Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.326
Filtrar
1.
Glycobiology ; 34(10)2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39244665

RESUMEN

Lipooligosaccharides are the most abundant cell surface glycoconjugates on the outer membrane of Gram-negative bacteria. They play important roles in host-microbe interactions. Certain Gram-negative pathogenic bacteria cap their lipooligosaccharides with the sialic acid, N-acetylneuraminic acid (Neu5Ac), to mimic host glycans that among others protects these bacteria from recognition by the hosts immune system. This process of molecular mimicry is not fully understood and remains under investigated. To explore the functional role of sialic acid-capped lipooligosaccharides at the molecular level, it is important to have tools readily available for the detection and manipulation of both Neu5Ac on glycoconjugates and the involved sialyltransferases, preferably in live bacteria. We and others have shown that the native sialyltransferases of some Gram-negative bacteria can incorporate extracellular unnatural sialic acid nucleotides onto their lipooligosaccharides. We here report on the expanded use of native bacterial sialyltransferases to incorporate neuraminic acids analogs with a reporter group into the lipooligosaccharides of a variety of Gram-negative bacteria. We show that this approach offers a quick strategy to screen bacteria for the expression of functional sialyltransferases and the ability to use exogenous CMP-Neu5Ac to decorate their glycoconjugates. For selected bacteria we also show this strategy complements two other glycoengineering techniques, Metabolic Oligosaccharide Engineering and Selective Exo-Enzymatic Labeling, and that together they provide tools to modify, label, detect and visualize sialylation of bacterial lipooligosaccharides.


Asunto(s)
Lipopolisacáridos , Sialiltransferasas , Sialiltransferasas/metabolismo , Sialiltransferasas/genética , Sialiltransferasas/química , Lipopolisacáridos/metabolismo , Lipopolisacáridos/química , Ácidos Neuramínicos/metabolismo , Ácidos Neuramínicos/química , Bacterias Gramnegativas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/química
2.
Biochemistry ; 63(19): 2440-2448, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39264328

RESUMEN

Nonvesicular lipid trafficking pathways are an important process in every domain of life. The mechanisms of these processes are poorly understood in part due to the difficulty in kinetic characterization. One important class of glycolipids, lipopolysaccharides (LPS), are the primary lipidic component of the outer membrane of Gram-negative bacteria. LPS are synthesized in the inner membrane and then trafficked to the cell surface by the lipopolysaccharide transport proteins, LptB2FGCADE. By characterizing the interaction of a fluorescent probe and LPS, we establish a quantitative assay to monitor the flux of LPS between proteoliposomes on the time scale of seconds. We then incorporate photocaged ATP into this system, which allows for light-based control of the initiation of LPS transport. This control allows us to measure the initial rate of LPS transport (3.0 min-1 per LptDE). We also find that the rate of LPS transport by the Lpt complex is independent of the structure of LPS. In contrast, we find the rate of LPS transport is dependent on the proper function of the LptDE complex. Mutants of the outer membrane Lpt components, LptDE, that cause defective LPS assembly in live cells display attenuated transport rates and slower ATP hydrolysis compared to wild type proteins. Analysis of these mutants reveals that the rates of ATP hydrolysis and LPS transport are correlated such that 1.2 ± 0.2 ATP are hydrolyzed for each LPS transported. This correlation suggests a model where the outer membrane components ensure the coupling of ATP hydrolysis and LPS transport by stabilizing a transport-active state of the Lpt bridge.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Lipopolisacáridos , Lipopolisacáridos/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Transporte Biológico , Escherichia coli/metabolismo , Escherichia coli/genética , Cinética , Adenosina Trifosfato/metabolismo , Proteolípidos/metabolismo , Proteolípidos/química , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Transportadoras de Casetes de Unión a ATP
3.
Microb Pathog ; 194: 106839, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39103126

RESUMEN

Histophilus somni is an important pathogen of the bovine respiratory disease complex, yet the mechanisms underlying its virulence remain poorly understood. It is known that H. somni can incorporate sialic acid into lipooligosaccharide (LOS), and sialylated H. somni is more resistant to phagocytosis and complement-mediated killing by serum compared to non-sialylated bacteria in vitro. However, the virulence of non-sialylated H. somni has not been evaluated in vivo using an animal model. In this study, we investigated the contribution of sialic acid to virulence by constructing an H. somni sialic acid uptake mutant (ΔnanP-ΔnanU) and comparing the parent and mutant strains in a mouse septicemia and mortality model. Intraperitoneal challenge of mice with wildtype H. somni (1 × 108 colony forming units/mouse, CFU) was lethal to all animals. Mice challenged with three different doses (1, 2, or 5 × 108 CFU/mouse) of an H. somni ΔnanP-ΔnanU sialic acid uptake mutant exhibited survival rates of 90 %, 60 %, and 0 % respectively. High-performance anion exchange chromatography analyses revealed that LOS prepared from both parent and the ΔnanP-ΔnanU mutant strains of H. somni were sialylated. These findings suggest the presence of de novo sialic acid synthesis pathway, although the genes associated with de novo sialic acid synthesis (neuB and neuC) were not identified by genomic analysis. The lower attenuation in mice is most likely attributed to the sialylated LOS of H. somni nanPU mutant.


Asunto(s)
Modelos Animales de Enfermedad , Lipopolisacáridos , Ácido N-Acetilneuramínico , Pasteurellaceae , Sepsis , Animales , Ratones , Ácido N-Acetilneuramínico/metabolismo , Pasteurellaceae/genética , Pasteurellaceae/patogenicidad , Pasteurellaceae/metabolismo , Virulencia/genética , Sepsis/microbiología , Sepsis/mortalidad , Lipopolisacáridos/metabolismo , Lipopolisacáridos/genética , Femenino , Mutación , Bovinos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
4.
Sci Rep ; 14(1): 19258, 2024 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164328

RESUMEN

Cyanobacteria possess special defense mechanisms to protect themselves against ultraviolet (UV) radiation. This study combines experimental and computational methods to identify the role of protective strategies in Nostoc species against UV-C radiation. To achieve this goal, various species of the genus Nostoc from diverse natural habitats in Iran were exposed to artificial UV-C radiation. The results indicated that UV-C treatment significantly reduced the photosynthetic pigments while simultaneously increasing the activity of antioxidant enzymes. Notably, N. sphaericum ISB97 and Nostoc sp. ISB99, the brown Nostoc species isolated from habitats with high solar radiations, exhibited greater resistance compared to the green-colored species. Additionally, an increase in scytonemin content occurred with a high expression of key genes associated with its synthesis (scyF and scyD) during the later stages of UV-C exposure in these species. The molecular docking of scytonemin with lipopolysaccharides of the cyanobacteria that mainly cover the extracellular matrix revealed the top/side positioning of scytonemin on the glycans of these lipopolysaccharides to form a UV-protective shield. These findings pave the way for exploring the molecular effects of scytonemin in forming the UV protection shield in cyanobacteria, an aspect that has been ambiguous until now.


Asunto(s)
Nostoc , Rayos Ultravioleta , Nostoc/metabolismo , Nostoc/efectos de la radiación , Simulación del Acoplamiento Molecular , Fenoles/metabolismo , Indoles/metabolismo , Indoles/química , Fotosíntesis/efectos de la radiación , Lipopolisacáridos/metabolismo
5.
J Med Chem ; 67(17): 15620-15675, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39172133

RESUMEN

Acinetobacter baumannii, a commonly multidrug-resistant Gram-negative bacterium responsible for large numbers of bloodstream and lung infections worldwide, is increasingly difficult to treat and constitutes a growing threat to human health. Structurally novel antibacterial chemical matter that can evade existing resistance mechanisms is essential for addressing this critical medical need. Herein, we describe our efforts to inhibit the essential A. baumannii lipooligosaccharide (LOS) ATP-binding cassette (ABC) transporter MsbA. An unexpected impurity from a phenotypic screening was optimized as a series of dimeric compounds, culminating with 1 (cerastecin D), which exhibited antibacterial activity in the presence of human serum and a pharmacokinetic profile sufficient to achieve efficacy against A. baumannii in murine septicemia and lung infection models.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Infecciones por Acinetobacter , Acinetobacter baumannii , Antibacterianos , Proteínas Bacterianas , Lipopolisacáridos , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Animales , Lipopolisacáridos/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Ratones , Humanos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/metabolismo , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Pruebas de Sensibilidad Microbiana
6.
Microbiol Spectr ; 12(9): e0099724, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39105587

RESUMEN

We previously reported that a linear cationic 12-amino acid cell-penetrating peptide (CPP) was bactericidal for Neisseria gonorrhoeae. In this study, our objectives were to determine the effect of cyclization of the linear CPP on its antibacterial activity for N. gonorrhoeae and cytotoxicity for human cells. We compared the bactericidal effect of 4-hour treatment with the linear CPP to that of CPPs cyclized by a thioether or a disulfide bond on human challenge and multi-drug resistant (MDR) strains of N. gonorrhoeae grown in cell culture media with 10% fetal bovine serum (FBS). The effect of lipooligosaccharide (LOS) sialylation on bactericidal activity was analyzed. We determined the ability of the CPPs to treat human cells infected in vitro with N. gonorrhoeae, to reduce the inflammatory response of human monocytic cells to gonococci, to kill strains of three commensal Neisseria species, and to inhibit gonococcal biofilms. The cyclized CPPs killed 100% of gonococci from all strains at 100 µM and >90% at 20 µM and were more potent than the linear form. The thioether-linked but not the disulfide-linked CPP was less cytotoxic for human cervical cells compared to the linear CPP. LOS sialylation had minimal effect on bactericidal activity. In treating infected human cells, the thioether-linked CPP at 20 µM killed >60% of extra- and intracellular bacteria and reduced TNF-α expression by THP-1 cells. The potency of the CPPs for the pathogenic and the commensal Neisseria was similar. The thioether-linked CPP partially eradicated gonococcal biofilms. Future studies will focus on determining efficacy in the female mouse model of gonorrhea.IMPORTANCENeisseria gonorrhoeae remains a major cause of sexually transmitted infections with 82 million cases worldwide in 2020, and 710,151 confirmed cases in the US in 2021, up 25% from 2017. N. gonorrhoeae can infect multiple tissues including the urethra, cervix, rectum, pharynx, and conjunctiva. The most serious sequelae are suffered by infected women as gonococci ascend to the upper reproductive tract and cause pelvic inflammatory disease, chronic pelvic pain, and infertility in 10%-20% of women. Control of gonococcal infection is widely recognized as increasingly challenging due to the lack of any vaccine. N. gonorrhoeae has quickly developed resistance to all but one class of antibiotics and the emergence of multidrug-resistant strains could result in untreatable infections. As such, gonorrhea is classified by the Center for Disease Control (CDC) as an urgent public health threat. The research presented herein on new therapeutics for gonorrhea has identified a cyclic cell-penetrating peptide (CPP) as a potent molecule targeting N. gonorrhoeae.


Asunto(s)
Antibacterianos , Péptidos de Penetración Celular , Gonorrea , Neisseria gonorrhoeae , Neisseria gonorrhoeae/efectos de los fármacos , Humanos , Gonorrea/tratamiento farmacológico , Gonorrea/microbiología , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/química , Antibacterianos/farmacología , Antibacterianos/química , Animales , Ratones , Femenino , Biopelículas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Ciclización , Lipopolisacáridos/metabolismo , Arginina/farmacología , Arginina/química
7.
Microbiology (Reading) ; 170(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39177453

RESUMEN

Escherichia coli (E. coli) is a major cause of urinary tract infections, bacteraemia, and sepsis. CFT073 is a prototypic, urosepsis isolate of sequence type (ST) 73. This laboratory, among others, has shown that strain CFT073 is resistant to serum, with capsule and other extracellular polysaccharides imparting resistance. The interplay of such polysaccharides remains under-explored. This study has shown that CFT073 mutants deficient in lipopolysaccharide (LPS) O-antigen and capsule display exquisite serum sensitivity. Additionally, O-antigen and LPS outer core mutants displayed significantly decreased surface K2 capsule, coupled with increased unbound K2 capsule being detected in the supernatant. The R1 core and O6 antigen are involved in the tethering of K2 capsule to the CFT073 cell surface, highlighting the importance of the R1 core in serum resistance. The dependence of capsule on LPS was shown to be post-transcriptional and related to changes in cell surface hydrophobicity. Furthermore, immunofluorescence microscopy suggested that the surface pattern of capsule is altered in such LPS core mutants, which display a punctate capsule pattern. Finally, targeting LPS biosynthesis using sub-inhibitory concentrations of a WaaG inhibitor resulted in increased serum sensitivity and decreased capsule in CFT073. Interestingly, the dependency of capsule on LPS has been observed previously in other Enterobacteria, indicating that the synergy between these polysaccharides is not just strain, serotype or species-specific but may be conserved across several pathogenic Gram-negative species. Therefore, using WaaG inhibitor derivatives to target LPS is a promising therapeutic strategy to reduce morbidity and mortality by reducing or eliminating surface capsule.


Asunto(s)
Cápsulas Bacterianas , Lipopolisacáridos , Lipopolisacáridos/metabolismo , Cápsulas Bacterianas/metabolismo , Cápsulas Bacterianas/genética , Humanos , Escherichia coli Patógena Extraintestinal/genética , Escherichia coli Patógena Extraintestinal/efectos de los fármacos , Escherichia coli Patógena Extraintestinal/metabolismo , Antígenos O/genética , Antígenos O/metabolismo , Infecciones por Escherichia coli/microbiología , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Mutación
8.
Mol Immunol ; 173: 100-109, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39094445

RESUMEN

Antimicrobial peptides (AMPs) are a promising alternative to antibiotics in the fight against multi-drug resistant and immune system-evading bacterial infections. Protegrins are porcine cathelicidins which have been identified in porcine leukocytes. Protegrin-1 is the best characterized family member and has broad antibacterial activity by interacting and permeabilizing bacterial membranes. Many host defense peptides (HDPs) like LL-37 or chicken cathelicidin 2 (CATH-2) have also been shown to have protective biological functions during infections. In this regard, it is interesting to study if Protegrin-1 has the immune modulating potential to suppress unnecessary immune activation by neutralizing endotoxins or by influencing the macrophage functionality in addition to its direct antimicrobial properties. This study showed that Protegrin-1 neutralized lipopolysaccharide- (LPS) and bacteria-induced activation of RAW macrophages by binding and preventing LPS from cell surface attachment. Furthermore, the peptide treatment not only inhibited bacterial phagocytosis by murine and porcine macrophages but also interfered with cell surface and intracellular bacterial survival. Lastly, Protegrin-1 pre-treatment was shown to inhibit the amastigote survival in Leishmania infected macrophages. These experiments describe an extended potential of Protegrin-1's protective role during microbial infections and add to the research towards clinical application of cationic AMPs.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Catelicidinas , Lipopolisacáridos , Macrófagos , Fagocitosis , Animales , Ratones , Antiinfecciosos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Factores Inmunológicos/farmacología , Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Células RAW 264.7 , Porcinos
9.
Int J Mol Sci ; 25(16)2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39201412

RESUMEN

Galectins are ß-galactoside-binding animal lectins involved in various biological functions, such as host defense. Galectin-2 and -3 are members of the galectin family that are expressed in the stomach, including the gastric mucosa and surface mucous cells. Galectin-3 exhibits aggregation and bactericidal activity against Helicobacter pylori in a ß-galactoside-dependent manner. We previously reported that galectin-2 has the same activity under neutral pH conditions. In this study, the H. pylori aggregation activity of galectin-2 was examined under weakly acidic conditions, in which H. pylori survived. Galectin-2 agglutinated H. pylori even at pH 6.0, but not at pH 5.0, correlating with its structural stability, as determined using circular dichroism. Additionally, galectin-2 binding to the lipopolysaccharide (LPS) of H. pylori cultured under weakly acidic conditions was investigated using affinity chromatography and Western blotting. Galectin-2 could bind to H. pylori LPS containing H type I, a Lewis antigen, in a ß-galactoside-dependent manner. In contrast, galectin-3 was structurally more stable than galectin-2 under acidic conditions and bound to H. pylori LPS containing H type I and Lewis X. In conclusion, galectin-2 and -3 might function cooperatively in the defense against H. pylori in the stomach under different pH conditions.


Asunto(s)
Galectina 2 , Helicobacter pylori , Lipopolisacáridos , Helicobacter pylori/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/química , Concentración de Iones de Hidrógeno , Galectina 2/metabolismo , Galectina 2/química , Humanos , Galectina 3/metabolismo , Galectina 3/química , Unión Proteica , Aglutinación , Galectinas/metabolismo , Galectinas/química
10.
Methods Mol Biol ; 2851: 193-199, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39210183

RESUMEN

Some gram-negative bacteria such as Escherichia coli and Salmonella spp. among intestinal bacteria might induce inflammation of human intestines. However, lactic acid bacteria (LAB) display anti-inflammatory activity, which improves the intestinal environment. Particularly, the cell surface protein on Pediococcus pentosaceus exhibits high LPS elimination activity, which is expected to provide anti-inflammatory activity in the intestines. This chapter describes that surface proteins are separable using Blue-Native PAGE, which relies upon the theory that protein binds with Coomassie brilliant blue to produce a negative charge for easy separation.


Asunto(s)
Lactobacillales , Lipopolisacáridos , Colorantes de Rosanilina , Lipopolisacáridos/metabolismo , Lactobacillales/metabolismo , Colorantes de Rosanilina/química , Colorantes de Rosanilina/metabolismo , Humanos , Electroforesis en Gel de Poliacrilamida Nativa/métodos , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo
11.
Nat Commun ; 15(1): 6958, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138228

RESUMEN

Outer membrane vesicles (OMVs) produced by Gram-negative bacteria have key roles in cell envelope homeostasis, secretion, interbacterial communication, and pathogenesis. The facultative intracellular pathogen Salmonella Typhimurium increases OMV production inside the acidic vacuoles of host cells by changing expression of its outer membrane proteins and modifying the composition of lipid A. However, the molecular mechanisms that translate pH changes into OMV production are not completely understood. Here, we show that the outer membrane protein PagC promotes OMV production through pH-dependent interactions between its extracellular loops and surrounding lipopolysaccharide (LPS). Structural comparisons and mutational studies indicate that a pH-responsive amino acid motif in PagC extracellular loops, containing PagC-specific histidine residues, is crucial for OMV formation. Molecular dynamics simulations suggest that protonation of histidine residues leads to changes in the structure and flexibility of PagC extracellular loops and their interactions with the surrounding LPS, altering membrane curvature. Consistent with that hypothesis, mimicking acidic pH by mutating those histidine residues to lysine increases OMV production. Thus, our findings reveal a mechanism for sensing and responding to environmental pH and for control of membrane dynamics by outer membrane proteins.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Lipopolisacáridos , Simulación de Dinámica Molecular , Salmonella typhimurium , Concentración de Iones de Hidrógeno , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Salmonella typhimurium/metabolismo , Salmonella typhimurium/genética , Lipopolisacáridos/metabolismo , Membrana Externa Bacteriana/metabolismo , Secuencias de Aminoácidos , Histidina/metabolismo
12.
J Extracell Vesicles ; 13(7): e12474, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39001704

RESUMEN

Gut microbiome dysbiosis is a major contributing factor to several pathological conditions. However, the mechanistic understanding of the communication between gut microbiota and extra-intestinal organs remains largely elusive. Extracellular vesicles (EVs), secreted by almost every form of life, including bacteria, could play a critical role in this inter-kingdom crosstalk and are the focus of present study. Here, we present a novel approach for isolating lipopolysaccharide (LPS)+ bacterial extracellular vesicles (bEVLPS) from complex biological samples, including faeces, plasma and the liver from lean and diet-induced obese (DIO) mice. bEVLPS were extensively characterised using nanoparticle tracking analyses, immunogold labelling coupled with transmission electron microscopy, flow cytometry, super-resolution microscopy and 16S sequencing. In liver tissues, the protein expressions of TLR4 and a few macrophage-specific biomarkers were assessed by immunohistochemistry, and the gene expressions of inflammation-related cytokines and their receptors (n = 89 genes) were measured using a PCR array. Faecal samples from DIO mice revealed a remarkably lower concentration of total EVs but a significantly higher percentage of LPS+ EVs. Interestingly, DIO faecal bEVLPS showed a higher abundance of Proteobacteria by 16S sequencing. Importantly, in DIO mice, a higher number of total EVs and bEVLPS consistently entered the hepatic portal vein and subsequently reached the liver, associated with increased expression of TLR4, macrophage markers (F4/80, CD86 and CD206), cytokines and receptors (Il1rn, Ccr1, Cxcl10, Il2rg and Ccr2). Furthermore, a portion of bEVLPS escaped liver and entered the peripheral circulation. In conclusion, bEV could be the key mediator orchestrating various well-established biological effects induced by gut bacteria on distant organs.


Asunto(s)
Vesículas Extracelulares , Microbioma Gastrointestinal , Lipopolisacáridos , Hígado , Vena Porta , Animales , Vesículas Extracelulares/metabolismo , Hígado/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Vena Porta/metabolismo , Ratones Endogámicos C57BL , Masculino , Bacterias/metabolismo , Receptor Toll-Like 4/metabolismo , Obesidad/metabolismo , Obesidad/microbiología , Heces/microbiología , Disbiosis/metabolismo , Disbiosis/microbiología
13.
Nat Commun ; 15(1): 5740, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982040

RESUMEN

Mycobacterial glycolipids are important cell envelope structures that drive host-pathogen interactions. Arguably, the most important are lipoarabinomannan (LAM) and its precursor, lipomannan (LM), which are trafficked from the bacterium to the host via unknown mechanisms. Arabinomannan is thought to be a capsular derivative of these molecules, lacking a lipid anchor. However, the mechanism by which this material is generated has yet to be elucidated. Here, we describe the identification of a glycoside hydrolase family 76 enzyme that we term LamH (Rv0365c in Mycobacterium tuberculosis) which specifically cleaves α-1,6-mannoside linkages within LM and LAM, driving its export to the capsule releasing its phosphatidyl-myo-inositol mannoside lipid anchor. Unexpectedly, we found that the catalytic activity of this enzyme is important for efficient exit from stationary phase cultures, potentially implicating arabinomannan as a signal for growth phase transition. Finally, we demonstrate that LamH is important for M. tuberculosis survival in macrophages.


Asunto(s)
Proteínas Bacterianas , Glicósido Hidrolasas , Lipopolisacáridos , Macrófagos , Mananos , Mycobacterium tuberculosis , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/crecimiento & desarrollo , Lipopolisacáridos/metabolismo , Mananos/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Glicósido Hidrolasas/metabolismo , Proteínas Bacterianas/metabolismo , Animales , Ratones , Humanos , Fosfatidilinositoles/metabolismo , Cápsulas Bacterianas/metabolismo
14.
Commun Biol ; 7(1): 883, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39030323

RESUMEN

Metamorphosis for many marine invertebrates is triggered by external cues, commonly produced by bacteria. For larvae of Hydroides elegans, lipopolysaccharide (LPS) from the biofilm-dwelling bacterium Cellulophaga lytica induces metamorphosis. To determine whether bacterial LPS is a common metamorphosis-inducing factor for this species, we compare larval responses to LPS from 3 additional inductive Gram-negative marine biofilm bacteria with commercially available LPS from 3 bacteria not known to induce metamorphosis. LPS from all the inductive bacteria trigger metamorphosis, while LPS from non-inductive isolated marine bacteria do not. We then ask, which part of the LPS is the inductive element, the lipid (Lipid-A) or the polysaccharide (O-antigen), and find it is the latter for all four inductive bacteria. Finally, we examine the LPS subunits from two strains of the same bacterial species, one inductive and the other not, and find the LPS and O-antigen to be inductive from only the inductive bacterial strain.


Asunto(s)
Metamorfosis Biológica , Poliquetos , Animales , Poliquetos/crecimiento & desarrollo , Poliquetos/fisiología , Poliquetos/microbiología , Lipopolisacáridos/metabolismo , Incrustaciones Biológicas , Larva/crecimiento & desarrollo , Larva/microbiología , Biopelículas/crecimiento & desarrollo
15.
Nat Commun ; 15(1): 5946, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009687

RESUMEN

The ATP-binding cassette (ABC) transporter, MsbA, plays a pivotal role in lipopolysaccharide (LPS) biogenesis by facilitating the transport of the LPS precursor lipooligosaccharide (LOS) from the cytoplasmic to the periplasmic leaflet of the inner membrane. Despite multiple studies shedding light on MsbA, the role of lipids in modulating MsbA-nucleotide interactions remains poorly understood. Here we use native mass spectrometry (MS) to investigate and resolve nucleotide and lipid binding to MsbA, demonstrating that the transporter has a higher affinity for adenosine 5'-diphosphate (ADP). Moreover, native MS shows the LPS-precursor 3-deoxy-D-manno-oct-2-ulosonic acid (Kdo)2-lipid A (KDL) can tune the selectivity of MsbA for adenosine 5'-triphosphate (ATP) over ADP. Guided by these studies, four open, inward-facing structures of MsbA are determined that vary in their openness. We also report a 2.7 Å-resolution structure of MsbA in an open, outward-facing conformation that is not only bound to KDL at the exterior site, but with the nucleotide binding domains (NBDs) adopting a distinct nucleotide-free structure. The results obtained from this study offer valuable insight and snapshots of MsbA during the transport cycle.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Adenosina Difosfato , Adenosina Trifosfato , Espectrometría de Masas , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Adenosina Trifosfato/metabolismo , Adenosina Difosfato/metabolismo , Espectrometría de Masas/métodos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Lipopolisacáridos/metabolismo , Lípido A/metabolismo , Lípido A/química , Unión Proteica , Modelos Moleculares , Cristalografía por Rayos X , Lípidos/química , Escherichia coli/metabolismo , Conformación Proteica
16.
mBio ; 15(8): e0151224, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39037275

RESUMEN

Staphylococcus aureus produces a plethora of virulence factors critical to its ability to establish an infection and cause disease. We have previously characterized a small membrane protein, MspA, which has pleiotropic effects on virulence and contributes to S. aureus pathogenicity in vivo. Here we report that mspA inactivation triggers overaccumulation of the essential cell wall component, lipoteichoic acid (LTA), which, in turn, decreases autolytic activity and leads to increased cell size due to a delay in cell separation. We show that MspA directly interacts with the enzymes involved in LTA biosynthesis (LtaA, LtaS, UgtP, and SpsB), interfering with their normal activities. MspA, in particular, interacts with the type I signal peptidase SpsB, limiting its cleavage of LtaS into its active form. These findings suggest that MspA contributes to maintaining a physiological level of LTA in the cell wall by interacting with and inhibiting the activity of SpsB, thereby uncovering a critical role for the MspA protein in regulating cell envelope biosynthesis and pathogenicity.IMPORTANCEThe S. aureus cell envelope, comprising the cytoplasmic membrane, a thick peptidoglycan layer, and the anionic polymers lipoteichoic acid and wall teichoic acids, is fundamental for bacterial growth and division, as well as being the main interface between the pathogen and the host. It has become increasingly apparent that the synthesis and turnover of cell envelope components also affect the virulence of S. aureus. In this study, we show that MspA, an effector of S. aureus virulence, contributes to the maintenance of normal levels of lipoteichoic acid in the cell wall, with implications on cell cycle and size. These findings further our understanding of the connections between envelope synthesis and pathogenicity and suggest that MspA represents a promising target for the development of future therapeutic strategies.


Asunto(s)
Proteínas Bacterianas , Pared Celular , Lipopolisacáridos , Staphylococcus aureus , Ácidos Teicoicos , Ácidos Teicoicos/biosíntesis , Ácidos Teicoicos/metabolismo , Lipopolisacáridos/biosíntesis , Lipopolisacáridos/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Pared Celular/metabolismo , Factores de Virulencia/metabolismo , Virulencia , Infecciones Estafilocócicas/microbiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Regulación Bacteriana de la Expresión Génica , Animales , Ratones , Serina Endopeptidasas
17.
J Appl Microbiol ; 135(7)2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-39066493

RESUMEN

AIMS: Microbial transformation to modify saponins and enhance their biological activities has received increasing attention in recent years. This study aimed to screen the strain that can biotransform notoginsenoside R1, identify the product and study its biological activity. METHODS AND RESULTS: A lactic acid bacteria strain S165 with glycosidase-producing activity was isolated from traditional Chinese fermented foods, which was identified and grouped according to API 50 CHL kit and 16S rDNA sequence analysis. Subsequently, notoginsenoside R1 underwent a 30-day fermentation period by the strain S165, and the resulting products were analyzed using High-performance liquid chromatography (HPLC), Ultra-performance liquid chromatography (UPLC)-mass spectrometry (MS)/MS, and 13C-Nuclear magnetic resonance (NMR) techniques. Employing a model of Lipopolysaccharide (LPS)-induced damage to Caco-2 cells, the damage of Caco-2 cells was detected by Hoechst 33 258 staining, and the activity of notoginsenoside R1 biotransformation product was investigated by CCK-8 and western blotting assay. The strain S165 was identified as Lactiplantibacillus plantarum and was used to biotransform notoginsenoside R1. Through a 30-day biotransformation, L. plantarum S165 predominantly converts notoginsenoside R1 into 3ß,12ß-dihydroxydammar-(E)-20(22),24-diene-6-O-ß-D-xylopyranosyl-(1→2)-ß-D-glucopyranoside, temporarily named notoginsenoside T6 (NGT6) according to HPLC, UPLC-MS/MS, and 13C-NMR analysis. Results from CCK-8 and Hoechst 33258 staining indicated that the ability notoginsenoside T6 to alleviate the intestinal injury induced by LPS in the Caco-2 cell was stronger than that of notoginsenoside R1. In addition, Western blotting result showed that notoginsenoside T6 could prevent intestinal injury by protecting tight junction proteins (Claudin-1, Occludin, and ZO-1). CONCLUSION: Notoginsenoside R1 was biotransformed into the notoginsenoside T6 by L. plantarum S165, and the biotransformed product showed an enhanced intestinal protective effect in vitro.


Asunto(s)
Ginsenósidos , Lipopolisacáridos , Ginsenósidos/metabolismo , Ginsenósidos/farmacología , Humanos , Células CACO-2 , Lipopolisacáridos/metabolismo , Fermentación , Biotransformación , Cromatografía Líquida de Alta Presión , Lactobacillus plantarum/metabolismo , Alimentos Fermentados/microbiología
18.
Front Cell Infect Microbiol ; 14: 1418651, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38933693

RESUMEN

Background: This study unveils the intricate functional association between cyclic di-3',5'-adenylic acid (c-di-AMP) signaling, cellular bioenergetics, and the regulation of lipopolysaccharide (LPS) profile in Porphyromonas gingivalis, a Gram-negative obligate anaerobe considered as a keystone pathogen involved in the pathogenesis of chronic periodontitis. Previous research has identified variations in P. gingivalis LPS profile as a major virulence factor, yet the underlying mechanism of its modulation has remained elusive. Methods: We employed a comprehensive methodological approach, combining two mutants exhibiting varying levels of c-di-AMP compared to the wild type, alongside an optimized analytical methodology that combines conventional mass spectrometry techniques with a novel approach known as FLATn. Results: We demonstrate that c-di-AMP acts as a metabolic nexus, connecting bioenergetic status to nuanced shifts in fatty acid and glycosyl profiles within P. gingivalis LPS. Notably, the predicted regulator gene cdaR, serving as a potent regulator of c-di-AMP synthesis, was found essential for producing N-acetylgalactosamine and an unidentified glycolipid class associated with the LPS profile. Conclusion: The multifaceted roles of c-di-AMP in bacterial physiology are underscored, emphasizing its significance in orchestrating adaptive responses to stimuli. Furthermore, our findings illuminate the significance of LPS variations and c-di-AMP signaling in determining the biological activities and immunostimulatory potential of P. gingivalis LPS, promoting a pathoadaptive strategy. The study expands the understanding of c-di-AMP pathways in Gram-negative species, laying a foundation for future investigations into the mechanisms governing variations in LPS structure at the molecular level and their implications for host-pathogen interactions.


Asunto(s)
Lipopolisacáridos , Porphyromonas gingivalis , Transducción de Señal , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/genética , Lipopolisacáridos/metabolismo , Factores de Virulencia/metabolismo , Regulación Bacteriana de la Expresión Génica , Metabolismo Energético , Fosfatos de Dinucleósidos/metabolismo , Ácidos Grasos/metabolismo , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
19.
Infect Genet Evol ; 123: 105625, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38906517

RESUMEN

The genus Pseudochrobactrum encompasses free-living bacteria phylogenetically close to Ochrobactrum opportunistic pathogens and to Brucella, facultative intracellular parasites causing brucellosis, a worldwide-extended and grave zoonosis. Recently, Pseudochrobactrum strains were isolated from Brucella natural hosts on Brucella selective media, potentially causing diagnostic confusions. Strikingly, P. algeriensis was isolated from cattle lymph nodes, organs that are inimical to bacteria. Here, we analyse P. algeriensis potential virulence factors in comparison with Ochrobactrum and Brucella. Consistent with genomic analyses, Western-Blot analyses confirmed that P. algeriensis lacks the ability to synthesize the N-formylperosamine O-polysaccharide characteristic of the lipopolysaccharide (LPS) of smooth Brucella core species. However, unlike other Pseudochrobactrum but similar to some early diverging brucellae, P. algeriensis carries genes potentially synthetizing a rhamnose-based O-polysaccharide LPS. Lipid A analysis by MALDI-TOF demonstrated that P. algeriensis LPS bears a lipid A with a reduced pathogen-associated molecular pattern, a trait shared with Ochrobactrum and Brucella that is essential to generate a highly stable outer membrane and to delay immune activation. Also, although not able to multiply intracellularly in macrophages, the analysis of P. algeriensis cell lipid envelope revealed the presence of large amounts of cationic aminolipids, which may account for the extremely high resistance of P. algeriensis to bactericidal peptides and could favor colonization of mucosae and transient survival in Brucella hosts. However, two traits critical in Brucella pathogenicity are either significantly different (T4SS [VirB]) or absent (erythritol catabolic pathway) in P. algeriensis. This work shows that, while diverging in other characteristics, lipidic envelope features relevant in Brucella pathogenicity are conserved in Brucellaceae. The constant presence of these features strongly suggests that reinforcement of the envelope integrity as an adaptive advantage in soil was maintained in Brucella because of the similarity of some environmental challenges, such as the action of cationic peptide antibiotics and host defense peptides. This information adds knowledge about the evolution of Brucellaceae, and also underlines the taxonomical differences of the three genera compared.


Asunto(s)
Filogenia , Animales , Factores de Virulencia/genética , Brucellaceae/genética , Brucellaceae/metabolismo , Brucella/genética , Brucella/clasificación , Lipopolisacáridos/metabolismo , Genoma Bacteriano , Ochrobactrum/genética , Bovinos , Brucelosis/microbiología , Lípido A/metabolismo , Lípido A/química
20.
Cell Host Microbe ; 32(6): 913-924.e7, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38870903

RESUMEN

Aspects of how Burkholderia escape the host's intrinsic immune response to replicate in the cell cytosol remain enigmatic. Here, we show that Burkholderia has evolved two mechanisms to block the activity of Ring finger protein 213 (RNF213)-mediated non-canonical ubiquitylation of bacterial lipopolysaccharide (LPS), thereby preventing the initiation of antibacterial autophagy. First, Burkholderia's polysaccharide capsule blocks RNF213 association with bacteria and second, the Burkholderia deubiquitylase (DUB), TssM, directly reverses the activity of RNF213 through a previously unrecognized esterase activity. Structural analysis provides insight into the molecular basis of TssM esterase activity, allowing it to be uncoupled from its isopeptidase function. Furthermore, a putative TssM homolog also displays esterase activity and removes ubiquitin from LPS, establishing this as a virulence mechanism. Of note, we also find that additional immune-evasion mechanisms exist, revealing that overcoming this arm of the host's immune response is critical to the pathogen.


Asunto(s)
Proteínas Bacterianas , Burkholderia , Lipopolisacáridos , Ubiquitinación , Lipopolisacáridos/metabolismo , Humanos , Burkholderia/inmunología , Proteínas Bacterianas/metabolismo , Esterasas/metabolismo , Evasión Inmune , Ubiquitina-Proteína Ligasas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Autofagia , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA