Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 19(10): 1100-1111, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30250184

RESUMEN

Females have an overall advantage over males in resisting Gram-negative bacteremias, thus hinting at sexual dimorphism of immunity during infections. Here, through intravital microscopy, we observed a sex-biased difference in the capture of blood-borne bacteria by liver macrophages, a process that is critical for the clearance of systemic infections. Complement opsonization was indispensable for the capture of enteropathogenic Escherichia coli (EPEC) in male mice; however, a faster complement component 3-independent process involving abundant preexisting antibodies to EPEC was detected in female mice. These antibodies were elicited predominantly in female mice at puberty in response to estrogen regardless of microbiota-colonization conditions. Estrogen-driven antibodies were maternally transferrable to offspring and conferred protection during infancy. These antibodies were conserved in humans and recognized specialized oligosaccharides integrated into the bacterial lipopolysaccharide and capsule. Thus, an estrogen-driven, innate antibody-mediated immunological strategy conferred protection to females and their offspring.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Infecciones por Escherichia coli/inmunología , Inmunidad Innata/inmunología , Caracteres Sexuales , Animales , Escherichia coli Enteropatógena , Estrógenos/inmunología , Femenino , Humanos , Lactante , Macrófagos del Hígado/inmunología , Masculino , Intercambio Materno-Fetal/inmunología , Ratones , Embarazo
2.
Nature ; 609(7925): 166-173, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35948634

RESUMEN

During infection, inflammatory monocytes are thought to be key for bacterial eradication, but this is hard to reconcile with the large numbers of neutrophils that are recruited for each monocyte that migrates to the afflicted tissue, and the much more robust microbicidal functions of the neutrophils. However, unlike neutrophils, monocytes have the capacity to convert to situationally specific macrophages that may have critical functions beyond infection control1,2. Here, using a foreign body coated with Staphylococcus aureus and imaging over time from cutaneous infection to wound resolution, we show that monocytes and neutrophils are recruited in similar numbers with low-dose infection but not with high-dose infection, and form a localization pattern in which monocytes surround the infection site, whereas neutrophils infiltrate it. Monocytes did not contribute to bacterial clearance but converted to macrophages that persisted for weeks after infection, regulating hypodermal adipocyte expansion and production of the adipokine hormone leptin. In infected monocyte-deficient mice there was increased persistent hypodermis thickening and an elevated leptin level, which drove overgrowth of dysfunctional blood vasculature and delayed healing, with a thickened scar. Ghrelin, which opposes leptin function3, was produced locally by monocytes, and reduced vascular overgrowth and improved healing post-infection. In sum, we find that monocytes function as a cellular rheostat by regulating leptin levels and revascularization during wound repair.


Asunto(s)
Leptina , Monocitos , Neovascularización Fisiológica , Infecciones Estafilocócicas , Staphylococcus aureus , Cicatrización de Heridas , Adipocitos/citología , Adipocitos/metabolismo , Animales , Cicatriz , Ghrelina/metabolismo , Leptina/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Monocitos/citología , Monocitos/metabolismo , Neutrófilos/citología , Neutrófilos/inmunología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/fisiología
3.
PLoS Pathog ; 17(9): e1009880, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34529737

RESUMEN

Staphylococcus aureus is a human commensal organism and opportunist pathogen, causing potentially fatal disease. The presence of non-pathogenic microflora or their components, at the point of infection, dramatically increases S. aureus pathogenicity, a process termed augmentation. Augmentation is associated with macrophage interaction but by a hitherto unknown mechanism. Here, we demonstrate a breadth of cross-kingdom microorganisms can augment S. aureus disease and that pathogenesis of Enterococcus faecalis can also be augmented. Co-administration of augmenting material also forms an efficacious vaccine model for S. aureus. In vitro, augmenting material protects S. aureus directly from reactive oxygen species (ROS), which correlates with in vivo studies where augmentation restores full virulence to the ROS-susceptible, attenuated mutant katA ahpC. At the cellular level, augmentation increases bacterial survival within macrophages via amelioration of ROS, leading to proliferation and escape. We have defined the molecular basis for augmentation that represents an important aspect of the initiation of infection.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Simbiosis/fisiología , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Sepsis/inmunología , Sepsis/microbiología , Infecciones Estafilocócicas/inmunología , Pez Cebra
4.
Hepatology ; 73(5): 1967-1984, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32761929

RESUMEN

BACKGROUND AND AIMS: Kupffer cells (KCs) are the resident intravascular phagocyte population of the liver and critical to the capture and killing of bacteria. Calcineurin/nuclear factor of activated T cells (NFAT) inhibitors (CNIs) such as tacrolimus are used to prevent rejection in solid organ transplant recipients. Although their effect on lymphocytes has been studied extensively, there are limited experimental data about if and how CNIs shape innate immunity, and whether this contributes to the higher rates of infection observed in patients taking CNIs. APPROACH AND RESULTS: Here, we investigated the impact of tacrolimus treatment on innate immunity and, more specifically, on the capability of Kupffer cells (KCs) to fight infections. Retrospective analysis of data of >2,700 liver transplant recipients showed that taking calcineurin inhibitors such as tacrolimus significantly increased the likelihood of Staphylococcus aureus infection. Using a mouse model of acute methicillin-resistant S. aureus (MRSA) bacteremia, most bacteria were sequestered in the liver and we found that bacteria were more likely to disseminate and kill the host in tacrolimus-treated mice. Using imaging, we unveiled the mechanism underlying this observation: the reduced capability of KCs to capture, phagocytose, and destroy bacteria in tacrolimus-treated animals. Furthermore, in a gene expression analysis of infected KCs, the triggering receptor expressed on myeloid cells 1 (TREM1) pathway was the one with the most significant down-regulation after tacrolimus treatment. TREM1 inhibition likewise inhibited KC bacteria capture. TREM1 levels on neutrophils as well as the overall neutrophil response after infection were unaffected by tacrolimus treatment. CONCLUSIONS: Our results indicate that tacrolimus treatment has a significant impact directly on KCs and on TREM1, thereby compromising their capacity to fend off infections.


Asunto(s)
Bacteriemia/etiología , Inmunosupresores/efectos adversos , Macrófagos del Hígado/efectos de los fármacos , Trasplante de Órganos/efectos adversos , Infecciones Estafilocócicas/etiología , Tacrolimus/efectos adversos , Animales , Femenino , Citometría de Flujo , Humanos , Inmunosupresores/uso terapéutico , Macrófagos del Hígado/fisiología , Masculino , Staphylococcus aureus Resistente a Meticilina , Ratones , Persona de Mediana Edad , Trasplante de Órganos/métodos , Fagocitosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Estudios Retrospectivos , Tacrolimus/uso terapéutico
5.
Hepatology ; 74(1): 296-311, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33219516

RESUMEN

BACKGROUND AND AIMS: Bacterial infections are common and severe in cirrhosis, but their pathogenesis is poorly understood. Dysfunction of liver macrophages may play a role, but information about their function in cirrhosis is limited. Our aims were to investigate the specific profile and function of liver macrophages in cirrhosis and their contribution to infections. Macrophages from human cirrhotic livers were characterized phenotypically by transcriptome analysis and flow cytometry; function was assessed in vivo by single photon emission computerized tomography in patients with cirrhosis. Serum levels of specific proteins and expression in peripheral monocytes were determined by ELISA and flow cytometry. In vivo phagocytic activity of liver macrophages was measured by spinning disk intravital microscopy in a mouse model of chronic liver injury. APPROACH AND RESULTS: Liver macrophages from patients with cirrhosis overexpressed proteins related to immune exhaustion, such as programmed death ligand 1 (PD-L1), macrophage receptor with collagenous structure (MARCO), and CD163. In vivo phagocytic activity of liver macrophages in patients with cirrhosis was markedly impaired. Monocytes from patients with cirrhosis showed overexpression of PD-L1 that paralleled disease severity, correlated with its serum levels, and was associated with increased risk of infections. Blockade of PD-L1 with anti-PD-L1 antibody caused a shift in macrophage phenotype toward a less immunosuppressive profile, restored liver macrophage in vivo phagocytic activity, and reduced bacterial dissemination. CONCLUSION: Liver cirrhosis is characterized by a remarkable impairment of phagocytic function of macrophages associated with an immunosuppressive transcriptome profile. The programmed cell death receptor 1/PD-L1 axis plays a major role in the impaired activity of liver macrophages. PD-L1 blockade reverses the immune suppressive profile and increases antimicrobial activity of liver macrophages in cirrhosis.


Asunto(s)
Antígeno B7-H1/metabolismo , Infecciones Bacterianas/inmunología , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Cirrosis Hepática/inmunología , Macrófagos/inmunología , Anciano , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Infecciones Bacterianas/prevención & control , Biopsia , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Hígado/inmunología , Hígado/patología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/patología , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Fagocitosis , Cultivo Primario de Células , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Índice de Severidad de la Enfermedad
6.
Cell Microbiol ; 20(11): e12869, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29904997

RESUMEN

Staphylococcus lugdunensis is a commensal bacterium that can cause serious infection suggesting an ability to circumvent aspects of host immunity. We demonstrate here that macrophages fail to kill ingested S. lugdunensis and the bacteria persist for extended periods, without replicating, within mature LAMP-1-positive phagolysosomes. Phagocytosed S. lugdunensis also do not intoxicate host cells in contrast to Staphylococcus aureus. Optimal survival of S. lugdunensis requires O-acetylated peptidoglycan because an oatA mutant, which is more sensitive to killing by lysozyme than wild type, survived to a lesser extent in macrophages. In vitro models of macrophage infection reveal that viable intracellular S. lugdunensis bacteria can be made to grow by pharmacologic perturbation of phagosome function or by phagocyte intoxication by S. aureus toxins. Remarkably, replicating S. lugdunensis is not constrained by LAMP-1 and phosphatidylserine-positive endomembranes, which is distinct from S. aureus that replicates within phagolysosomes. In vivo, S. lugdunensis can also reside in the murine Kupffer cell where the bacteria persist without replicating and require oatA to resist killing in vivo. The intracellular environment of the macrophage represents a niche where S. lugdunensis can exist while protected from extracellular immune factors and may serve as a reservoir from which these bacteria could disseminate.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Evasión Inmune , Macrófagos/microbiología , Infecciones Estafilocócicas/inmunología , Staphylococcus lugdunensis/patogenicidad , Animales , Toxinas Bacterianas/farmacología , Células Cultivadas , Femenino , Humanos , Macrófagos del Hígado/microbiología , Macrófagos del Hígado/patología , Proteínas de Membrana de los Lisosomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Peptidoglicano/genética , Peptidoglicano/metabolismo , Fagosomas/microbiología , Células RAW 264.7 , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/química , Staphylococcus aureus/patogenicidad , Staphylococcus lugdunensis/fisiología
7.
Cell Tissue Res ; 371(3): 489-503, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29204747

RESUMEN

Humans are well equipped to defend themselves against bacteria. The innate immune system employs diverse mechanisms to recognize, control and initiate a response that can destroy millions of different microbes. Microbes that evade the sophisticated innate immune system are able to escape detection and could become pathogens. The pathogens Streptococcus pneumoniae and Staphylococcus aureus are particularly successful due to the development of a wide variety of virulence strategies for bacterial pathogenesis and they invest significant efforts towards mechanisms that allow for neutrophil evasion. Neutrophils are a primary cellular defense and can rapidly kill invading microbes, which is an indispensable function for maintaining host health. This review compares the key features of Streptococcus pneumoniae and Staphylococcus aureus in epidemiology, with a specific focus on virulence mechanisms utilized to evade neutrophils in bacterial pathogenesis. It is important to understand the complex interactions between pathogenic bacteria and neutrophils so that we can disrupt the ability of pathogens to cause disease.


Asunto(s)
Evasión Inmune , Neutrófilos/inmunología , Staphylococcus aureus/fisiología , Streptococcus pneumoniae/fisiología , Animales , Muerte Celular , Humanos , Viabilidad Microbiana
8.
Annu Rev Microbiol ; 67: 629-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23834243

RESUMEN

The pathogen Staphylococcus aureus is well adapted to its human host. Neutrophil-mediated killing is a crucial defense system against S. aureus; however, the pathogen has evolved many strategies to resist killing. We first describe the discrete steps of neutrophil activation and migration to the site of infection and the killing of microbes by neutrophils in general. We then highlight the different approaches utilized by S. aureus to resist the different steps of neutrophil attack. Various molecules are discussed in their evolutionary context. Most of the molecules secreted by S. aureus to combat neutrophil attacks at the site of infection show clear human specificity. Many elements of human neutrophil defenses appear redundant, and so the evasion strategies of staphylococci display redundant functions as well. All efforts by S. aureus to resist neutrophil-mediated killing stress the importance of these mechanisms in the pathophysiology of staphylococcal diseases. However, the highly human-specific nature of most host-pathogen interactions hinders the in vivo establishment of their contribution to staphylococcal pathophysiology.


Asunto(s)
Evasión Inmune , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología
9.
Methods ; 128: 12-19, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28522327

RESUMEN

It is central to the field of bacterial pathogenesis to define how bacteria are killed by phagocytic cells. During phagocytosis, the microbe is localized to the phagolysosome where crucial defense mechanisms such as acidification and production of reactive oxygen species (ROS) are initiated. This process has extensively been studied in vitro, however many resident tissue phagocytes will phenotypically change upon isolation from their natural environment. Therefore, interrogation of phagocytosis and phagosomal function of cells in the context of their natural tissue environment enhances our understanding of the biological process in vivo. This article outlines a real-time intravital microscopy protocol that utilizes fluorescent dyes to study the process of phagocytosis, which reveals acidification and oxidation of individual bacteria inside host cells of living animals. The novelty of this technique exists in use of bacteria that are covalently labelled with the fluorescent dyes Oxyburst and pHrodo, which respectively report on oxidation or acidification. Intravital microscopy is applied to visualize the uptake and subsequent oxidation or acidification of reporter bacteria in the organ of interest. Fluorescently labelled antibodies can be used to counter stain for host immune cells such as neutrophils and macrophages, along with reference stains to identify all bacteria. Although these assays were originally developed to assess the uptake and survival ofStaphylococcus aureusin liver resident macrophages (Kupffer cells), this protocol may be adapted to investigate any bacterium-host cell interaction.


Asunto(s)
Microscopía Intravital/métodos , Macrófagos del Hígado/microbiología , Fagosomas/microbiología , Coloración y Etiquetado/métodos , Staphylococcus aureus/crecimiento & desarrollo , Animales , Proteínas Fluorescentes Verdes/análisis , Macrófagos del Hígado/química , Macrófagos del Hígado/fisiología , Ratones , Fagosomas/química , Fagosomas/fisiología , Staphylococcus aureus/química
10.
PLoS Pathog ; 8(3): e1002606, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457627

RESUMEN

Staphylococcus aureus virulence has been associated with the production of phenol soluble modulins (PSM). PSM are known to activate, attract and lyse neutrophils. However, the functional characterizations were generally performed in the absence of human serum. Here, we demonstrate that human serum can inhibit all the previously-described activities of PSM. We observed that serum can fully block both the cell lysis and FPR2 activation of neutrophils. We show a direct interaction between PSM and serum lipoproteins in human serum and whole blood. Subsequent analysis using purified high, low, and very low density lipoproteins (HDL, LDL, and VLDL) revealed that they indeed neutralize PSM. The lipoprotein HDL showed highest binding and antagonizing capacity for PSM. Furthermore, we show potential intracellular production of PSM by S. aureus upon phagocytosis by neutrophils, which opens a new area for exploration of the intracellular lytic capacity of PSM. Collectively, our data show that in a serum environment the function of PSM as important extracellular toxins should be reconsidered.


Asunto(s)
Toxinas Bacterianas/metabolismo , Células HL-60/metabolismo , Lipoproteínas/sangre , Staphylococcus aureus/metabolismo , Factores de Virulencia/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/toxicidad , Calcio/metabolismo , Señalización del Calcio , Células HL-60/inmunología , Humanos , Pruebas de Neutralización , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fenol/química , Unión Proteica , Solubilidad , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Factores de Virulencia/inmunología , Factores de Virulencia/toxicidad
11.
Cell Microbiol ; 15(10): 1753-65, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23601501

RESUMEN

Neutrophil recruitment is essential in clearing pneumococcal infections. The first step in neutrophil extravasation involves the interaction between P-selectin on activated endothelium and P-Selectin Glycoprotein 1 (PSGL-1) on neutrophils. Here, we identify pneumococcal Zinc metalloproteinase C as a potent inhibitor of PSGL-1. ZmpC degrades the N-terminal domain of PSGL-1, thereby disrupting the initial rolling of neutrophils on activated human umbilical vein endothelial cells. Furthermore, mice infected with wild-type strain in the model of pneumococcal pneumonia showed lower lungs neutrophil infiltration compare to animals infected with ZmpC mutant. In addition, we confirmed the association of zmpC with serotype 8 and 11A and found it to be associated with serotype 33F as well. In conclusion, wereport PSGL-1 as a novel target for ZmpC and show that ZmpC inhibits neutrophil extravasation during pneumococcal pneumonia.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Neutrófilos/inmunología , Streptococcus pneumoniae/fisiología , Animales , Adhesión Celular , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Eliminación de Gen , Humanos , Pulmón/inmunología , Pulmón/patología , Metaloendopeptidasas/genética , Ratones , Neumonía Neumocócica/patología , Proteolisis , Streptococcus pneumoniae/genética
12.
Microbiol Spectr ; 12(6): e0007124, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38700321

RESUMEN

Novel antimicrobial agents are needed to combat antimicrobial resistance. This study tested novel pentafluorosulfanyl-containing triclocarban analogs for their potential antibacterial efficacy. Standard procedures were used to produce pentafluorosulfanyl-containing triclocarban analogs. Twenty new compounds were tested against seven Gram-positive and Gram-negative indicator strains as well as 10 clinical isolates for their antibacterial and antibiofilm activity. Mechanistic investigations focused on damage to cell membrane, oxidizing reduced thiols, iron-sulfur clusters, and oxidative stress to explain the compounds' activity. Safety profiles were assessed using cytotoxicity experiments in eukaryotic cell lines. Following screening, selected components had significantly better antibacterial and antibiofilm activity against Gram-positive bacteria in lower concentrations in comparison to ciprofloxacin and gentamycin. For instance, one compound had a minimum inhibitory concentration of <0.0003 mM, but ciprofloxacin had 0.08 mM. Mechanistic studies show that these novel compounds do not affect reduced thiol content, iron-sulfur clusters, or hydrogen peroxide pathways. Their impact comes from Gram-positive bacterial cell membrane damage. Tests on cell culture toxicity and host component safety showed promise. Novel diarylurea compounds show promise as Gram-positive antimicrobials. These compounds offer prospects for study and optimization. IMPORTANCE: The rise of antibiotic resistance among bacterial pathogens poses a significant threat to global health, underscoring the urgent need for novel antimicrobial agents. This study presents research on a promising class of novel compounds with potent antibacterial properties against Gram-positive bacteria, notably Staphylococcus aureus and MRSA. What sets these novel analogs apart is their superior efficacy at substantially lower concentrations compared with commonly used antibiotics like ciprofloxacin and gentamycin. Importantly, these compounds act by disrupting the bacterial cell membrane, offering a unique mechanism that could potentially circumvent existing resistance mechanisms. Preliminary safety assessments also highlight their potential for therapeutic use. This study not only opens new avenues for combating antibiotic-resistant infections but also underscores the importance of innovative chemical approaches in addressing the global antimicrobial resistance crisis.


Asunto(s)
Antibacterianos , Carbanilidas , Bacterias Grampositivas , Pruebas de Sensibilidad Microbiana , Carbanilidas/farmacología , Carbanilidas/química , Antibacterianos/farmacología , Antibacterianos/química , Bacterias Grampositivas/efectos de los fármacos , Humanos , Biopelículas/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Ciprofloxacina/farmacología
13.
Infect Immun ; 81(5): 1830-41, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23509138

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) strains of the pulsed-field type USA300 are primarily responsible for the current community-associated epidemic of MRSA infections in the United States. The success of USA300 is partly attributed to the ability of the pathogen to avoid destruction by human neutrophils (polymorphonuclear leukocytes [PMNs]), which are crucial to the host immune response to S. aureus infection. In this work, we investigated the contribution of bicomponent pore-forming toxins to the ability of USA300 to withstand attack from primary human PMNs. We demonstrate that in vitro growth conditions influence the expression, production, and availability of leukotoxins by USA300, which in turn impact the cytotoxic potential of this clone toward PMNs. Interestingly, we also found that upon exposure to PMNs, USA300 preferentially activates the promoter of the lukAB operon, which encodes the recently identified leukocidin AB (LukAB). LukAB elaborated by extracellular S. aureus forms pores in the plasma membrane of PMNs, leading to PMN lysis, highlighting a contribution of LukAB to USA300 virulence. We now show that LukAB also facilitates the escape of bacteria engulfed within PMNs, in turn enabling the replication and outgrowth of S. aureus. Together, these results suggest that upon encountering PMNs S. aureus induces the production of LukAB, which serves as an extra- and intracellular weapon to protect the bacterium from destruction by human PMNs.


Asunto(s)
Toxinas Bacterianas/metabolismo , Evasión Inmune/inmunología , Leucocidinas/metabolismo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Infecciones Estafilocócicas/inmunología , Humanos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Fagocitosis/fisiología , Infecciones Estafilocócicas/epidemiología , Infecciones Estafilocócicas/microbiología , Virulencia/fisiología , Factores de Virulencia/metabolismo
14.
Science ; 381(6662): eabq5202, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37676943

RESUMEN

Kupffer cells (KCs) are localized in liver sinusoids but extend pseudopods to parenchymal cells to maintain their identity and serve as the body's central bacterial filter. Liver cirrhosis drastically alters vascular architecture, but how KCs adapt is unclear. We used a mouse model of liver fibrosis and human tissue to examine immune adaptation. Fibrosis forced KCs to lose contact with parenchymal cells, down-regulating "KC identity," which rendered them incapable of clearing bacteria. Commensals stimulated the recruitment of monocytes through CD44 to a spatially distinct vascular compartment. There, recruited monocytes formed large aggregates of multinucleated cells (syncytia) that expressed phenotypical KC markers and displayed enhanced bacterial capture ability. Syncytia formed via CD36 and were observed in human cirrhosis as a possible antimicrobial defense that evolved with fibrosis.


Asunto(s)
Infecciones de Transmisión Sanguínea , Células Gigantes , Macrófagos del Hígado , Cirrosis Hepática , Animales , Humanos , Ratones , Células Gigantes/inmunología , Células Gigantes/microbiología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/microbiología , Cirrosis Hepática/inmunología , Cirrosis Hepática/microbiología , Cirrosis Hepática/patología , Infecciones de Transmisión Sanguínea/inmunología , Modelos Animales de Enfermedad
15.
Biomedicines ; 10(8)2022 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-36009351

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) infections pose a global health threat, especially with the continuous development of antibiotic resistance. As an opportunistic pathogen, MRSA infections have a high mortality rate worldwide. Although classically described as an extracellular pathogen, many studies have shown over the past decades that MRSA also has an intracellular aspect to its infectious cycle, which has been observed in vitro in both non-professional as well as professional phagocytes. In vivo, MRSA has been shown to establish an intracellular niche in liver Kupffer cells upon bloodstream infection. The staphylococci have evolved various evasion strategies to survive the antimicrobial environment of phagolysosomes and use these compartments to hide from immune cells and antibiotics. Ultimately, the host cells get overwhelmed by replicating bacteria, leading to cell lysis and bacterial dissemination. In this review, we describe the different intracellular aspects of MRSA infection and briefly mention S. aureus evasion strategies. We discuss how this intracellular niche of bacteria may assist in antibiotic tolerance development, and lastly, we describe various new antibacterial strategies that target the intracellular bacterial niche.

16.
Redox Biol ; 49: 102225, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34959099

RESUMEN

BACKGROUND: Neutrophils play a role in innate immunity and are critical for clearance of Staphylococcus aureus. Current understanding of neutrophil bactericidal effects is that NADPH oxidase produces reactive oxygen species (ROS), mediating bacterial killing. Neutrophils also contain numerous mitochondria; since these organelles lack oxidative metabolism, their function is unclear. We hypothesize that mitochondria in human neutrophils contribute to the bactericidal capacity of S. aureus. METHODS: and Findings: Using human neutrophils isolated from healthy volunteers (n = 13; 7 females, 6 males), we show that mitochondria are critical in the immune response to S. aureus. Using live-cell and fixed confocal, and transmission electron microscopy, we show mitochondrial tagging of bacteria prior to ingestion and surrounding of phagocytosed bacteria immediately upon engulfment. Further, we demonstrate that mitochondria are ejected from intact neutrophils and engage bacteria during vital NETosis. Inhibition of the mitochondrial electron transport chain at Complex III, but not Complex I, attenuates S. aureus killing by 50 ± 7%, comparable to the NADPH oxidase inhibitor apocynin. Similarly, mitochondrial ROS scavenging using MitoTEMPO attenuates bacterial killing 112 ± 60% versus vehicle control. Antimycin A treatment also reduces mitochondrial ROS production by 50 ± 12% and NETosis by 53 ± 5%. CONCLUSIONS: We identify a previously unrecognized role for mitochondria in human neutrophils in the killing of S. aureus. Inhibition of electron transport chain Complex III significantly impairs antimicrobial activity. This is the first demonstration that vital NETosis, an early event in the antimicrobial response, occurring within 5 min of bacterial exposure, depends on the function of mitochondrial Complex III. Mitochondria join NADPH oxidase as bactericidal ROS generators that mediate the bactericidal activities of human neutrophils.


Asunto(s)
Neutrófilos , Staphylococcus aureus , Femenino , Humanos , Masculino , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Fagocitosis , Especies Reactivas de Oxígeno/metabolismo , Staphylococcus aureus/metabolismo
17.
JCI Insight ; 7(2)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34908534

RESUMEN

Acute respiratory distress syndrome (ARDS) is a life-threatening syndrome, constituted by respiratory failure and diffuse alveolar damage that results from dysregulated local and systemic immune activation, causing pulmonary vascular, parenchymal, and alveolar damage. SARS-CoV-2 infection has become the dominant cause of ARDS worldwide, and emerging evidence implicates neutrophils and their cytotoxic arsenal of effector functions as central drivers of immune-mediated lung injury in COVID-19 ARDS. However, key outstanding questions are whether COVID-19 drives a unique program of neutrophil activation or effector functions that contribute to the severe pathogenesis of this pandemic illness and whether this unique neutrophil response can be targeted to attenuate disease. Using a combination of high-dimensional single-cell analysis and ex vivo functional assays of neutrophils from patients with COVID-19 ARDS, compared with those with non-COVID ARDS (caused by bacterial pneumonia), we identified a functionally distinct landscape of neutrophil activation in COVID-19 ARDS that was intrinsically programmed during SARS-CoV-2 infection. Furthermore, neutrophils in COVID-19 ARDS were functionally primed to produce high amounts of neutrophil extracellular traps. Surprisingly, this unique pathological program of neutrophil priming escaped conventional therapy with dexamethasone, thereby revealing a promising target for adjunctive immunotherapy in severe COVID-19.


Asunto(s)
COVID-19/inmunología , Trampas Extracelulares/inmunología , Activación Neutrófila , Neutrófilos/inmunología , Síndrome de Dificultad Respiratoria/inmunología , SARS-CoV-2/inmunología , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neutrófilos/patología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Síndrome de Dificultad Respiratoria/patología , Índice de Severidad de la Enfermedad
18.
Biomaterials ; 287: 121632, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35728409

RESUMEN

Infections with Staphylococcus aureus (S. aureus) have been reported from various organs ranging from asymptomatic colonization to severe infections and sepsis. Although considered an extracellular pathogen, S. aureus can invade and persist in professional phagocytes such as monocytes and macrophages. Its capability to persist and manipulate macrophages is considered a critical step to evade host antimicrobial reactions. We leveraged a recently established human liver-on-chip model to demonstrate that S. aureus specifically targets macrophages as essential niche facilitating bacterial persistence and phenotype switching to small colony variants (SCVs). In vitro, M2 polarization was found to favor SCV-formation and was associated with increased intracellular bacterial loads in macrophages, increased cell death, and impaired recruitment of circulating monocytes to sites of infection. These findings expand the knowledge about macrophage activation in the liver and its impact on bacterial persistence and dissemination in the course of infection.

19.
Nat Commun ; 13(1): 3279, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672409

RESUMEN

Invariant NKT (iNKT) cells comprise a heterogeneous group of non-circulating, tissue-resident T lymphocytes that recognize glycolipids, including alpha-galactosylceramide (αGalCer), in the context of CD1d, but whether peripheral iNKT cell subsets are terminally differentiated remains unclear. Here we show that mouse and human liver-resident αGalCer/CD1d-binding iNKTs largely correspond to a novel Zbtb16+Tbx21+Gata3+MaflowRorc- subset that exhibits profound transcriptional, phenotypic and functional plasticity. Repetitive in vivo encounters of these liver iNKT (LiNKT) cells with intravenously delivered αGalCer/CD1d-coated nanoparticles (NP) trigger their differentiation into immunoregulatory, IL-10+IL-21-producing Zbtb16highMafhighTbx21+Gata3+Rorc- cells, termed LiNKTR1, expressing a T regulatory type 1 (TR1)-like transcriptional signature. This response is LiNKT-specific, since neither lung nor splenic tissue-resident iNKT cells from αGalCer/CD1d-NP-treated mice produce IL-10 or IL-21. Additionally, these LiNKTR1 cells suppress autoantigen presentation, and recognize CD1d expressed on conventional B cells to induce IL-10+IL-35-producing regulatory B (Breg) cells, leading to the suppression of liver and pancreas autoimmunity. Our results thus suggest that LiNKT cells are plastic for further functional diversification, with such plasticity potentially targetable for suppressing tissue-specific inflammatory phenomena.


Asunto(s)
Linfocitos B Reguladores , Células T Asesinas Naturales , Animales , Antígenos CD1d/metabolismo , Autoinmunidad , Linfocitos B Reguladores/metabolismo , Galactosilceramidas , Interleucina-10/metabolismo , Hígado/metabolismo , Ratones
20.
Pathogens ; 10(4)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33805913

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) first emerged after methicillin was introduced to combat penicillin resistance, and its prevalence in Canada has increased since the first MRSA outbreak in the early 1980s. We reviewed the existing literature on MRSA prevalence in Canada over time and in diverse populations across the country. MRSA prevalence increased steadily in the 1990s and 2000s and remains a public health concern in Canada, especially among vulnerable populations, such as rural, remote, and Indigenous communities. Antibiotic resistance patterns and risk factors for MRSA infection were also reported. All studies reported high susceptibility (>85%) to trimethoprim-sulfamethoxazole, with no significant resistance reported for vancomycin, linezolid, or rifampin. While MRSA continues to have susceptibility to several antibiotics, the high and sometimes variable resistance rates to other drugs underscores the importance of antimicrobial stewardship. Risk factors for high MRSA infection rates related to infection control measures, low socioeconomic status, and personal demographic characteristics were also reported. Additional surveillance, infection control measures, enhanced anti-microbial stewardship, and community education programs are necessary to decrease MRSA prevalence and minimize the public health risk posed by this pathogen.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA