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.
Antimicrob Agents Chemother ; 68(9): e0023524, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39072634

RESUMEN

In response to the antimicrobial resistance crisis, we have developed a powerful and versatile therapeutic platform, the Antibacterial Drone (ABD) system. The ABD consists of a highly mobile staphylococcal pathogenicity island re-purposed to deliver genes encoding antibacterial proteins. The chromosomally located island is induced by a co-resident helper phage, packaged in phage-like particles, and released in very high numbers upon phage-induced lysis. ABD particles specifically adsorb to bacteria causing an infection and deliver their DNA to these bacteria, where the bactericidal cargo genes are expressed, kill the bacteria, and cure the infection. Here, we report a major advance of the system, incorporation of the gene encoding a secreted, bactericidal, species-specific lytic enzyme, lysostsphin. This ABD not only kills the bacterium that has been attacked by the ABD, but also any surrounding bacteria that are sensitive to the lytic enzyme which is released by secretion and by lysis of the doomed cell. So while the killing field is thus expanded, there are no civilian casualties (bacteria that are insensitive to the ABD and its cargo protein(s) are not inadvertently killed). Without amplifying the number of ABD particles (which are not re-packaged), the expression and release of the cargo gene's product dramatically extend the effective reach of the ABD. A cargo gene that encodes a secreted bactericidal protein also enables the treatment of a mixed bacterial infection in which one of the infecting organisms is insensitive to the ABD delivery system but is sensitive to the ABD's secreted cargo protein.


Asunto(s)
Antibacterianos , Lisostafina , Antibacterianos/farmacología , Lisostafina/farmacología , Islas Genómicas/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Bacteriófagos/genética
2.
Artículo en Inglés | MEDLINE | ID: mdl-31932377

RESUMEN

We present a case of endocarditis wherein organisms cultured from different valve leaflets yielded different daptomycin susceptibilities from each other and from organisms obtained from peripheral blood culture. Genomic analyses showed mutations in mprF, purR, and agrA Pharmacokinetic simulations showed consistent activity of daptomycin plus beta-lactam against all subpopulations. This represents an opportunity to understand S. aureus evolution and fitness in vivo on daptomycin therapy and the role of beta-lactams to prevent the selection of daptomycin-resistant subpopulations.


Asunto(s)
Daptomicina/farmacología , Endocarditis Bacteriana/microbiología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Válvula Tricúspide/microbiología , Válvula Tricúspide/patología , Aminoaciltransferasas/genética , Proteínas Bacterianas/genética , Daptomicina/uso terapéutico , Endocarditis Bacteriana/tratamiento farmacológico , Endocarditis Bacteriana/genética , Humanos , Pruebas de Sensibilidad Microbiana , Mutación/genética , Proteínas Represoras/genética , Válvula Tricúspide/efectos de los fármacos , Secuenciación Completa del Genoma
3.
Antibiotics (Basel) ; 9(1)2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31861446

RESUMEN

Immune biomarkers can stratify mortality risk in staphylococcal bacteremia. Microbial biomarkers may provide more consistent signals during early infection. We demonstrate that in ST45/USA600 bacteremia, bacterial membrane vesicle production in vitro predicts clinical mortality (773 vs. 116 RFU, survivors vs. decedents, p < 0.0001). Using a threshold of 301 relative fluorescence units (RFU), the sensitivity and specificity of the membrane vesicles to predict mortality are 78% and 90%, respectively. This platform is facile, scalable and can be integrated into clinical microbiology lab workflows.

4.
Microb Pathog ; 115: 239-250, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29274459

RESUMEN

Alarming increase of death due to S. aureus sepsis demands newer treatment strategies. Enhancement of antibiotic resistant S. aureus strains caused increased mortality. Only antibiotic treatment for Staphylococcal sepsis has been found insufficient to improve outcomes. In the innate immune response, phagocytosis mediated killing of pathogen and further triggering of intracellular signaling cascades by the PRRs culminates in the release of a variety of pro inflammatory cytokines, which orchestrate together in the early host response to infection. Increased production of inflammatory cytokines not only delineate pathogen burden but also affects host cell by triggering inflammation. Therefore, combinational therapy of Ascorbic acid is used along with antibiotics Ofloxacin (OFX) or Chloramphenicol (CHL) to kill S. aureus by mouse peritoneal macrophages. For this ROS like H2O2, superoxide anion and NO production was accessed, TLR2 and COX2 expression was monitored. Pro-inflammatory cytokines along with antioxidant levels were also analyzed. Ascorbic acid along with antibiotics OFX or CHL promoted bacterial clearance at early infection by increasing H2O2 and O2-.NO production has been found to decrease, providing protection against harmful per-oxynitril ion. Increase in TLR-2 expression resulted in enhanced phagocytosis and subsequently more killing. Treatment with Ascorbic acid decreased proinflammatory cytokines and inflammatory markers like iNOS and COX2. This combination increased antioxidant enzymes like SOD, Catalase, GSH as well as decreased LPO, thus balancing ROS and antioxidant status inside the cell. Thus in-vitro augmentation of bacterial clearance along with regulated inflammation as found by decrease in proinflammatory cytokines like TNF-α IFN-γ,IL-6 and inflammatory markers like COX2 may be considered as a novel and important therapeutic strategy.


Asunto(s)
Antibacterianos/farmacología , Ácido Ascórbico/farmacología , Cloranfenicol/farmacología , Inflamación/inmunología , Macrófagos Peritoneales/inmunología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/inmunología , Ofloxacino/farmacología , Animales , Antioxidantes/análisis , Ciclooxigenasa 2/biosíntesis , Citocinas/inmunología , Citocinas/metabolismo , Quimioterapia Combinada , Peróxido de Hidrógeno/análisis , Macrófagos Peritoneales/microbiología , Masculino , Ratones , Óxido Nítrico/biosíntesis , Fagocitosis/inmunología , Infecciones Estafilocócicas/microbiología , Superóxidos/análisis , Receptor Toll-Like 2/biosíntesis
5.
Microb Pathog ; 105: 307-320, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28242423

RESUMEN

Death due to sepsis by S. aureus is rapidly increasing because of their potent weaponries against macrophage mediated killing. Macrophages serve as intracellular reservoirs of S. aureus. Although significant resources have been invested during the last decade in new treatments for sepsis, only antibiotic therapy has failed to improve outcomes. Moreover the host pathogen interaction resulted in host cell death triggering inflammation. So, successful therapy requires amalgamation of therapies to delineate pathogen along with providing protection to host cell. With this idea, LNMMA, the iNOS inhibitor is used along with antibiotics Ofloxacin or Chloramphenicol on S. aureus infected mouse peritoneal macrophage. ROS like H2O2, O2- production has been measured. NO inhibition by iNOS inhibitor and antioxidant levels has been analysed. COX2, TLR2 and iNOS expression along with proinflammatory cytokine level was studied. It was found that the use of iNOS inhibitor LNMMA along with antibiotics not only enhances bacterial clearance but also decreases proinflammatory responses in Staphylococcus aureus infected macrophages. Inhibition of TLR2 as well as COX2 has also been found in combined treatment groups. The use of iNOS inhibitor LNMMA plus Ofloxacin or Chloramphenicol pretreatment enhanced bacterial clearance by increasing ROS. Decreases in NO protect the cell from harmful peroxynitril as well as inflammatory damage by changes in iNOS, COX2 activity along with reduced proinflammatory cytokines like TNFα, IFNγ, IL1-ß etc. Changes in antioxidant level has been found. This in-vitro realm of augmented bacterial clearance and regulated inflammation may be considered as a novel and important therapeutic intervention.


Asunto(s)
Cloranfenicol/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/microbiología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Ofloxacino/farmacología , Staphylococcus aureus/efectos de los fármacos , omega-N-Metilarginina/farmacología , Animales , Antibacterianos/farmacología , Antioxidantes/metabolismo , Ciclooxigenasa 2/biosíntesis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oxígeno/metabolismo , Distribución Aleatoria , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Receptor Toll-Like 2/biosíntesis
6.
J Inflamm (Lond) ; 13: 36, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27932936

RESUMEN

BACKGROUND: Macrophages serve as intracellular reservoirs of S. aureus. Recent in vitro studies have confirmed high level resistance by S. aureus to macrophage mediated killing and the intracellular persistence of Staphylococci may play an important role in the pathogenesis. Since this localization protects them from both cell-mediated and humoral immune responses, therefore, a successful anti-staphylococcal therapy should include the elimination of intracellular bacteria, further protecting the host cells from staphylococci-induced cell death. So, only antibiotic therapy may not be helpful, successful therapy needs combination of drugs not only for elimination of pathogen but also for rescuing the host cell for S. aureus induced cell death. METHODS: In keeping with this idea an in vitro study has been done to examine the effect of Riboflavin along with antibiotics on phagocytosis, hydorgen peroxide, superoxide production, antioxidant enzyme levels, and cytokine levels in mouse macrophages for amelioration of the Staphylococcus aureus burden. The immune boosting effects of Riboflavin have been validated through perturbations of redox homeostasis and pro-inflammatory cytokines measurements. RESULTS: It was observed that the supplementation of Vitamin B-2 (Riboflavin) not only enhances macrophage function as previously reported but also decreases pro-inflammatory responses in Staphylococcus aureus infected macrophages. The observed influence of Riboflavin on enhanced antimicrobial effects such as enhanced phagocytosis of macrophages exposed to S. aureus, hydrogen peroxide or superoxide production when combined with either ciprofloxacin (CIP) or Azithromycin (AZM) and decrease in pro-inflammatory responses of IFN-γ, IL-6, IL-1ß. Riboflavin treatment also decreased NO and TNF-α level possibly by inhibiting the NF-κß pathway. The increased antioxidant enzymes like glutathione reductase, SOD and GSH level helped in maintaining a stable redox state in the cell. CONCLUSION: Riboflavin plus antibiotic pretreatment not only enhances macrophage functions but also decreases proinflammatory responses in Staphylococcus aureus infected macrophages indicating better bacterial clearance and regulated inflammation which may be considered as a novel and important therapeutic intervention.

7.
J Inflamm Res ; 8: 29-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25653549

RESUMEN

This study aimed to determine any alteration in the killing of Staphylococcus aureus in murine peritoneal macrophages when chloroquine (CQ) is used alone compared with when it is used in combination with ciprofloxacin (CIP) or azithromycin (AZM). The study also aimed to find out the implication of reactive oxygen species (ROS) production and cytokine release in the intracellular killing of S. aureus in macrophages. We present here data obtained with a model of S. aureus-infected mouse peritoneal macrophages in which the intracellular growth of the bacteria and the influence of antibiotics was monitored for 30, 60, and 90 minutes in the presence or absence of CQ along with the production of ROS and alteration in levels of antioxidant enzymes and cytokines. It was observed that S. aureus-triggered cytokine response was regulated when macrophages were co-cultured with CQ and AZM as compared with CQ stimulation only. It can be suggested that action of AZM in mediating bacterial killing is enhanced by the presence of CQ, indicating enhanced uptake of AZM during early infection that may be essential for bacteria killing by AZM. Reduction of oxidative stress burden on the S. aureus-infected macrophages may pave the way for better killing of internalized S. aureus by CQ plus ciprofloxacin (CIP) or CQ plus AZM. Based on these observations, one may speculate that in an inflammatory milieu, CQ loaded with AZM elicits a stronger proinflammatory response by increasing the intracellular uptake of AZM or CIP, thus enabling the immune system to mount a more robust and prolonged response against intracellular pathogens.

8.
Inflammation ; 38(1): 224-44, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25266881

RESUMEN

Among the known Toll-like receptors (TLRs), Toll-like receptor 2 (TLR2) is a key sensor for detecting Staphylococcus aureus invasion. But the function of TLR2 during S. aureus infection in different cell populations is unclear. Two different cell subtypes were chosen to study the interaction of S. aureus with TLR2 because macrophages are extremely different from one compartment to another and their capacity to respond to live bacteria or bacterial products differs from one site to another. The contribution of TLR2 to the host innate response against acute live S. aureus infection and heat-killed S. aureus (HKSA) using anti-TLR2 antibody in murine peritoneal macrophages and resident fresh bone marrow cells has been investigated here. TLR2 blocking before infection induces the release of interleukin (IL)-10 by macrophages thereby inhibiting excessive production of oxidants by activating antioxidant enzymes. TLR2-blocked peritoneal macrophages showed impaired release of tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ) and IL-6 in response to both live and heat-killed S. aureus infection except bone marrow cells. TLR2-mediated free radical production and killing of S. aureus were modulated by TLR2 blocking in peritoneal macrophages and resident bone marrow cells. This study supported that S. aureus persists in resident bone marrow cells in a state of quiescence.


Asunto(s)
Células de la Médula Ósea/metabolismo , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Staphylococcus aureus , Receptor Toll-Like 2/fisiología , Animales , Células de la Médula Ósea/microbiología , Células Cultivadas , Macrófagos Peritoneales/microbiología , Ratones , Infecciones Estafilocócicas/metabolismo , Receptor Toll-Like 2/antagonistas & inhibidores
9.
Inflammation ; 38(3): 1050-69, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25429758

RESUMEN

We hypothesized that if internalization of Staphylococcus aureus could be blocked by using cytochalasin D (an inhibitor of phagocytosis and phagolysosome fusion), then the intracellular entry and survival of the pathogen in host's phagocytic cells recruited to the inflammatory site can be restricted. At the same time, if we use antimicrobial agents (e.g., ciprofloxacin and azithromycin) having potent intracellular and extracellular microbicidal activity against the bacterium that have not entered into the phagosome and remains adhered to the phagocytic cell membrane, then they can be eradicated from the site of infection without compromising the host cell. To validate this, role of ciprofloxacin (CIP) and azithromycin (AZM) in eliminating S. aureus by suppressing the phagocytic activity of macrophages with cytochalasin D before infection was investigated. CIP and AZM were used either alone or in combination with cytochalasin D. Supernatant and lysate obtained from the culture of macrophages were used for quantification of reactive oxygen species, lysozymes, antioxidant enzymes, and cytokines produced. Azithromycin was better than ciprofloxacin in combination with cytochalasin D for eradicating S. aureus and regulating cytokine release. Further studies are required for ensuring proper delivery of this combination at the site of infection.


Asunto(s)
Azitromicina/uso terapéutico , Ciprofloxacina/uso terapéutico , Citocalasina D/uso terapéutico , Macrófagos/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Antibacterianos/uso terapéutico , Antiinflamatorios/uso terapéutico , Antioxidantes/metabolismo , Quimioterapia Combinada , Glutatión/metabolismo , Peróxido de Hidrógeno/metabolismo , Inflamación/tratamiento farmacológico , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Muramidasa/metabolismo , Fagocitosis/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/inmunología , Staphylococcus aureus/aislamiento & purificación , Staphylococcus aureus/patogenicidad , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA