Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 7(1): 572, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750133

RESUMEN

Long-chain fatty acids with antimicrobial properties are abundant on the skin and mucosal surfaces, where they are essential to restrict the proliferation of opportunistic pathogens such as Staphylococcus aureus. These antimicrobial fatty acids (AFAs) elicit bacterial adaptation strategies, which have yet to be fully elucidated. Characterizing the pervasive mechanisms used by S. aureus to resist AFAs could open new avenues to prevent pathogen colonization. Here, we identify the S. aureus lipase Lip2 as a novel resistance factor against AFAs. Lip2 detoxifies AFAs via esterification with cholesterol. This is reminiscent of the activity of the fatty acid-modifying enzyme (FAME), whose identity has remained elusive for over three decades. In vitro, Lip2-dependent AFA-detoxification was apparent during planktonic growth and biofilm formation. Our genomic analysis revealed that prophage-mediated inactivation of Lip2 was rare in blood, nose, and skin strains, suggesting a particularly important role of Lip2 for host - microbe interactions. In a mouse model of S. aureus skin colonization, bacteria were protected from sapienic acid (a human-specific AFA) in a cholesterol- and lipase-dependent manner. These results suggest Lip2 is the long-sought FAME that exquisitely manipulates environmental lipids to promote bacterial growth in otherwise inhospitable niches.


Asunto(s)
Ácidos Grasos , Lipasa , Staphylococcus aureus , Staphylococcus aureus/metabolismo , Ácidos Grasos/metabolismo , Animales , Ratones , Lipasa/metabolismo , Lipasa/genética , Humanos , Infecciones Estafilocócicas/microbiología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Femenino , Infecciones Cutáneas Estafilocócicas/microbiología
2.
Trends Microbiol ; 31(7): 723-734, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36822953

RESUMEN

The corneocyte layers forming the upper surface of mammalian skin are embedded in a lamellar-membrane matrix which repels harmful molecules while retaining solutes from subcutaneous tissues. Only certain bacterial and fungal taxa colonize skin surfaces. They have ways to use epidermal lipids as nutrients while resisting antimicrobial fatty acids. Skin microorganisms release lipophilic microbe-associated molecular pattern (MAMP) molecules which are largely retained by the epidermal lipid barrier. Skin barrier defects, as in atopic dermatitis, impair lamellar-membrane integrity, resulting in altered skin microbiomes, which then include the pathogen Staphylococcus aureus. The resulting increased penetration of MAMPs and toxins promotes skin inflammation. Elucidating how microorganisms manipulate the epidermal lipid barrier will be key for better ways of preventing inflammatory skin disorders.


Asunto(s)
Dermatitis Atópica , Microbiota , Animales , Piel , Epidermis , Dermatitis Atópica/microbiología , Ácidos Grasos , Mamíferos
3.
Trends Microbiol ; 30(3): 241-253, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34218980

RESUMEN

Fatty acids (FAs) are potent antimicrobials which hold great promise as viable alternatives or complements to conventional antibiotics. Intriguingly, bacteria are well equipped to use environmental FAs as energy sources and/or building blocks for their membrane lipids. Furthermore, these microbes display a wide array of mechanisms to prevent or mitigate FA toxicity. In this review we discuss strategies that bacteria use to thrive despite extensive exposure to host-derived antimicrobial FAs. We also highlight the altered response of these FA-adapted bacteria to antibiotics. Given the ubiquitous nature of FAs in various host environments, deciphering bacterial adaptation strategies to FAs is of prime importance. This knowledge may pave the way for a rational design of FA-based combination therapies with antibiotics.


Asunto(s)
Bacterias , Ácidos Grasos , Adaptación Fisiológica , Antibacterianos/farmacología , Antibacterianos/uso terapéutico
4.
mSphere ; 5(5)2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32999082

RESUMEN

Staphylococcus aureus is a major pathogen, which colonizes one in three otherwise healthy humans. This significant spread of S. aureus is largely due to its ability to circumvent innate immune responses, including antimicrobial fatty acids (AFAs) on the skin and in nasal secretions. In response to AFAs, S. aureus swiftly induces resistance mechanisms, which have yet to be completely elucidated. Here, we identify membrane vesicle (MV) release as a resistance strategy used by S. aureus to sequester host-specific AFAs. MVs protect S. aureus against a wide array of AFAs. Strikingly, beside MV production, S. aureus modulates MV composition upon exposure to AFAs. MVs purified from bacteria grown in the presence of linoleic acid display a distinct protein content and are enriched in lipoproteins, which strongly activate Toll-like receptor 2 (TLR2). Cumulatively, our findings reveal the protective capacities of MVs against AFAs, which are counteracted by an increased TLR2-mediated innate immune response.IMPORTANCE The nares of one in three humans are colonized by Staphylococcus aureus In these environments, and arguably on all mucosal surfaces, bacteria encounter fatty acids with antimicrobial properties. Our study uncovers that S. aureus releases membrane vesicles (MVs) that act as decoys to protect the bacterium against antimicrobial fatty acids (AFAs). The AFA-neutralizing effects of MVs were neither strain specific nor restricted to one particular AFA. Hence, MVs may represent "public goods" playing an overlooked role in shaping bacterial communities in AFA-rich environments such as the skin and nose. Intriguingly, in addition to MV biogenesis, S. aureus modulates MV composition in response to exposure to AFAs, including an increased release of lipoproteins. These MVs strongly stimulate the innate immunity via Toll-like receptor 2 (TLR2). TLR2-mediated inflammation, which helps to fight infections, may exacerbate inflammatory disorders like atopic dermatitis. Our study highlights intricate immune responses preventing infections from colonizing bacteria.


Asunto(s)
Vesículas Extracelulares/inmunología , Ácidos Grasos/inmunología , Inmunidad Innata , Staphylococcus aureus/inmunología , Vesículas Extracelulares/química , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Ácido Linoleico , Proteómica , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad
5.
Sci Rep ; 10(1): 14838, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32908165

RESUMEN

The Staphylococcus aureus type VII secretion system (T7SS) exports several proteins that are pivotal for bacterial virulence. The mechanisms underlying T7SS-mediated staphylococcal survival during infection nevertheless remain unclear. Here we report that S. aureus lacking T7SS components are more susceptible to host-derived antimicrobial fatty acids. Unsaturated fatty acids such as linoleic acid (LA) elicited an increased inhibition of S. aureus mutants lacking T7SS effectors EsxC, EsxA and EsxB, or the membrane-bound ATPase EssC, compared to the wild-type (WT). T7SS mutants generated in different S. aureus strain backgrounds also displayed an increased sensitivity to LA. Analysis of bacterial membrane lipid profiles revealed that the esxC mutant was less able to incorporate LA into its membrane phospholipids. Although the ability to bind labelled LA did not differ between the WT and mutant strains, LA induced more cell membrane damage in the T7SS mutants compared to the WT. Furthermore, proteomic analyses of WT and mutant cell fractions revealed that, in addition to compromising membranes, T7SS defects induce oxidative stress and hamper their response to LA challenge. Thus, our findings indicate that T7SS contribute to maintaining S. aureus membrane integrity and homeostasis when bacteria encounter antimicrobial fatty acids.


Asunto(s)
Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Ácidos Grasos/metabolismo , Staphylococcus aureus/metabolismo , Sistemas de Secreción Tipo VII/metabolismo , Regulación Bacteriana de la Expresión Génica
6.
Biomaterials ; 217: 119249, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31279102

RESUMEN

Intracellular persistence of bacteria represents a clinical challenge as bacteria can thrive in an environment protected from antibiotics and immune responses. Novel targeting strategies are critical in tackling antibiotic resistant infections. Synthetic antimicrobial peptides (SAMPs) are interesting candidates as they exhibit a very high antimicrobial activity. We first compared the activity of a library of ammonium and guanidinium polymers with different sequences (statistical, tetrablock and diblock) synthesized by RAFT polymerization against methicillin-resistant S. aureus (MRSA) and methicillin-sensitive strains (MSSA). As the guanidinium SAMPs were the most potent, they were used to treat intracellular S. aureus in keratinocytes. The diblock structure was the most active, reducing the amount of intracellular MSSA and MRSA by two-fold. We present here a potential treatment for intracellular, multi-drug resistant bacteria, using a simple and scalable strategy.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Guanidina/química , Guanidina/farmacología , Espacio Intracelular/microbiología , Polímeros/química , Polímeros/farmacología , Staphylococcus aureus/efectos de los fármacos , Células A549 , Compuestos de Amonio/farmacología , Animales , Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Fluorescencia , Guanidina/síntesis química , Humanos , Pruebas de Sensibilidad Microbiana , Polímeros/síntesis química , Ovinos , Relación Estructura-Actividad , Pruebas de Toxicidad
8.
Nat Microbiol ; 2: 16188, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27748756

RESUMEN

Helicobacter pylori (Hp) strains that carry the cag type IV secretion system (cag-T4SS) to inject the cytotoxin-associated antigen A (CagA) into host cells are associated with peptic ulcer disease and gastric adenocarcinoma. CagA translocation by Hp is mediated by ß1 integrin interaction of the cag-T4SS. However, other cellular receptors or bacterial outer membrane adhesins essential for this process are unknown. Here, we identify the HopQ protein as a genuine Hp adhesin, exploiting defined members of the carcinoembryonic antigen-related cell adhesion molecule family (CEACAMs) as host cell receptors. HopQ binds the amino-terminal IgV-like domain of human CEACAM1, CEACAM3, CEACAM5 or CEACAM6 proteins, thereby enabling translocation of the major pathogenicity factor CagA into host cells. The HopQ-CEACAM interaction is characterized by a remarkably high affinity (KD from 23 to 268 nM), which is independent of CEACAM glycosylation, identifying CEACAMs as bona fide protein receptors for Hp. Our data suggest that the HopQ-CEACAM interaction contributes to gastric colonization or Hp-induced pathologies, although the precise role and functional consequences of this interaction in vivo remain to be determined.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Antígenos Bacterianos/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno , Transporte de Proteínas , Línea Celular , Humanos , Unión Proteica
9.
PLoS Pathog ; 12(5): e1005608, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27171273

RESUMEN

Attachment to the host mucosa is a key step in bacterial pathogenesis. On the apical surface of epithelial cells, members of the human carcinoembryonic antigen (CEA) family are abundant glycoproteins involved in cell-cell adhesion and modulation of cell signaling. Interestingly, several gram-negative bacterial pathogens target these receptors by specialized adhesins. The prototype of a CEACAM-binding pathogen, Neisseria gonorrhoeae, utilizes colony opacity associated (Opa) proteins to engage CEA, as well as the CEA-related cell adhesion molecules CEACAM1 and CEACAM6 on human epithelial cells. By heterologous expression of neisserial Opa proteins in non-pathogenic E. coli we find that the Opa protein-CEA interaction is sufficient to alter gene expression, to increase integrin activity and to promote matrix adhesion of infected cervical carcinoma cells and immortalized vaginal epithelial cells in vitro. These CEA-triggered events translate in suppression of exfoliation and improved colonization of the urogenital tract by Opa protein-expressing E. coli in CEA-transgenic compared to wildtype mice. Interestingly, uropathogenic E. coli expressing an unrelated CEACAM-binding protein of the Afa/Dr adhesin family recapitulate the in vitro and in vivo phenotype. In contrast, an isogenic strain lacking the CEACAM-binding adhesin shows reduced colonization and does not suppress epithelial exfoliation. These results demonstrate that engagement of human CEACAMs by distinct bacterial adhesins is sufficient to blunt exfoliation and to promote host infection. Our findings provide novel insight into mucosal colonization by a common UPEC pathotype and help to explain why human CEACAMs are a preferred epithelial target structure for diverse gram-negative bacteria to establish a foothold on the human mucosa.


Asunto(s)
Adhesión Bacteriana/fisiología , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Membrana Mucosa/microbiología , Sistema Urogenital/microbiología , Escherichia coli Uropatógena/patogenicidad , Animales , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Membrana Mucosa/metabolismo , Sistema Urogenital/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA