Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nature ; 535(7613): 511-6, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27466123

RESUMEN

The vast majority of systemic bacterial infections are caused by facultative, often antibiotic-resistant, pathogens colonizing human body surfaces. Nasal carriage of Staphylococcus aureus predisposes to invasive infection, but the mechanisms that permit or interfere with pathogen colonization are largely unknown. Whereas soil microbes are known to compete by production of antibiotics, such processes have rarely been reported for human microbiota. We show that nasal Staphylococcus lugdunensis strains produce lugdunin, a novel thiazolidine-containing cyclic peptide antibiotic that prohibits colonization by S. aureus, and a rare example of a non-ribosomally synthesized bioactive compound from human-associated bacteria. Lugdunin is bactericidal against major pathogens, effective in animal models, and not prone to causing development of resistance in S. aureus. Notably, human nasal colonization by S. lugdunensis was associated with a significantly reduced S. aureus carriage rate, suggesting that lugdunin or lugdunin-producing commensal bacteria could be valuable for preventing staphylococcal infections. Moreover, human microbiota should be considered as a source for new antibiotics.


Asunto(s)
Antibacterianos/metabolismo , Péptidos Cíclicos/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus lugdunensis/metabolismo , Simbiosis , Tiazolidinas/metabolismo , Animales , Antibacterianos/biosíntesis , Portador Sano/microbiología , Modelos Animales de Enfermedad , Farmacorresistencia Microbiana , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Microbiota/fisiología , Nariz/microbiología , Sigmodontinae , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/patogenicidad
2.
EMBO Rep ; 19(12)2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30337494

RESUMEN

Chitin is the second most abundant polysaccharide in nature and linked to fungal infection and asthma. However, bona fide immune receptors directly binding chitin and signaling immune activation and inflammation have not been clearly identified because polymeric crude chitin with unknown purity and molecular composition has been used. By using defined chitin (N-acetyl-glucosamine) oligomers, we here identify six-subunit-long chitin chains as the smallest immunologically active motif and the innate immune receptor Toll-like receptor (TLR2) as a primary fungal chitin sensor on human and murine immune cells. Chitin oligomers directly bind TLR2 with nanomolar affinity, and this fungal TLR2 ligand shows overlapping and distinct signaling outcomes compared to known mycobacterial TLR2 ligands. Unexpectedly, chitin oligomers composed of five or less subunits are inactive, hinting to a size-dependent system of immuno-modulation that appears conserved in plants and humans. Since blocking of the chitin-TLR2 interaction effectively prevents chitin-mediated inflammation in vitro and in vivo, our study highlights the chitin-TLR2 interaction as a potential target for developing novel therapies in chitin-related pathologies and fungal disease.


Asunto(s)
Quitina/química , Quitina/metabolismo , Hongos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Receptor Toll-Like 2/metabolismo , Animales , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Quitinasas/metabolismo , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Factores Inmunológicos/farmacología , Ligandos , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células THP-1 , Receptor Toll-Like 1/agonistas , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 2/química , Zimosan/metabolismo
3.
Angew Chem Int Ed Engl ; 58(27): 9234-9238, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31059155

RESUMEN

Lugdunin, a novel thiazolidine cyclopeptide, exhibits micromolar activity against methicillin-resistant Staphylococcus aureus (MRSA). For structure-activity relationship (SAR) studies, synthetic analogues obtained from alanine and stereo scanning as well as peptides with modified thiazolidine rings were tested for antimicrobial activity. The thiazolidine ring and the alternating d- and l-amino acid backbone are essential. Notably, the non-natural enantiomer displays equal activity, thus indicating the absence of a chiral target. The antibacterial activity strongly correlates with dissipation of the membrane potential in S. aureus. Lugdunin equalizes pH gradients in artificial membrane vesicles, thereby maintaining membrane integrity, which demonstrates that proton translocation is the mode of action (MoA). The incorporation of extra tryptophan or propargyl moieties further expands the diversity of this class of thiazolidine cyclopeptides.


Asunto(s)
Antiinfecciosos/síntesis química , Péptidos Cíclicos/química , Tiazolidinas/química , Alanina/química , Secuencia de Aminoácidos , Antiinfecciosos/química , Antiinfecciosos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/farmacología , Protones , Estereoisomerismo , Relación Estructura-Actividad , Tiazolidinas/síntesis química , Tiazolidinas/farmacología
5.
mBio ; 15(9): e0057824, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39133006

RESUMEN

Lugdunin is a microbiome-derived antibacterial agent with good activity against Gram-positive pathogens in vitro and in animal models of nose colonization and skin infection. We have previously shown that lugdunin depletes bacterial energy resources by dissipating the membrane potential of Staphylococcus aureus. Here, we explored the mechanism of action of lugdunin in more detail and show that lugdunin quickly depolarizes cytoplasmic membranes of different bacterial species and acidifies the cytoplasm of S. aureus within minutes due to protonophore activity. Varying the salt species and concentrations in buffers revealed that not only protons are transported, and we demonstrate the binding of the monovalent cations K+, Na+, and Li+ to lugdunin. By comparing known ionophores with various ion transport mechanisms, we conclude that the ion selectivity of lugdunin largely resembles that of 15-mer linear peptide gramicidin A. Direct interference with the main bacterial metabolic pathways including DNA, RNA, protein, and cell wall biosyntheses can be excluded. The previously observed synergism of lugdunin with dermcidin-derived peptides such as DCD-1 in killing S. aureus is mechanistically based on potentiated membrane depolarization. We also found that lugdunin was active against certain eukaryotic cells, however strongly depending on the cell line and growth conditions. While adherent lung epithelial cell lines were almost unaffected, more sensitive cells showed dissipation of the mitochondrial membrane potential. Lugdunin seems specifically adapted to its natural environment in the respiratory tract. The ionophore mechanism is refractory to resistance development and benefits from synergy with host-derived antimicrobial peptides. IMPORTANCE: The vast majority of antimicrobial peptides produced by members of the microbiome target the bacterial cell envelope by many different mechanisms. These compounds and their producers have evolved side-by-side with their host and were constantly challenged by the host's immune system. These molecules are optimized to be well tolerated at their physiological site of production, and their modes of action have proven efficient in vivo. Imbalancing the cellular ion homeostasis is a prominent mechanism among antibacterial natural products. For instance, over 120 naturally occurring polyether ionophores are known to date, and antimicrobial peptides with ionophore activity have also been detected in microbiomes. In this study, we elucidated the mechanism underlying the membrane potential-dissipating activity of the thiazolidine-containing cycloheptapeptide lugdunin, the first member of the fibupeptides discovered in a commensal bacterium from the human nose, which is a promising future probiotic candidate that is not prone to resistance development.


Asunto(s)
Antibacterianos , Ionóforos , Microbiota , Staphylococcus aureus , Humanos , Antibacterianos/farmacología , Cationes/farmacología , Cationes/metabolismo , Sinergismo Farmacológico , Ionóforos/farmacología , Lipopéptidos/farmacología , Lipopéptidos/metabolismo , Pruebas de Sensibilidad Microbiana , Microbiota/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/metabolismo
6.
Nat Microbiol ; 9(1): 200-213, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38110697

RESUMEN

Antagonistic bacterial interactions often rely on antimicrobial bacteriocins, which attack only a narrow range of target bacteria. However, antimicrobials with broader activity may be advantageous. Here we identify an antimicrobial called epifadin, which is produced by nasal Staphylococcus epidermidis IVK83. It has an unprecedented architecture consisting of a non-ribosomally synthesized peptide, a polyketide component and a terminal modified amino acid moiety. Epifadin combines a wide antimicrobial target spectrum with a short life span of only a few hours. It is highly unstable under in vivo-like conditions, potentially as a means to limit collateral damage of bacterial mutualists. However, Staphylococcus aureus is eliminated by epifadin-producing S. epidermidis during co-cultivation in vitro and in vivo, indicating that epifadin-producing commensals could help prevent nasal S. aureus carriage. These insights into a microbiome-derived, previously unknown antimicrobial compound class suggest that limiting the half-life of an antimicrobial may help to balance its beneficial and detrimental activities.


Asunto(s)
Antiinfecciosos , Infecciones Estafilocócicas , Humanos , Staphylococcus aureus , Péptidos Antimicrobianos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/metabolismo
7.
J Med Chem ; 64(7): 4034-4058, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33779184

RESUMEN

A new solid-phase peptide synthesis and bioprofiling of the antimicrobial activity of lugdunin, a fibupeptide, enable a comprehensive structure-activity relationship (SAR) study (MRSA Staphylococcus aureus). Distinct lugdunin analogues with variation of the three important amino acids Val2, Trp3, and Leu4 are readily available based on the established high-output synthesis. This efficient synthesis concept takes advantage of the presynthesized thiazolidine building block. To gain further knowledge of SAR, d-Val2, and d-Leu4 were replaced with aliphatic amino acids. For l-Trp3 derivatization, a set of non-natural aromatic amino acids with manifold substitution and annulation patterns precisely shows structural imperatives, starting from the exchange of d-Val6 → d-Trp6 with a 2-fold improved biological activity. d-Trp6-lugdunin analogues with additional variation of d-Val2 and d-Leu4 residues were designed and synthesized followed by antimicrobial profiling. For the first time, these SAR studies deliver valuable information on the tolerance of other amino acids to d-Val2, l-Trp3, and d-Leu4 in the sequence of lugdunin.


Asunto(s)
Antibacterianos/farmacología , Péptidos Cíclicos/farmacología , Tiazolidinas/farmacología , Antibacterianos/síntesis química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Péptidos Cíclicos/síntesis química , Relación Estructura-Actividad , Tiazolidinas/síntesis química
8.
J Exp Med ; 218(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34554188

RESUMEN

Activity of the NLRP3 inflammasome, a critical mediator of inflammation, is controlled by accessory proteins, posttranslational modifications, cellular localization, and oligomerization. How these factors relate is unclear. We show that a well-established drug target, Bruton's tyrosine kinase (BTK), affects several levels of NLRP3 regulation. BTK directly interacts with NLRP3 in immune cells and phosphorylates four conserved tyrosine residues upon inflammasome activation, in vitro and in vivo. Furthermore, BTK promotes NLRP3 relocalization, oligomerization, ASC polymerization, and full inflammasome assembly, probably by charge neutralization, upon modification of a polybasic linker known to direct NLRP3 Golgi association and inflammasome nucleation. As NLRP3 tyrosine modification by BTK also positively regulates IL-1ß release, we propose BTK as a multifunctional positive regulator of NLRP3 regulation and BTK phosphorylation of NLRP3 as a novel and therapeutically tractable step in the control of inflammation.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Tirosina/metabolismo , Animales , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Nat Commun ; 10(1): 2730, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227691

RESUMEN

Recently our groups discovered lugdunin, a new cyclic peptide antibiotic that inhibits Staphylococcus aureus epithelial colonization in humans and rodents. In this work, we analyzed its immuno-modulatory and antimicrobial potential as a single agent or in combination with other microbiota- or host-derived factors. We show that pretreatment of primary human keratinocytes or mouse skin with lugdunin in combination with microbiota-derived factors results in a significant reduction of S. aureus colonization. Moreover, lugdunin increases expression and release of LL-37 and CXCL8/MIP-2 in human keratinocytes and mouse skin, and results in the recruitment of monocytes and neutrophils in vivo, both by a TLR/MyD88-dependent mechanism. Interestingly, S. aureus elimination by lugdunin is additionally achieved by synergistic antimicrobial activity with LL-37 and dermcidin-derived peptides. In summary, our results indicate that lugdunin provides multi-level protection against S. aureus and may thus become a promising treatment option for S. aureus skin infections in the future.


Asunto(s)
Antibacterianos/farmacología , Inmunidad Innata/efectos de los fármacos , Péptidos Cíclicos/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Tiazolidinas/farmacología , Animales , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/inmunología , Queratinocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/efectos de los fármacos , Microbiota/inmunología , Péptidos/inmunología , Péptidos Cíclicos/uso terapéutico , Cultivo Primario de Células , Piel/efectos de los fármacos , Piel/inmunología , Piel/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Tiazolidinas/uso terapéutico , Catelicidinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA