Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(43): 11488-11493, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073076

RESUMEN

Sepsis, resulting from uncontrolled inflammatory responses to bacterial infections, continues to cause high morbidity and mortality worldwide. Currently, effective sepsis treatments are lacking in the clinic, and care remains primarily supportive. Here we report the development of macrophage biomimetic nanoparticles for the management of sepsis. The nanoparticles, made by wrapping polymeric cores with cell membrane derived from macrophages, possess an antigenic exterior the same as the source cells. By acting as macrophage decoys, these nanoparticles bind and neutralize endotoxins that would otherwise trigger immune activation. In addition, these macrophage-like nanoparticles sequester proinflammatory cytokines and inhibit their ability to potentiate the sepsis cascade. In a mouse Escherichia coli bacteremia model, treatment with macrophage mimicking nanoparticles, termed MΦ-NPs, reduced proinflammatory cytokine levels, inhibited bacterial dissemination, and ultimately conferred a significant survival advantage to infected mice. Employing MΦ-NPs as a biomimetic detoxification strategy shows promise for improving patient outcomes, potentially shifting the current paradigm of sepsis management.


Asunto(s)
Membrana Celular/química , Citocinas/química , Endotoxinas/química , Infecciones por Escherichia coli/terapia , Nanopartículas/química , Sepsis/terapia , Animales , Bacteriemia/terapia , Línea Celular , Lipopolisacáridos/farmacología , Lipoproteínas/química , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Receptor Toll-Like 4
2.
J Biol Chem ; 292(27): 11243-11249, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28416607

RESUMEN

Streptococcus agalactiae (group B Streptococcus, GBS) is a leading cause of invasive bacterial infections in newborns, typically acquired vertically during childbirth secondary to maternal vaginal colonization. Human milk oligosaccharides (HMOs) have important nutritional and biological activities that guide the development of the immune system of the infant and shape the composition of normal gut microbiota. In this manner, HMOs help protect against pathogen colonization and reduce the risk of infection. In the course of our studies of HMO-microbial interactions, we unexpectedly uncovered a novel HMO property to directly inhibit the growth of GBS independent of host immunity. By separating different HMO fractions through multidimensional chromatography, we found the bacteriostatic activity to be confined to specific non-sialylated HMOs and synergistic with a number of conventional antibiotic agents. Phenotypic screening of a GBS transposon insertion library identified a mutation within a GBS-specific gene encoding a putative glycosyltransferase that confers resistance to HMOs, suggesting that HMOs may function as an alternative substrate to modify a GBS component in a manner that impairs growth kinetics. Our study uncovers a unique antibacterial role for HMOs against a leading neonatal pathogen and expands the potential therapeutic utility of these versatile molecules.


Asunto(s)
Antibacterianos/farmacología , Leche Humana/química , Oligosacáridos/farmacología , Streptococcus agalactiae/crecimiento & desarrollo , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Femenino , Humanos , Oligosacáridos/química , Oligosacáridos/aislamiento & purificación , Streptococcus agalactiae/genética
3.
Front Pediatr ; 7: 410, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31737584

RESUMEN

Group B Streptococcus (GBS) emerged as a leading cause of invasive infectious disease in neonates in the 1970s, but has recently been identified as an escalating public health threat in non-pregnant adults, particularly those of advanced aged or underlying medical conditions. GBS infection can rapidly develop into life-threatening disease despite prompt administration of effective antibiotics and initiation of state-of-the-art intensive care protocols and technologies due to deleterious bacterial virulence factors, such as the GBS pore-forming toxin ß-hemolysin/cytolysin (ß-H/C). ß-H/C is known to have noxious effects on a wide range of host cells and tissues, including lung epithelial cell injury, blood brain barrier weakening, and immune cell apoptosis. Neonatal and adult survivors of GBS infection are at a high risk for substantial long-term health issues and neurologic disabilities due to perturbations in organ systems caused by bacterial- and host- mediated inflammatory stressors. Previously engineered anti-virulence inhibitors, such as monoclonal antibodies and small molecular inhibitors, generally require customized design for each different pathogenic toxin and do not target deleterious host pro-inflammatory responses that may cause organ injury, septic shock, or death. By simply wrapping donor red blood cells (RBCs) around polymeric cores, we have created biomimetic "nanosponges." Because nanoparticles retain the same repertoire of cell membrane receptors as their host cell, they offer non-specific and all-purpose toxin decoy strategies with a broad ability to sequester and neutralize various bacterial toxins and host pro-inflammatory chemokines and cytokines to attenuate the course of infectious disease. This proof-of-concept study successfully demonstrated that intervention with nanosponges reduced the hemolytic activity of live GBS and stabilized ß-H/C in a dose-dependent manner. Nanosponge treatment also decreased lung epithelial and macrophage cell death following exposure to live GBS bacteria and stabilized ß-H/C, improved neutrophil killing of GBS, and diminished GBS-induced macrophage IL-1ß production. Our results, therefore, suggest biomimetic nanosponges provide a titratable detoxification therapy that may provide a first-in-class treatment option for GBS infection by sequestering and inhibiting ß-H/C activity.

4.
Toxins (Basel) ; 10(12)2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30562923

RESUMEN

For many of the most important human bacterial infections, invasive disease severity is fueled by the cell damaging and pro-inflammatory effects of secreted pore-forming toxins (PFTs). Isogenic PFT-knockout mutants, e.g., Staphylococcus aureus lacking α-toxin or Streptococcus pneumoniae deficient in pneumolysin, show attenuation in animal infection models. This knowledge has inspired multi-model investigations of strategies to neutralize PFTs or counteract their toxicity as a novel pharmacological approach to ameliorate disease pathogenesis in clinical disease. Promising examples of small molecule, antibody or nanotherapeutic drug candidates that directly bind and neutralize PFTs, block their oligomerization or membrane receptor interactions, plug establishment membrane pores, or boost host cell resiliency to withstand PFT action have emerged. The present review highlights these new concepts, with a special focus on ß-PFTs produced by leading invasive human Gram-positive bacterial pathogens. Such anti-virulence therapies could be applied as an adjunctive therapy to antibiotic-sensitive and -resistant strains alike, and further could be free of deleterious effects that deplete the normal microflora.


Asunto(s)
Infecciones Bacterianas/tratamiento farmacológico , Proteínas Citotóxicas Formadoras de Poros/antagonistas & inhibidores , Animales , Humanos , Proteínas Citotóxicas Formadoras de Poros/clasificación , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Vacunas/uso terapéutico
5.
Adv Healthc Mater ; 7(13): e1701366, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29436150

RESUMEN

Neutralization of bacterial toxins has become a compelling approach to treating bacterial infections as it may pose less selective pressure for the development of bacterial resistance. Currently, the majority of toxin neutralization platforms act by targeting the molecular structure of the toxin, which requires toxin identification and customized design for different diseases. Therefore, their development has been challenged by the enormous number and complexity of bacterial toxins. Herein, biomimetic toxin nanosponges are formulated by coating membranes of human red blood cells (hRBCs) onto polymeric nanoparticles, which act as a toxin decoy to absorb and neutralize a broad-spectrum of hemolytic toxins regardless of their molecular structure. When tested with model pore-forming toxins, including melittin, α-hemolysin of methicillin-resistant Staphylococcus aureus, listeriolysin O of Listeria monocytogenes, and streptolysin O of Group A Streptococcus, the hRBC nanosponges are able to completely inhibit toxin-induced hemolysis in a concentration-dependent manner. In addition, the nanosponge-detained toxins show no cytotoxicity when tested on human umbilical vein endothelial cells and no lethality when injected into mice, which together indicate effective toxin neutralization. Overall, these results demonstrate the broad applicability and high effectiveness of the hRBC nanosponges as a novel antivirulence platform against hemolytic toxins from various strains of bacteria.


Asunto(s)
Toxinas Bacterianas/química , Membrana Eritrocítica/química , Nanoestructuras/química , Animales , Proteínas Bacterianas/química , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Materiales Biomiméticos , Proteínas Hemolisinas/química , Hemólisis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Meliteno/química , Ratones Endogámicos ICR , Estreptolisinas/química
6.
Front Pharmacol ; 8: 477, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28769806

RESUMEN

Group A Streptococcus (GAS), an important human-specific Gram-positive bacterial pathogen, is associated with a broad spectrum of disease, ranging from mild superficial infections such as pharyngitis and impetigo, to serious invasive infections including necrotizing fasciitis and streptococcal toxic shock syndrome. The GAS pore-forming streptolysin O (SLO) is a well characterized virulence factor produced by nearly all GAS clinical isolates. High level expression of SLO is epidemiologically linked to intercontinental dissemination of hypervirulent clonotypes and poor clinical outcomes. SLO can trigger macrophage and neutrophil cell death and/or the inactivation of immune cell functions, and promotes tissue injury and bacterial survival in animal models of infection. In the present work, we describe how the pharmacological presentation of red blood cell (RBC) derived biomimetic nanoparticles ("nanosponges") can sequester SLO and block the ability of GAS to damage host cells, thereby preserving innate immune function and increasing bacterial clearance in vitro and in vivo. Nanosponge administration protected human neutrophils, macrophages, and keratinocytes against SLO-mediated cytotoxicity. This therapeutic intervention prevented SLO-induced macrophage apoptosis and increased neutrophil extracellular trap formation, allowing increased GAS killing by the respective phagocytic cell types. In a murine model of GAS necrotizing skin infection, local administration of the biomimetic nanosponges was associated with decreased lesion size and reduced bacterial colony-forming unit recovery. Utilization of a toxin decoy and capture platform that inactivates the secreted SLO before it contacts the host cell membrane, presents a novel virulence factor targeted strategy that could be a powerful adjunctive therapy in severe GAS infections where morbidity and mortality are high despite antibiotic treatment.

7.
ACS Nano ; 11(12): 11923-11930, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29116753

RESUMEN

Colloidal gels consisting of oppositely charged nanoparticles are increasingly utilized for drug delivery and tissue engineering. Meanwhile, cell membrane-coated nanoparticles are becoming a compelling biomimetic system for innovative therapeutics. Here, we demonstrate the successful use of cell membrane-coated nanoparticles as building blocks to formulate a colloidal gel that gelates entirely based on material self-assembly without chemical cross-linking. Specifically, we prepare red blood cell membrane-coated nanosponges and mix them with an appropriate amount of cationic nanoparticles, resulting in a spontaneously formed gel-like complex. Rheological test shows that the nanosponge colloidal gel has pronounced shear-thinning property, which makes it an injectable formulation. The gel formulation not only preserves the nanosponges' toxin neutralization capability but also greatly prolongs their retention time after subcutaneous injection into mouse tissue. When tested in a mouse model of subcutaneous group A Streptococcus infection, the nanosponge colloidal gel shows significant antibacterial efficacy by markedly reducing skin lesion development. Overall, the nanosponge colloidal gel system is promising as an injectable formulation for therapeutic applications such as antivirulence treatment for local bacterial infections.


Asunto(s)
Membrana Eritrocítica/química , Nanopartículas/química , Coloides/síntesis química , Coloides/química , Geles/síntesis química , Geles/química , Tamaño de la Partícula , Propiedades de Superficie
8.
Mol Cell Endocrinol ; 427: 73-85, 2016 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-26992564

RESUMEN

Oxysterol binding protein-related protein 2 (ORP2) is a lipid binding protein that has been implicated in various cellular processes, including lipid sensing, cholesterol efflux, and endocytosis. We recently identified ORP2 as a member of a protein complex that regulates glucocorticoid biosynthesis. Herein, we examine the effect of silencing ORP2 on adrenocortical function and show that the ORP2 knockdown cells exhibit reduced amounts of multiple steroid metabolites, including progesterone, 11-deoxycortisol, and cortisol, but have increased concentrations of androgens, and estrogens. Moreover, silencing ORP2 suppresses the expression of most proteins required for cortisol production and reduces the expression of steroidogenic factor 1 (SF1). ORP2 silencing also increases cellular cholesterol, concomitant with decreased amounts of 22-hydroxycholesterol and 7-ketocholesterol, two molecules that have been shown to bind to ORP2. Further, we show that ORP2 binds to liver X receptor (LXR) and is required for nuclear LXR expression. LXR and ORP2 are recruited to the CYP11B1 promoter in response to cAMP signaling. Additionally, ORP2 is required for the expression of other LXR target genes, including ABCA1 and the LDL receptor (LDLR). In summary, we establish a novel role for ORP2 in regulating steroidogenic capacity and cholesterol homeostasis in the adrenal cortex.


Asunto(s)
Colesterol/metabolismo , Hidrocortisona/biosíntesis , Receptores de Esteroides/fisiología , Corteza Suprarrenal/metabolismo , Línea Celular , Deshidroepiandrosterona/biosíntesis , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Homeostasis/efectos de los fármacos , Humanos , Receptores de Esteroides/genética , Esteroides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA