-rRaEptA can remodel the surface of Escherichia coli and rendering it resistant to colistin, suggesting this point-mutation of P309K is necessary for EptA-mediated lipid A modification. Moreover, the virulence of RA-LZ01ΔRaEptA was attenuated compared with RA-LZ01 both in vivo and vitro. Taken together, the results represent the RaEptA involved in the colistin resistance and pathogenicity, and the P309K mutation might alter bacterial adaptation and increase the spread of colistin resistance from R. anatipestifer to other gram-negative bacteria. The findings of this study suggest another scenario for the spread of colistin resistance genes and should be considered by a wide audience.


Subject(s)
Anti-Bacterial Agents , Colistin , Colistin/pharmacology , Colistin/chemistry , Anti-Bacterial Agents/pharmacology , Virulence/genetics , Lipid A/chemistry , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Phenotype , Transferases
5.
Glycoconj J ; 41(2): 119-131, 2024 04.
Article in English | MEDLINE | ID: mdl-38642279

ABSTRACT

Gram-negative bacteria living in marine waters have evolved peculiar adaptation strategies to deal with the numerous stress conditions that characterize aquatic environments. Among the multiple mechanisms for efficient adaptation, these bacteria typically exhibit chemical modifications in the structure of the lipopolysaccharide (LPS), which is a fundamental component of their outer membrane. In particular, the glycolipid anchor to the membrane of marine bacteria LPSs, i.e. the lipid A, frequently shows unusual chemical structures, which are reflected in equally singular immunological properties with potential applications as immune adjuvants or anti-sepsis drugs. In this work, we determined the chemical structure of the lipid A from Cellulophaga pacifica KMM 3664T isolated from the Sea of Japan. This bacterium showed to produce a heterogeneous mixture of lipid A molecules that mainly display five acyl chains and carry a single phosphate and a D-mannose disaccharide on the glucosamine backbone. Furthermore, we proved that C. pacifica KMM 3664T LPS acts as a weaker activator of Toll-like receptor 4 (TLR4) compared to the prototypical enterobacterial Salmonella typhimurium LPS. Our results are relevant to the future development of novel vaccine adjuvants and immunomodulators inspired by marine LPS chemistry.


Subject(s)
Lipid A , Lipid A/chemistry , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/chemistry , Animals , Lipopolysaccharides/chemistry , Mice
6.
J Nanobiotechnology ; 22(1): 483, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138475

ABSTRACT

The mortality of ovarian cancer (OC) has long been the highest among gynecological malignancies. Although OC is considered to be an immunogenic tumor, the effect of immunotherapy is not satisfactory. The immunosuppressive microenvironment is one reason for this, and the absence of recognized effective antigens for vaccines is another. Chemotherapy, as one of the most commonly used treatment for OC, can produce chemotherapy-associated antigens (CAAs) during treatment and show the effect of in situ vaccine. Herein, we designed an antigen capture nano-vaccine NP-TP1@M-M with tumor targeting peptide TMTP1 and dendritic cell (DC) receptor mannose assembled on the surface and adjuvant monophosphoryl lipid A (MPLA) encapsulated in the core of poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles. PLGA itself possessed the ability of antigen capture. TMTP1 was a tumor-homing peptide screened by our research team, which held extensive and excellent tumor targeting ability. After these modifications, NP-TP1@M-M could capture and enrich more tumor-specific antigens after chemotherapy, stimulate DC maturation, activate the adaptive immunity and combined with immune checkpoint blockade to maximize the release of the body's immune potential, providing an eutherapeutic strategy for the treatment of OC.


Subject(s)
Antigens, Neoplasm , B7-H1 Antigen , Cancer Vaccines , Nanoparticles , Ovarian Neoplasms , Female , Ovarian Neoplasms/drug therapy , Animals , Mice , Cancer Vaccines/therapeutic use , Nanoparticles/chemistry , Cell Line, Tumor , Antigens, Neoplasm/immunology , Humans , Dendritic Cells/drug effects , Peptides/chemistry , Peptides/pharmacology , Lipid A/analogs & derivatives , Lipid A/chemistry , Lipid A/pharmacology , Immunotherapy/methods , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Immune Checkpoint Inhibitors/pharmacology , Nanovaccines
7.
Int J Mol Sci ; 25(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474006

ABSTRACT

The lipopolysaccharide (LPS) that resides on the outermost surface and protects Gram-negative bacteria from host defenses is one of the key components leading to Salmonella infection, particularly the endotoxic lipid A domain of LPS. Lipid A modifications have been associated with several genes such as the arnT that encodes 4-amino-4-deoxy-L-arabinose transferase, which can be critical for bacteria to resist cationic antimicrobial peptides and interfere with host immune recognition. However, the association of arnT with virulence is not completely understood. Thus, this study aimed to elucidate the interrelationship of the major lipid A modification gene arnT with Salmonella Typhimurium virulence. We observed that the arnT-deficient S. Typhimurium (JOL2943), compared to the wild type (JOL401), displayed a significant decrease in several virulence phenotypes such as polymyxin B resistance, intracellular survival, swarming, and biofilm and extracellular polymeric substance (EPS) production. Interestingly, the cell-surface hydrophobicity, adhesion, and invasion characteristics remained unaffected. Additionally, LPS isolated from the mutant induced notably lower levels of endotoxicity-related cytokines in RAW and Hela cells and mice, particularly IL-1ß with a nine-fold decrease, than WT. In terms of in vivo colonization, JOL2943 showed diminished presence in internal organs such as the spleen and liver by more than 60%, while ileal infectivity remained similar to JOL401. Overall, the arnT deletion rendered the strain less virulent, with low endotoxicity, maintained gut infectivity, and reduced colonization in internal organs. With these ideal characteristics, it can be further explored as a potential attenuated Salmonella strain for therapeutics or vaccine delivery systems.


Subject(s)
Lipid A , Salmonella typhimurium , Humans , Animals , Mice , Salmonella typhimurium/genetics , Lipid A/chemistry , Lipopolysaccharides/chemistry , Virulence , Extracellular Polymeric Substance Matrix , HeLa Cells , Bacterial Proteins/genetics
8.
Int J Mol Sci ; 25(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38928052

ABSTRACT

Bacterial endotoxins (lipopolysaccharides (LPSs)) are important mediators of inflammatory processes induced by Gram-negative microorganisms. LPSs are the key inducers of septic shock due to a Gram-negative bacterial infection; thus, the structure and functions of LPSs are of specific interest. Often, highly purified bacterial endotoxins must be isolated from small amounts of biological material. Each of the currently available methods for LPS extraction has certain limitations. Herein, we describe a rapid and simple microscale method for extracting LPSs. The method consists of the following steps: ultrasonic destruction of the bacterial material, LPS extraction via heating, LPS purification with organic solvents, and treatment with proteinase K. LPSs that were extracted by using this method contained less than 2-3% protein and 1% total nucleic acid. We also demonstrated the structural integrity of the O-antigen and lipid A via the sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) methods, respectively. We demonstrated the ability of the extracted LPSs to induce typical secretion of cytokines and chemokines by primary macrophages. Overall, this method may be used to isolate purified LPSs with preserved structures of both the O-antigen and lipid A and unchanged functional activity from small amounts of bacterial biomass.


Subject(s)
Lipopolysaccharides , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Mice , Macrophages/metabolism , Lipid A/chemistry , Lipid A/isolation & purification , Cytokines/metabolism , Endopeptidase K/metabolism , Endopeptidase K/chemistry , Electrophoresis, Polyacrylamide Gel/methods
9.
Angew Chem Int Ed Engl ; 63(24): e202402922, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38581637

ABSTRACT

Lipopolysaccharide (LPS), a cell surface component of Gram-negative bacteria, activates innate immunity. Its active principle is the terminal glycolipid lipid A. Acetobacter pasteurianus is a Gram-negative bacterium used in the fermentation of traditional Japanese black rice vinegar (kurozu). In this study, we focused on A. pasteurianus lipid A, which is a potential immunostimulatory component of kurozu. The active principle structure of A. pasteurianus lipid A has not yet been identified. Herein, we first systematically synthesized three types of A. pasteurianus lipid As containing a common and unique tetrasaccharide backbone. We developed an efficient method for constructing the 2-trehalosamine skeleton utilizing borinic acid-catalyzed glycosylation to afford 1,1'-α,α-glycoside in high yield and stereoselectivity. A common tetrasaccharide intermediate with an orthogonal protecting group pattern was constructed via [2+2] glycosylation. After introducing various fatty acids, all protecting groups were removed to achieve the first chemical synthesis of three distinct types of A. pasteurianus lipid As. After evaluating their immunological function using both human and murine cell lines, we identified the active principles of A. pasteurianus LPS. We also found the unique anomeric structure of A. pasteurianus lipid A contributes to its high chemical stability.


Subject(s)
Acetobacter , Lipid A , Lipid A/chemistry , Lipid A/immunology , Lipid A/chemical synthesis , Humans , Mice , Acetobacter/chemistry , Animals , Oligosaccharides/chemistry , Oligosaccharides/chemical synthesis , Glycosylation
10.
J Bacteriol ; 205(5): e0006723, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37070977

ABSTRACT

Gram-negative bacteria have a unique cell surface that can be modified to maintain bacterial fitness in diverse environments. A well-defined example is the modification of the lipid A component of lipopolysaccharide (LPS), which promotes resistance to polymyxin antibiotics and antimicrobial peptides. In many organisms, such modifications include the addition of the amine-containing constituents 4-amino-4-deoxy-l-arabinose (l-Ara4N) and phosphoethanolamine (pEtN). Addition of pEtN is catalyzed by EptA, which uses phosphatidylethanolamine (PE) as its substrate donor, resulting in production of diacylglycerol (DAG). DAG is then quickly recycled into glycerophospholipid (GPL) synthesis by the DAG kinase A (DgkA) to produce phosphatidic acid, the major GPL precursor. Previously, we hypothesized that loss of DgkA recycling would be detrimental to the cell when LPS is heavily modified. Instead, we found that DAG accumulation inhibits EptA activity, preventing further degradation of PE, the predominant GPL of the cell. However, DAG inhibition of pEtN addition results in complete loss of polymyxin resistance. Here, we selected for suppressors to find a mechanism of resistance independent of DAG recycling or pEtN modification. Disrupting the gene encoding the adenylate cyclase, cyaA, fully restored antibiotic resistance without restoring DAG recycling or pEtN modification. Supporting this, disruptions of genes that reduce CyaA-derived cAMP formation (e.g., ptsI) or disruption of the cAMP receptor protein, Crp, also restored resistance. We found that loss of the cAMP-CRP regulatory complex was necessary for suppression and that resistance arises from a substantial increase in l-Ara4N-modified LPS, bypassing the need for pEtN modification. IMPORTANCE Gram-negative bacteria can alter the structure of their LPS to promote resistance to cationic antimicrobial peptides, including polymyxin antibiotics. Polymyxins are considered last-resort antibiotics for treatment against multidrug-resistant Gram-negative organisms. Here, we explore how changes in general metabolism and carbon catabolite repression pathways can alter LPS structure and influence polymyxin resistance.


Subject(s)
Lipopolysaccharides , Polymyxin B , Polymyxin B/pharmacology , Lipopolysaccharides/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Cyclic AMP Receptor Protein/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Polymyxins/pharmacology , Lipid A/chemistry , Drug Resistance, Bacterial/genetics
11.
Chembiochem ; 24(10): e202300183, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37042436

ABSTRACT

Marine bacteria, which are often described as chemical gold, are considered an exceptional source of new therapeutics. Considerable research interest has been given to lipopolysaccharides (LPSs), the main components of the Gram-negative outer membrane. LPS and its lipid A portion from marine bacteria are known to exhibit a tricky chemistry that has been often associated with intriguing properties such as behaving as immune adjuvants or anti-sepsis molecules. In this scenario, we report the structural determination of the lipid A from three marine bacteria within the Cellulophaga genus, which showed to produce an extremely heterogenous blend of tetra- to hexa-acylated lipid A species, mostly carrying one phosphate and one D-mannose on the glucosamine disaccharide backbone. The ability of the three LPSs in activating TLR4 signaling revealed a weaker immunopotential by C. baltica NNO 15840T and C. tyrosinoxydans EM41T , while C. algicola ACAM 630T behaved as a more potent TLR4 activator.


Subject(s)
Flavobacteriaceae , Gammaproteobacteria , Lipid A/chemistry , Toll-Like Receptor 4 , Lipopolysaccharides/chemistry
12.
Microb Pathog ; 174: 105889, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36435436

ABSTRACT

Lipid A plays an important role in the pathogenicity and antimicrobial resistance of Vibrio parahaemolyticus, but little is known about the structure and biosynthesis of lipid A in V. parahaemolyticus. In this study, lipid A species were either directly extracted or obtained by the acid hydrolysis of lipopolysaccharide from V. parahaemolyticus ATCC33846 cells and analyzed by thin-layer chromatography and high-performance liquid chromatography-tandem mass spectrometry. Several lipid A species in V. parahaemolyticus cells were characterized, and two of these species were not connected to polysaccharides. One free lipid A species has the similar structure as the hexa-acylated lipid A in Escherichia coli, and the other is a hepta-acylated lipid A with an additional secondary C16:0 acyl chain. Three lipid A species were isolated by the acid hydrolysis of lipopolysaccharide: the 1st one has the similar structure as the hexa-acylated lipid A in E. coli, the 2nd one is a hepta-acylated lipid A with an additional secondary C16:0 acyl chain and a secondary 2-OH C12:0 acyl chain, and the 3rd one is equal to the 2nd species with a phosphoethanolamine modification. These results are important for understanding the biosynthesis of lipid A in V. parahaemolyticus.


Subject(s)
Lipopolysaccharides , Vibrio parahaemolyticus , Lipid A/chemistry , Escherichia coli , Mass Spectrometry
13.
Infect Immun ; 90(8): e0020822, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35862709

ABSTRACT

Detection of Gram-negative bacterial lipid A by the extracellular sensor, myeloid differentiation 2 (MD2)/Toll-like receptor 4 (TLR4), or the intracellular inflammasome sensors, CASP4 and CASP5, induces robust inflammatory responses. The chemical structure of lipid A, specifically its phosphorylation and acylation state, varies across and within bacterial species, potentially allowing pathogens to evade or suppress host immunity. Currently, it is not clear how distinct alterations in the phosphorylation or acylation state of lipid A affect both human TLR4 and CASP4/5 activation. Using a panel of engineered lipooligosaccharides (LOS) derived from Yersinia pestis with defined lipid A structures that vary in their acylation or phosphorylation state, we identified that differences in phosphorylation state did not affect TLR4 or CASP4/5 activation. However, the acylation state differentially impacted TLR4 and CASP4/5 activation. Specifically, all tetra-, penta-, and hexa-acylated LOS variants examined activated CASP4/5-dependent responses, whereas TLR4 responded to penta- and hexa-acylated LOS but did not respond to tetra-acylated LOS or penta-acylated LOS lacking the secondary acyl chain at the 3' position. As expected, lipid A alone was sufficient for TLR4 activation. In contrast, both core oligosaccharide and lipid A were required for robust CASP4/5 inflammasome activation in human macrophages, whereas core oligosaccharide was not required to activate mouse macrophages expressing CASP4. Our findings show that human TLR4 and CASP4/5 detect both shared and nonoverlapping LOS/lipid A structures, which enables the innate immune system to recognize a wider range of bacterial LOS/lipid A and would thereby be expected to constrain the ability of pathogens to evade innate immune detection.


Subject(s)
Lipid A , Toll-Like Receptor 4 , Acylation , Animals , Humans , Inflammasomes , Lipid A/chemistry , Lipopolysaccharides , Macrophages , Mice , Toll-Like Receptor 4/metabolism
14.
Infect Immun ; 90(8): e0020122, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35862717

ABSTRACT

Immune sensing of the Gram-negative bacterial membrane glycolipid lipopolysaccharide (LPS) is both a critical component of host defense against bacterial infection and a contributor to the hyperinflammatory response, potentially leading to sepsis and death. Innate immune activation by LPS is due to the lipid A moiety, an acylated di-glucosamine molecule that can activate inflammatory responses via the extracellular sensor Toll-like receptor 4 (TLR4)/myeloid differentiation 2 (MD2) or the cytosolic sensor caspase-11 (Casp11). The number and length of acyl chains present on bacterial lipid A structures vary across bacterial species and strains, which affects the magnitude of TLR4 and Casp11 activation. TLR4 and Casp11 are thought to respond similarly to various lipid A structures, as tetra-acylated lipid A structures do not activate either sensor, whereas hexa-acylated structures activate both sensors. However, the precise features of lipid A that determine the differential activation of each receptor remain poorly defined, as direct analysis of extracellular and cytosolic responses to the same sources and preparations of LPS/lipid A structures have been limited. To address this question, we used rationally engineered lipid A isolated from a series of bacterial acyl-transferase mutants that produce novel, structurally defined molecules. Intriguingly, we found that the location of specific secondary acyl chains on lipid A resulted in differential recognition by TLR4 or Casp11, providing new insight into the structural features of lipid A required to activate either TLR4 or Casp11. Our findings indicate that TLR4 and Casp11 sense nonoverlapping areas of lipid A chemical space, thereby constraining the ability of Gram-negative pathogens to evade innate immunity.


Subject(s)
Lipid A , Toll-Like Receptor 4 , Acylation , Animals , Caspases , Lipid A/chemistry , Lipopolysaccharides , Mice , Toll-Like Receptor 4/metabolism
15.
Microbiology (Reading) ; 168(4)2022 04.
Article in English | MEDLINE | ID: mdl-35394417

ABSTRACT

The lipopolysaccharide (LPS) is a characteristic molecule of the outer leaflet of the Gram-negative bacterial outer membrane, which consists of lipid A, core oligosaccharide, and O antigen. The lipid A is embedded in outer membrane and provides an efficient permeability barrier, which is particularly important to reduce the permeability of antibiotics, toxic cationic metals, and antimicrobial peptides. LPS, an important modulator of innate immune responses ranging from localized inflammation to disseminated sepsis, displays a high level of structural and functional heterogeneity, which arise due to regulated differences in the acylation of the lipid A and the incorporation of non-stoichiometric modifications in lipid A and the core oligosaccharide. This review focuses on the current mechanistic understanding of the synthesis and assembly of the lipid A molecule and its most salient non-stoichiometric modifications.


Subject(s)
Lipid A , Lipopolysaccharides , Bacterial Outer Membrane , Bacterial Outer Membrane Proteins/metabolism , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism , Lipid A/chemistry , Lipid A/metabolism , Lipopolysaccharides/chemistry
16.
J Antimicrob Chemother ; 77(9): 2441-2447, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35770844

ABSTRACT

OBJECTIVES: Neisseria gonorrhoeae is an exclusively human pathogen that commonly infects the urogenital tract resulting in gonorrhoea. Empirical treatment of gonorrhoea with antibiotics has led to multidrug resistance and the need for new therapeutics. Inactivation of lipooligosaccharide phosphoethanolamine transferase A (EptA), which attaches phosphoethanolamine to lipid A, results in attenuation of the pathogen in infection models. Small molecules that inhibit EptA are predicted to enhance natural clearance of gonococci via the human innate immune response. METHODS: A library of small-fragment compounds was tested for the ability to enhance susceptibility of the reference strain N. gonorrhoeae FA1090 to polymyxin B. The effect of these compounds on lipid A synthesis and viability in models of infection were tested. RESULTS: Three compounds, 135, 136 and 137, enhanced susceptibility of strain FA1090 to polymyxin B by 4-fold. Pre-treatment of bacterial cells with all three compounds resulted in enhanced killing by macrophages. Only lipid A from bacterial cells exposed to compound 137 showed a 17% reduction in the level of decoration of lipid A with phosphoethanolamine by MALDI-TOF MS analysis and reduced stimulation of cytokine responses in THP-1 cells. Binding of 137 occurred with higher affinity to purified EptA than the starting material, as determined by 1D saturation transfer difference NMR. Treatment of eight MDR strains with 137 increased susceptibility to polymyxin B in all cases. CONCLUSIONS: Small molecules have been designed that bind to EptA, inhibit addition of phosphoethanolamine to lipid A and can sensitize N. gonorrhoeae to killing by macrophages.


Subject(s)
Gonorrhea , Neisseria gonorrhoeae , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Peptides , Drug Resistance, Bacterial , Ethanolaminephosphotransferase/metabolism , Ethanolamines , Gonorrhea/drug therapy , Humans , Lipid A/chemistry , Microbial Sensitivity Tests , Polymyxin B/pharmacology
17.
Chemistry ; 28(35): e202200547, 2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35439332

ABSTRACT

TLR4 is a key pattern recognition receptor that can sense pathogen- and danger- associated molecular patterns to activate the downstream signaling pathways which results in the upregulation of transcription factors and expression of interferons and cytokines to mediate protective pro-inflammatory responses involved in immune defense. Bacterial lipid A is the primary TLR4 ligand with very complex, species-specific, and barely predictable structure-activity relationships. Given that therapeutic targeting of TLR4 is an emerging tool for management of a variety of human diseases, the development of novel TLR4 activating biomolecules other than lipid A is of vast importance. We report on design, chemical synthesis and immunobiology of novel glycan-based lipid A-mimicking molecules that can activate human and murine TLR4-mediated signaling with picomolar affinity. Exploiting crystal structure - based design we have created novel disaccharide lipid A mimetics (DLAMs) where the inherently flexible ß(1→6)-linked diglucosamine backbone of lipid A is exchanged with a conformationally restrained non-reducing ßGlcN(1↔1')ßGlcN scaffold. Excellent stereoselectivity in a challenging ß,ß-1,1' glycosylation was achieved by tuning the reactivities of donor and acceptor molecules using protective group manipulation strategy. Divergent streamlined synthesis of ß,ß-1,1'-linked diglucosamine-derived glycolipids entailing multiple long-chain (R)-3- acyloxyacyl residues and up two three phosphate groups was developed. Specific 3D-molecular shape and conformational rigidity of unnatural ß,ß-1,1'-linked diglucosamine combined with carefully optimized phosphorylation and acylation pattern ensured efficient induction of the TLR4-mediated signaling in a species-independent manner.


Subject(s)
Lipid A , Toll-Like Receptor 4 , Adjuvants, Immunologic/chemistry , Animals , Disaccharides/chemistry , Humans , Immunotherapy , Lipid A/chemistry , Mice , Prospective Studies , Toll-Like Receptor 4/agonists
18.
Semin Immunol ; 39: 4-13, 2018 10.
Article in English | MEDLINE | ID: mdl-30396811

ABSTRACT

The development of the CAF family adjuvant was initiated around 20 years ago when Statens Serum Institut was preparing its first generation protein based recombinant subunit vaccine against tuberculosis for clinical testing, but realized that there were no clinically relevant adjuvants available that would support the strong CMI response needed. Since then the aim for the adjuvant research at Statens Serum Institut has been to provide adjuvants with distinct immunogenicity profiles correlating with protection for any given infectious disease. Two of the adjuvants CAF01 and CAF09 are currently being evaluated in human clinical trials. The purpose of this review is to give an overview of the immunocorrelates of those CAF adjuvants furthest in development. We further aim at giving an overview of the mechanism of action of the CAF adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Glycolipids/pharmacology , Immunity, Cellular/drug effects , Immunogenicity, Vaccine , Lipid A/analogs & derivatives , Quaternary Ammonium Compounds/pharmacology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/chemistry , Animals , Glycolipids/chemistry , Humans , Immunity, Humoral/drug effects , Lipid A/chemistry , Lipid A/pharmacology , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Mice , Quaternary Ammonium Compounds/chemistry , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/microbiology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/microbiology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/microbiology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
19.
Semin Immunol ; 39: 22-29, 2018 10.
Article in English | MEDLINE | ID: mdl-30366662

ABSTRACT

Lipopolysaccharide (LPS) is a well-defined agonist of Toll-like receptor (TLR) 4 that activates innate immune responses and influences the development of the adaptive response during infection with Gram-negative bacteria. Many years ago, Dr. Edgar Ribi separated the adjuvant activity of LPS from its toxic effects, an effort that led to the development of monophosphoryl lipid A (MPL). MPL, derived from Salmonella minnesota R595, has progressed through clinical development and is now used in various product-enabling formulations to support the generation of antigen-specific responses in several commercial and preclinical vaccines. We have generated several synthetic lipid A molecules, foremost glucopyranosyl lipid adjuvant (GLA) and second-generation lipid adjuvant (SLA), and have advanced these to clinical trial for various indications. In this review we summarize the potential and current positioning of TLR4-based adjuvant formulations in approved and emerging vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Glucosides/pharmacology , Immunogenicity, Vaccine , Lipid A/analogs & derivatives , Tuberculosis/prevention & control , Adjuvants, Immunologic/chemistry , Alum Compounds/chemistry , Animals , Glucosides/chemistry , HIV Infections/immunology , HIV Infections/prevention & control , HIV Infections/virology , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Leishmaniasis/immunology , Leishmaniasis/parasitology , Leishmaniasis/prevention & control , Leprosy/immunology , Leprosy/parasitology , Leprosy/prevention & control , Lipid A/chemistry , Lipid A/pharmacology , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Mice , Schistosomiasis/immunology , Schistosomiasis/parasitology , Schistosomiasis/prevention & control , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccines/administration & dosage , Vaccines/chemistry , Vaccines/immunology
20.
Semin Immunol ; 39: 30-34, 2018 10.
Article in English | MEDLINE | ID: mdl-30122362

ABSTRACT

The discovery and wide spread use of vaccines have saved millions of lives in the past few decades. Vaccine adjuvants represent an integral part of the modern vaccines. Despite numerous efforts, however, only a handful of vaccine adjuvants is currently available for human use. A comprehensive understanding of the mechanisms of action of adjuvants is pivotal to harness the potential of existing and new adjuvants in mounting desirable immune responses to counter human pathogens. Decomposing the host response to vaccines and its components at systems level has recently been made possible owing to the recent advancements in Omics technology and cutting edge immunological assays powered by systems biology approaches. This approach has begun to shed light on the molecular signatures of several human vaccines and adjuvants. This review is an attempt to provide an overview of the recent efforts in systems analysis of vaccine adjuvants that are currently in clinic.


Subject(s)
Adjuvants, Immunologic/pharmacology , HIV Infections/prevention & control , Immunogenicity, Vaccine , Influenza, Human/prevention & control , Malaria, Falciparum/prevention & control , Systems Analysis , Adjuvants, Immunologic/chemistry , Animals , Drug Combinations , Glucosides/chemistry , Glucosides/pharmacology , HIV Infections/immunology , HIV Infections/virology , Humans , Immunity, Innate/drug effects , Influenza, Human/immunology , Influenza, Human/virology , Lipid A/chemistry , Lipid A/pharmacology , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Polysorbates/chemistry , Polysorbates/pharmacology , Squalene/chemistry , Squalene/pharmacology , Systems Biology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vaccines/administration & dosage , Vaccines/chemistry , Vaccines/immunology , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL