Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Bioconjug Chem ; 35(4): 489-498, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38591251

RESUMEN

The role of the intestinal microbiota in host health is increasingly revealed in its contributions to disease states. The host-microbiome interaction is multifactorial and dynamic. One of the factors that has recently been strongly associated with host physiological responses is peptidoglycan from bacterial cell walls. Peptidoglycan from gut commensal bacteria activates peptidoglycan sensors in human cells, including the nucleotide-binding oligomerization domain-containing protein 2. When present in the gastrointestinal tract, both the polymeric form (sacculi) and depolymerized fragments can modulate host physiology, including checkpoint anticancer therapy efficacy, body temperature and appetite, and postnatal growth. To utilize this growing area of biology toward therapeutic prescriptions, it will be critical to directly analyze a key feature of the host-microbiome interaction from living hosts in a reproducible and noninvasive way. Here we show that metabolically labeled peptidoglycan/sacculi can be readily isolated from fecal samples collected from both mice and humans. Analysis of fecal samples provided a noninvasive route to probe the gut commensal community including the metabolic synchronicity with the host circadian clock. Together, these results pave the way for noninvasive diagnostic tools to interrogate the causal nature of peptidoglycan in host health and disease.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Animales , Ratones , Peptidoglicano , Bacterias/metabolismo
2.
Angew Chem Int Ed Engl ; 63(3): e202313870, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38051128

RESUMEN

Staphylococcus aureus (S. aureus) has evolved the ability to persist after uptake into host immune cells. This intracellular niche enables S. aureus to potentially escape host immune responses and survive the lethal actions of antibiotics. While the elevated tolerance of S. aureus to small-molecule antibiotics is likely to be multifactorial, we pose that there may be contributions related to permeation of antibiotics into phagocytic vacuoles, which would require translocation across two mammalian bilayers. To empirically test this, we adapted our recently developed permeability assay to determine the accumulation of FDA-approved antibiotics into phagocytic vacuoles of live macrophages. Bioorthogonal reactive handles were metabolically anchored within the surface of S. aureus, and complementary tags were chemically added to antibiotics. Following phagocytosis of tagged S. aureus cells, we were able to specifically analyze the arrival of antibiotics within the phagosomes of infected macrophages. Our findings enabled the determination of permeability differences between extra- and intracellular S. aureus, thus providing a roadmap to dissect the contribution of antibiotic permeability to intracellular pathogens.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Animales , Staphylococcus aureus/fisiología , Antibacterianos/farmacología , Macrófagos , Fagosomas , Fagocitosis , Infecciones Estafilocócicas/tratamiento farmacológico , Mamíferos
3.
Angew Chem Int Ed Engl ; 62(20): e202217777, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36700874

RESUMEN

The general lack of permeability of small molecules observed for Mycobacterium tuberculosis (Mtb) is most ascribed to its unique cell envelope. More specifically, the outer mycomembrane is hypothesized to be the principal determinant for access of antibiotics to their molecular targets. We describe a novel assay that combines metabolic tagging of the peptidoglycan, which sits directly beneath the mycomembrane, click chemistry of test molecules, and a fluorescent labeling chase step, to measure the permeation of small molecules. We showed that the assay workflow was robust and compatible with high-throughput analysis in mycobacteria by testing a small panel of azide-tagged molecules. The general trend is similar across the two types of mycobacteria with some notable exceptions. We anticipate that this assay platform will lay the foundation for medicinal chemistry efforts to understand and improve uptake of both existing drugs and newly-discovered compounds into mycobacteria.


Asunto(s)
Mycobacterium tuberculosis , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Pared Celular/química , Pared Celular/metabolismo , Transporte Biológico , Antibacterianos/química , Antibacterianos/metabolismo
4.
Biochemistry ; 61(13): 1404-1414, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35687722

RESUMEN

A primary component of all known bacterial cell walls is the peptidoglycan (PG) layer, which is composed of repeating units of sugars connected to short and unusual peptides. The various steps within PG biosynthesis are targets of potent antibiotics as proper assembly of the PG is essential for cellular growth and survival. Synthetic mimics of PG have proven to be indispensable tools to study the bacterial cell structure, growth, and remodeling. Yet, a common component of PG, meso-diaminopimelic acid (m-DAP) at the third position of the stem peptide, remains challenging to access synthetically and is not commercially available. Here, we describe the synthesis and metabolic processing of a selenium-based bioisostere of m-DAP (selenolanthionine) and show that it is installed within the PG of live bacteria by the native cell wall crosslinking machinery in mycobacterial species. This PG probe has an orthogonal release mechanism that could be important for downstream proteomics studies. Finally, we describe a bead-based assay that is compatible with high-throughput screening of cell wall enzymes. We envision that this probe will supplement the current methods available for investigating PG crosslinking in m-DAP-containing organisms.


Asunto(s)
Mycobacterium , Selenio , Pared Celular/química , Ácido Diaminopimélico/metabolismo , Mycobacterium/metabolismo , Peptidoglicano/química
5.
Chembiochem ; 23(20): e202200412, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36018606

RESUMEN

The surfaces of most Gram-positive bacterial cells, including that of Staphylococcus aureus (S. aureus), are heavily decorated with proteins that coordinate cellular interactions with the extracellular space. In S. aureus, sortase A is the principal enzyme responsible for covalently anchoring proteins, which display the sorting signal LPXTG, onto the peptidoglycan (PG) matrix. Considerable efforts have been made to understand the role of this signal peptide in the sortase-mediated reaction. In contrast, much less is known about how the primary structure of the other substrate involved in the reaction (PG stem peptide) could impact sortase activity. To assess the sortase activity, a library of synthetic analogs of the stem peptide that mimic naturally existing variations found in the S. aureus PG primary sequence were evaluated. Using a combination of two unique assays, we showed that there is broad tolerability of substrate variations that are effectively processed by sortase A. While some of these stem peptide derivatives are naturally found in mature PG, they are not known to be present in the PG precursor, lipid II. These results suggest that sortase A could process both lipid II and mature PG as acyl-acceptor strands that might reside near the membrane, which has not been previously described.


Asunto(s)
Aminoaciltransferasas , Staphylococcus aureus , Peptidoglicano/metabolismo , Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Relación Estructura-Actividad , Señales de Clasificación de Proteína
6.
Chembiochem ; 23(8): e202100521, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35199442

RESUMEN

Current immunotherapeutics often work by directing components of the immune system to recognize biomarkers on the surface of cancer cells to generate an immune response. However, variable changes in biomarker distribution and expression can result in inconsistent patient response. The development of a more universal tumor-homing strategy has the potential to improve selectivity and extend therapy to cancers with decreased expression or absence of specific biomarkers. Here, we designed a bifunctional agent that exploits the inherent acidic microenvironment of most solid tumors to selectively graft the surface of cancer cells with a formyl peptide receptor ligand (FPRL). Our approach is based on the pH(Low) insertion peptide (pHLIP), a unique peptide that selectively targets tumors in vivo by anchoring to cancer cells in a pH-dependent manner. We establish that selectively remodeling cancer cells with a pHLIP-based FPRL activates formyl peptide receptors on recruited immune cells, potentially initiating an immune response towards tumors.


Asunto(s)
Neoplasias , Receptores de Formil Péptido , Línea Celular Tumoral , Factores Quimiotácticos , Humanos , Ligandos , Neoplasias/tratamiento farmacológico , Péptidos/metabolismo , Péptidos/farmacología , Receptores de Formil Péptido/metabolismo
7.
Bioconjug Chem ; 33(5): 767-772, 2022 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-35499914

RESUMEN

Bacterial cell walls represent one of the most prominent targets of antibacterial agents. These agents include natural products (e.g., vancomycin) and proteins stemming from the innate immune system (e.g., peptidoglycan-recognition proteins and lysostaphin). Among bacterial pathogens that infect humans, Staphylococcus aureus (S. aureus) continues to impose a tremendous healthcare burden across the globe. S. aureus has evolved countermeasures that can directly restrict the accessibility of innate immune proteins, effectively protecting itself from threats that target key cell well components. We recently described a novel assay that directly reports on the accessibility of molecules to the peptidoglycan layer within the bacterial cell wall of S. aureus. The assay relies on site-specific chemical remodeling of the peptidoglycan with a biorthogonal handle. Here, we disclose the application of our assay to a screen of a nonredundant transposon mutant library for susceptibility of the peptidoglycan layer with the goal of identifying genes that contribute to the control of cell surface accessibility. We discovered several genes that resulted in higher accessibility levels to the peptidoglycan layer and showed that these genes modulate sensitivity to lysostaphin. These results indicate that this assay platform can be leveraged to gain further insight into the biology of bacterial cell surfaces.


Asunto(s)
Lisostafina , Staphylococcus aureus , Antibacterianos/metabolismo , Antibacterianos/farmacología , Pared Celular/química , Humanos , Lisostafina/química , Lisostafina/metabolismo , Lisostafina/farmacología , Peptidoglicano/química , Vancomicina/metabolismo
8.
Anal Chem ; 91(20): 13251-13259, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31545025

RESUMEN

Infrared chemical microscopy through mechanical probing of light-matter interactions by atomic force microscopy (AFM) bypasses the diffraction limit. One increasingly popular technique is photoinduced force microscopy (PiFM), which utilizes the mechanical heterodyne signal detection between cantilever mechanical resonant oscillations and the photoinduced force from the light-matter interaction. So far, PiFM has been operated in only one heterodyne configuration. In this Article, we generalize heterodyne configurations of PiFM by introducing two new schemes: harmonic heterodyne detection and sequential heterodyne detection. In harmonic heterodyne detection, the laser repetition rate matches integer fractions of the difference between the two mechanical resonant modes of the AFM cantilever. The high harmonic of the beating from the photothermal expansion mixes with the AFM cantilever oscillation to provide the PiFM signal. In sequential heterodyne detection, the combination of the repetition rate of laser pulses and the polarization modulation frequency matches the difference between two AFM mechanical modes, leading to detectable PiFM signals. These two generalized heterodyne configurations for PiFM deliver new avenues for chemical imaging and broadband spectroscopy at ∼10 nm spatial resolution. They are suitable for a wide range of heterogeneous materials across various disciplines: from structured polymer film, to polaritonic boron nitride materials, to isolated bacterial peptidoglycan cell walls. The generalized heterodyne configurations introduce flexibility for the implementation of PiFM and the related tapping-mode AFM-IR and provide possibilities for an additional modulation channel in PiFM for targeted signal extraction with nanoscale spatial resolution.

9.
Bioconjug Chem ; 28(9): 2310-2315, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28737895

RESUMEN

Peptidoglycan (PG) scaffolds are critical components of bacterial cell walls. They counter internal turgor pressure to prevent lysis and protect against external insults. It was recently discovered that various types of bacteria release large quantities of PG building blocks (d-amino acids) into their surrounding medium. Contrarily, cultured bacteria were also found to incorporate d-amino acids (both natural and synthetic) from the medium directly into their PG scaffold. These two processes may potentially function, in concert, to metabolically remodel PG in live host organisms. However, demonstration that bacteria can decorate their cell surfaces with exogenous d-amino acids was limited to in vitro culture conditions. We present the first evidence that bacteria remodel their PG with exogenous d-amino acids in a live host animal. A tetrazine click partner was conjugated onto the side chain of a d-amino acid to capture incorporation into the bacterial PG scaffold using a complementary click-reactive fluorophore. Staphylococcus aureus infected Caenorhabditis elegans treated with exogenous d-amino acids readily revealed in vivo PG labeling. These results suggest that extracellular d-amino acids may provide pathogens with a mode of late-stage in vivo cell-surface remodeling.


Asunto(s)
Aminoácidos/metabolismo , Caenorhabditis elegans/microbiología , Pared Celular/metabolismo , Interacciones Huésped-Patógeno , Peptidoglicano/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Animales , Caenorhabditis elegans/fisiología , Modelos Animales de Enfermedad , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/veterinaria
10.
Angew Chem Int Ed Engl ; 56(30): 8839-8843, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28570012

RESUMEN

The surge in drug-resistant bacterial infections threatens to overburden healthcare systems worldwide. Bacterial cell walls are essential to bacteria, thus making them unique targets for the development of antibiotics. We describe a cellular reporter to directly monitor the phenotypic switch in drug-resistant bacteria with temporal resolution. Vancomycin-resistant enterococci (VRE) escape the bactericidal action of vancomycin by chemically modifying their cell-wall precursors. A synthetic cell-wall analogue was developed to hijack the biosynthetic rewiring of drug-resistant cells in response to antibiotics. Our study provides the first in vivo VanX reporter agent that responds to cell-wall alteration in drug-resistant bacteria. Cellular reporters that reveal mechanisms related to antibiotic resistance can potentially have a significant impact on the fundamental understanding of cellular adaption to antibiotics.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Microbiana/efectos de los fármacos , Enterococcus faecium/efectos de los fármacos , Vancomicina/farmacología , Antibacterianos/química , Pared Celular/efectos de los fármacos , Enterococcus faecium/citología , Citometría de Flujo , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Vancomicina/química
11.
J Biol Chem ; 290(51): 30540-50, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26499795

RESUMEN

Peptidoglycan is an essential and highly conserved mesh structure that surrounds bacterial cells. It plays a critical role in retaining a defined cell shape, and, in the case of pathogenic Gram-positive bacteria, it lies at the interface between bacterial cells and the host organism. Intriguingly, bacteria can metabolically incorporate unnatural D-amino acids into the peptidoglycan stem peptide directly from the surrounding medium, a process mediated by penicillin binding proteins (PBPs). Metabolic peptidoglycan remodeling via unnatural D-amino acids has provided unique insights into peptidoglycan biosynthesis of live bacteria and has also served as the basis of a synthetic immunology strategy with potential therapeutic implications. A striking feature of this process is the vast promiscuity displayed by PBPs in tolerating entirely unnatural side chains. However, the chemical space and physical features of this side chain promiscuity have not been determined systematically. In this report, we designed and synthesized a library of variants displaying diverse side chains to comprehensively establish the tolerability of unnatural D-amino acids by PBPs in both Gram-positive and Gram-negative organisms. In addition, nine Bacillus subtilis PBP-null mutants were evaluated with the goal of identifying a potential primary PBP responsible for unnatural D-amino acid incorporation and gaining insights into the temporal control of PBP activity. We empirically established the scope of physical parameters that govern the metabolic incorporation of unnatural D-amino acids into bacterial peptidoglycan.


Asunto(s)
Bacillus subtilis/química , Marcaje Isotópico/métodos , Proteínas de Unión a las Penicilinas/química , Peptidoglicano/química , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Mutación , Proteínas de Unión a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/metabolismo , Peptidoglicano/genética , Peptidoglicano/metabolismo
12.
Bioconjug Chem ; 27(12): 2850-2853, 2016 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-27801580

RESUMEN

A strategy has been devised for increasing the cellular selectivity of membrane-disrupting antibiotics based on the attachment of a facially amphiphilic sterol. Using Amphotericin B (AmB) as a prototype, covalent attachment of cholic acid bound to a series of α,ω-diamines has led to a dramatic reduction in hemolytic activity, a significant reduction in toxicity toward HEK293T cells, and significant retention of antifungal activity.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Anfotericina B/química , Anfotericina B/farmacología , Antibacterianos/efectos adversos , Antifúngicos/química , Antifúngicos/farmacología , Candida/efectos de los fármacos , Ácido Cólico/química , Células HEK293/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Relación Estructura-Actividad
13.
Angew Chem Int Ed Engl ; 55(29): 8401-4, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27225706

RESUMEN

ß-Lactams represent one of the most important classes of antibiotics discovered to date. These agents block Lipid II processing and cell wall biosynthesis through inactivation of penicillin-binding proteins (PBPs). PBPs enzymatically load cell wall building blocks from Lipid II carrier molecules onto the growing cell wall scaffold during growth and division. Lipid II, a bottleneck in cell wall biosynthesis, is the target of some of the most potent antibiotics in clinical use. Despite the immense therapeutic value of this biosynthetic pathway, the PBP-Lipid II association has not been established in live cells. To determine this key interaction, we designed an unnatural d-amino acid dipeptide that is metabolically incorporated into Lipid II molecules. By hijacking the peptidoglycan biosynthetic machinery, photoaffinity probes were installed in combination with click partners within Lipid II, thereby allowing, for the first time, demonstration of PBP interactions in vivo with Lipid II.


Asunto(s)
Proteínas de Unión a las Penicilinas/química , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Bacillus subtilis/citología , Bacillus subtilis/metabolismo , Pared Celular/metabolismo , Estructura Molecular , Proteínas de Unión a las Penicilinas/metabolismo , Uridina Difosfato Ácido N-Acetilmurámico/química , Uridina Difosfato Ácido N-Acetilmurámico/metabolismo
14.
Biopolymers ; 104(4): 351-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25653048

RESUMEN

During the past few decades there has been a rapid emergence of multidrug resistant bacteria afflicting human patients. At the same time, reduced output from pharmaceutical industry in this area precipitated a sharp decrease in the approval of new antibiotics. The combination of these factors potentially compromises the ability to effectively combat bacterial infections. While traditional drug discovery efforts continue in the pursuit of small molecule agents that disrupt bacterial growth, non-traditional efforts could serve to complement antimicrobial strategies. We recently demonstrated our ability to remodel the surface of bacterial cells using unnatural D-amino acids displaying the antigenic dinitrophenyl (DNP) handle. These immune stimulant D-amino acids derivatives were metabolically incorporated onto the peptidoglycan of bacteria via a promiscuous surface-anchored transpeptidase. The covalent modification of DNP moieties onto the peptidoglycan led to the anti-DNP antibody opsonization of the bacterial cell surface. Herein, we show that the amidation of the C-terminus to generate DNP-displaying D-amino carboxamide drastically improves antibody recruitment. Antibody opsonization using the D-amino carboxamide agent is observed at lower concentrations than the D-amino acid counterpart. In addition, the recruitment of endogenous antibodies in pooled human serum to the DNP-modified bacterial cell surface is demonstrated for the first time. We envision that the C-terminus amidation of DNP-conjugated D-amino acids could potentially facilitate translation of these results to in vivo animal disease models.


Asunto(s)
Anticuerpos/química , Bacillus subtilis/química , Dinitrofenoles/química , Peptidoglicano/química , Bacillus subtilis/metabolismo , Humanos , Peptidoglicano/metabolismo
15.
Angew Chem Int Ed Engl ; 54(21): 6158-62, 2015 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-25832713

RESUMEN

Bacterial peptidoglycan is a mesh-like network comprised of sugars and oligopeptides. Transpeptidases cross-link peptidoglycan oligopeptides to provide vital cell wall rigidity and structural support. It was recently discovered that the same transpeptidases catalyze the metabolic incorporation of exogenous D-amino acids onto bacterial cell surfaces with vast promiscuity for the side-chain identity. It is now shown that this enzymatic promiscuity is not exclusive to side chains, but that C-terminus variations can also be accommodated across a diverse range of bacteria. Atomic force microscopy analysis revealed that the incorporation of C-terminus amidated D-amino acids onto bacterial surfaces substantially reduced the cell wall stiffness. We exploited the promiscuity of bacterial transpeptidases to develop a novel assay for profiling different bacterial species.


Asunto(s)
Aminoácidos/metabolismo , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Peptidoglicano/metabolismo , Peptidil Transferasas/metabolismo , Aminoácidos/análisis , Bacillus/química , Bacillus/metabolismo , Bacterias/química , Secuencia de Carbohidratos , Pared Celular/química , Metaboloma , Datos de Secuencia Molecular , Peptidoglicano/análisis , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo
16.
ACS Infect Dis ; 10(5): 1602-1611, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38592927

RESUMEN

Antibiotic resistance is an alarming public health concern that affects millions of individuals across the globe each year. A major challenge in the development of effective antibiotics lies in their limited ability to permeate cells, noting that numerous susceptible antibiotic targets reside within the bacterial cytosol. Consequently, improving the cellular permeability is often a key consideration during antibiotic development, underscoring the need for reliable methods to assess the permeability of molecules across cellular membranes. Currently, methods used to measure permeability often fail to discriminate between the arrival within the cytoplasm and the overall association of molecules with the cell. Additionally, these techniques typically possess throughput limitations. In this work, we describe a luciferase-based assay designed for assessing the permeability of molecules in the cytosolic compartment of Gram-negative bacteria. Our findings demonstrate a robust system that can elucidate the kinetics of intracellular antibiotic accumulation in live bacterial cells in real time.


Asunto(s)
Antibacterianos , Citosol , Escherichia coli , Mediciones Luminiscentes , Antibacterianos/farmacología , Escherichia coli/metabolismo , Escherichia coli/genética , Citosol/metabolismo , Citosol/química , Pruebas de Sensibilidad Microbiana , Permeabilidad de la Membrana Celular
17.
Front Immunol ; 15: 1337973, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38665920

RESUMEN

Cytotoxic T lymphocytes are the primary effector immune cells responsible for protection against cancer, as they target peptide neoantigens presented through the major histocompatibility complex (MHC) on cancer cells, leading to cell death. Targeting peptide-MHC (pMHC) complex offers a promising strategy for immunotherapy due to their specificity and effectiveness against cancer. In this work, we exploit the acidic tumor micro-environment to selectively deliver antigenic peptides to cancer using pH(low) insertion peptides (pHLIP). We demonstrated the delivery of MHC binding peptides directly to the cytoplasm of melanoma cells resulted in the presentation of antigenic peptides on MHC, and activation of T cells. This work highlights the potential of pHLIP as a vehicle for the targeted delivery of antigenic peptides and its presentation via MHC-bound complexes on cancer cell surface for activation of T cells with implications for enhancing anti-cancer immunotherapy.


Asunto(s)
Presentación de Antígeno , Proteínas de la Membrana , Oligopéptidos , Humanos , Presentación de Antígeno/inmunología , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Inmunoterapia/métodos , Acidosis/inmunología , Activación de Linfocitos/inmunología , Microambiente Tumoral/inmunología , Ratones , Linfocitos T Citotóxicos/inmunología , Péptidos/inmunología , Concentración de Iones de Hidrógeno , Melanoma/inmunología , Melanoma/terapia
18.
Elife ; 132024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38639995

RESUMEN

Mechanisms by which Mycobacterium tuberculosis (Mtb) evades pathogen recognition receptor activation during infection may offer insights for the development of improved tuberculosis (TB) vaccines. Whilst Mtb elicits NOD-2 activation through host recognition of its peptidoglycan-derived muramyl dipeptide (MDP), it masks the endogenous NOD-1 ligand through amidation of glutamate at the second position in peptidoglycan side-chains. As the current BCG vaccine is derived from pathogenic mycobacteria, a similar situation prevails. To alleviate this masking ability and to potentially improve efficacy of the BCG vaccine, we used CRISPRi to inhibit expression of the essential enzyme pair, MurT-GatD, implicated in amidation of peptidoglycan side-chains. We demonstrate that depletion of these enzymes results in reduced growth, cell wall defects, increased susceptibility to antibiotics, altered spatial localization of new peptidoglycan and increased NOD-1 expression in macrophages. In cell culture experiments, training of a human monocyte cell line with this recombinant BCG yielded improved control of Mtb growth. In the murine model of TB infection, we demonstrate that depletion of MurT-GatD in BCG, which is expected to unmask the D-glutamate diaminopimelate (iE-DAP) NOD-1 ligand, yields superior prevention of TB disease compared to the standard BCG vaccine. In vitro and in vivo experiments in this study demonstrate the feasibility of gene regulation platforms such as CRISPRi to alter antigen presentation in BCG in a bespoke manner that tunes immunity towards more effective protection against TB disease.


Tuberculosis is the leading cause of death from an infectious disease worldwide, partially due to a lack of access to drug treatments in certain countries where the disease is common. The only available tuberculosis vaccine ­ known as the BCG vaccine ­ is useful for preventing cases in young children, but is ineffective in teenagers and adults. So, there is a need to develop new vaccines that offer better, and longer lasting, durable protection in people of all ages. During an infection, our immune system recognizes markers known as PAMPs on the surface of bacteria, viruses or other disease-causing pathogens. The recognition of PAMPs by the immune system enables the body to distinguish foreign invading organisms from its own cells and tissues, thus triggering a response that fights the infection. If the body encounters the infectious agent again in the future, the immune system is able to quickly recognize and eliminate it before it can cause disease. Vaccines protect us by mimicking the appearance of the pathogen to trigger the first immune response without causing the illness. The BCG vaccine contains live bacteria that are closely related to the bacterium responsible for tuberculosis called Mycobacterium tuberculosis. Both M. tuberculosis and the live bacteria used in the BCG vaccine are able to hide an important PAMP, known as the NOD-1 ligand, from the immune system, making it harder for the body to detect them. The NOD-1 ligand forms part of the bacterial cell wall and modifying the BCG bacterium so it cannot disguise this PAMP may lead to a new, more effective vaccine. To investigate this possibility, Shaku et al. used a gene editing approach to develop a modified version of the BCG bacterium which is unable to hide its NOD-1 ligand when treated with a specific drug. Immune cells trained with the modified BCG vaccine were more effective at controlling the growth of M. tuberculosis than macrophages trained using the original vaccine. Furthermore, mice vaccinated with the modified BCG vaccine were better able to limit M. tuberculosis growth in their lungs than mice that had received the original vaccine. These findings offer a new candidate vaccine in the fight against tuberculosis. Further studies will be needed to modify the vaccine for use in humans. More broadly, this work demonstrates that gene editing can be used to expose a specific PAMP present in a live vaccine. This may help develop more effective vaccines for other diseases in the future.


Asunto(s)
Vacuna BCG , Mycobacterium tuberculosis , Peptidoglicano , Tuberculosis , Animales , Peptidoglicano/metabolismo , Ratones , Vacuna BCG/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Tuberculosis/microbiología , Humanos , Ratones Endogámicos C57BL , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/metabolismo , Femenino , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Modelos Animales de Enfermedad , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
19.
mBio ; 15(5): e0011924, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38587424

RESUMEN

Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophilic influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5'-monophosphate-N-acetylneuraminic acid, which is scavenged from the host using LOS sialyltransferase (Lst) since Gc cannot make its sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress the oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea. IMPORTANCE: Neisseria gonorrhoeae, the bacterium that causes gonorrhea, is an urgent global health concern due to increasing infection rates, widespread antibiotic resistance, and its ability to thwart protective immune responses. The mechanisms by which Gc subverts protective immune responses remain poorly characterized. One way N. gonorrhoeae evades human immunity is by adding sialic acid that is scavenged from the host onto its lipooligosaccharide, using the sialyltransferase Lst. Here, we found that sialylation enhances N. gonorrhoeae survival from neutrophil assault and inhibits neutrophil activation, independently of the complement system. Our results implicate bacterial binding of sialic acid-binding lectins (Siglecs) on the neutrophil surface, which dampens neutrophil antimicrobial responses. This work identifies a new role for sialylation in protecting N. gonorrhoeae from cellular innate immunity, which can be targeted to enhance the human immune response in gonorrhea.


Asunto(s)
Gonorrea , Ácido N-Acetilneuramínico , Neisseria gonorrhoeae , Activación Neutrófila , Neutrófilos , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Humanos , Ácido N-Acetilneuramínico/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética , Gonorrea/inmunología , Gonorrea/microbiología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Lipopolisacáridos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Estallido Respiratorio , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune
20.
bioRxiv ; 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38293026

RESUMEN

Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophil influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5'-monophosphate-N-acetylneuraminic acid (CMP-NANA) scavenged from the host using LOS sialyltransferase (Lst), since Gc cannot make its own sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda