Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Appl Microbiol Biotechnol ; 107(14): 4567-4580, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37284893

RESUMEN

Acinetobacter baumannii is a Gram-negative bacterial pathogen that exhibits high intrinsic resistance to antimicrobials, with treatment often requiring the use of last-resort antibiotics. Antibiotic-resistant strains have become increasingly prevalent, underscoring a need for new therapeutic interventions. The aim of this study was to use A. baumannii outer membrane vesicles as immunogens to generate single-domain antibodies (VHHs) against bacterial cell surface targets. Llama immunization with the outer membrane vesicle preparations from four A. baumannii strains (ATCC 19606, ATCC 17961, ATCC 17975, and LAC-4) elicited a strong heavy-chain IgG response, and VHHs were selected against cell surface and/or extracellular targets. For one VHH, OMV81, the target antigen was identified using a combination of gel electrophoresis, mass spectrometry, and binding studies. Using these techniques, OMV81 was shown to specifically recognize CsuA/B, a protein subunit of the Csu pilus, with an equilibrium dissociation constant of 17 nM. OMV81 specifically bound to intact A. baumannii cells, highlighting its potential use as a targeting agent. We anticipate the ability to generate antigen-specific antibodies against cell surface A. baumannii targets could provide tools for further study and treatment of this pathogen. KEY POINTS: •Llama immunization with bacterial OMV preparations for VHH generation •A. baumannii CsuA/B, a pilus subunit, identified by mass spectrometry as VHH target •High-affinity and specific VHH binding to CsuA/B and A. baumannii cells.


Asunto(s)
Acinetobacter baumannii , Camélidos del Nuevo Mundo , Animales , Acinetobacter baumannii/metabolismo , Membrana Celular/metabolismo , Antibacterianos/metabolismo , Proteínas de la Membrana/metabolismo
2.
Environ Chem Lett ; 19(4): 2789-2808, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33907538

RESUMEN

The iron dependence of antibiotic-resistant microbes represents an Achilles' heel that can be exploited broadly. The growing global problem of antibiotic resistance of microbial pathogens wherein microbes become resistant to the very antibiotics used against them during infection is linked not only to our health uses but also to agribusiness practices and the changing environment. Here we review mechanisms of microbial iron acquisition and host iron withdrawal defense, and the influence of iron withdrawal on the antimicrobial activity of antibiotics. Antibiotic-resistant microbes are unaltered in their iron requirements, but iron withdrawal from microbes enhances the activities of various antibiotics and importantly suppresses outgrowth of antibiotic-exposed resistant microbial survivors. Of the three therapeutic approaches available to exploit microbial iron susceptibility, including (1) use of gallium as a non-functional iron analogue, (2) Trojan horse conjugates of microbial siderophores carrying antibiotics, and (3) new generation iron chelators, purposely designed as anti-microbials, the latter offers various advantages. For instance, these novel anti-microbial chelators overcome the limitations of conventional clinically-used hematological chelators which display host toxicity and are not useful antimicrobials. 3-Hydroxypyridin-4-one-containing polymeric chelators appear to have the highest potential. DIBI (developmental code name) is a well-developed lead candidate, being a low molecular weight, water-soluble copolymer with enhanced iron binding characteristics, strong anti-microbial and anti-inflammatory activities, low toxicity for animals and demonstrated freedom from microbial resistance development. DIBI has been shown to enhance antibiotic efficacy for antibiotic-resistant microbes during infection, and it also prevents recovery growth and resistance development during microbe exposure to various antibiotics. Because DIBI bolsters innate iron withdrawal defenses of the infected host, it has potential to provide a host-directed anti-infective therapy.

3.
Microb Pathog ; 145: 104229, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32353579

RESUMEN

Helicobacter pylori infection and associated diseases remain a major public health problem worldwide. Much effort has been made over the last several decades in vaccine development, but there is no licensed vaccine on the market. We have previously reported that oral immunization with H. pylori lysates and double mutant heat-labile toxin (dmLT) affords prophylactic protection against H. pylori infection in mice. In the present study, we investigated the effects of oral immunization with recombinant H. pylori protein antigens (NAP/UreA/UreB) and dmLT on H. pylori challenge in BALB/c mice. We found that oral immunization with candidate antigens and dmLT significantly reduced the gastric colonization of H. pylori 6 weeks after challenge, as compared to unimmunized mice. Moreover, the subunit vaccine appeared to provide a better protection than the bacterial lysate vaccine. The immunized mice showed enhanced antigen-specific lymphocyte proliferation, and serum IgG and mucosal IgA responses. Furthermore, the immunization increased the proportion of CD4+ IL-17+ lymphocytes in spleen and mesenteric lymph nodes, and enhanced the production of IL-17, IL-16, IL-6 and TNF-α in lymphocyte culture supernatants. Taken together, our results suggest that oral vaccination with recombinant H. pylori antigens (NAP/UreA/UreB) and dmLT confers more effective prophylactic protection against H. pylori infection than whole bacterial lysates in BALB/c mice. The reduction of H. pylori colonization was associated with the induction of antigen-specific Th17 and local mucosal IgA immune responses.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Adyuvantes Inmunológicos , Animales , Anticuerpos Antibacterianos , Antígenos Bacterianos/genética , Vacunas Bacterianas , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/genética , Calor , Inmunización , Ratones , Ratones Endogámicos BALB C , Vacunación
4.
Infect Immun ; 87(11)2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31405959

RESUMEN

Porcine mucin has been commonly used to enhance the infectivity of bacterial pathogens, including Acinetobacter baumannii, in animal models, but the mechanisms for enhancement by mucin remain relatively unknown. In this study, using the mouse model of intraperitoneal (i.p.) mucin-enhanced A. baumannii infection, we characterized the kinetics of bacterial replication and dissemination and the host innate immune responses, as well as their potential contribution to mucin-enhanced bacterial virulence. We found that mucin, either admixed with or separately injected with the challenge bacterial inoculum, was able to enhance the tissue and blood burdens of A. baumannii strains of different virulence. Intraperitoneal injection of A. baumannii-mucin or mucin alone induced a significant but comparable reduction of peritoneal macrophages and lymphocytes, accompanied by a significant neutrophil recruitment and early interleukin-10 (IL-10) responses, suggesting that the resulting inflammatory cellular and cytokine responses were largely induced by the mucin. Depletion of peritoneal macrophages or neutralization of endogenous IL-10 activities showed no effect on the mucin-enhanced infectivity. However, pretreatment of mucin with iron chelator DIBI, but not deferoxamine, partially abolished its virulence enhancement ability, and replacement of mucin with iron significantly enhanced the bacterial burdens in the peritoneal cavity and lung. Taken together, our results favor the hypothesis that iron at least partially contributes to the mucin-enhanced infectivity of A. baumannii in this model.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/patogenicidad , Mucinas/metabolismo , Peritonitis/microbiología , Animales , Células 3T3 BALB , Femenino , Interleucina-10/metabolismo , Interleucina-10/farmacología , Macrófagos Peritoneales , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes , Organismos Libres de Patógenos Específicos , Virulencia
5.
Infect Immun ; 87(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718286

RESUMEN

Acinetobacter baumannii is an important nosocomial pathogen. Mechanisms that allow A. baumannii to cause human infection are still poorly understood. Iron is an essential nutrient for bacterial growth in vivo, and the multiplicity of iron uptake systems in A. baumannii suggests that iron acquisition contributes to the ability of A. baumannii to cause infection. In Gram-negative bacteria, active transport of ferrisiderophores and heme relies on the conserved TonB-ExbB-ExbD energy-transducing complex, while active uptake of ferrous iron is mediated by the Feo system. The A. baumannii genome invariably contains three tonB genes (tonB1, tonB2, and tonB3), whose role in iron uptake is poorly understood. Here, we generated A. baumannii mutants with knockout mutations in the feo and/or tonB gene. We report that tonB3 is essential for A. baumannii growth under iron-limiting conditions, whereas tonB1, tonB2, and feoB appear to be dispensable for ferric iron uptake. tonB3 deletion resulted in reduced intracellular iron content despite siderophore overproduction, supporting a key role of TonB3 in iron uptake. In contrast to the case for tonB1 and tonB2, the promoters of tonB3 and feo contain functional Fur boxes and are upregulated in iron-poor media. Both TonB3 and Feo systems are required for growth in complement-free human serum and contribute to resistance to the bactericidal activity of normal human serum, but only TonB3 appears to be essential for virulence in insect and mouse models of infection. Our findings highlight a central role of the TonB3 system for A. baumannii pathogenicity. Hence, TonB3 represents a promising target for novel antibacterial therapies and for the generation of attenuated vaccine strains.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/patogenicidad , Proteínas de Transporte de Catión/metabolismo , Hierro/metabolismo , Acinetobacter baumannii/genética , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico Activo , Proteínas de Transporte de Catión/genética , Femenino , Hemo/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Sideróforos/metabolismo , Virulencia
6.
Artículo en Inglés | MEDLINE | ID: mdl-31209004

RESUMEN

Acinetobacter baumannii is a major cause of nosocomial infections especially hospital-acquired pneumonia. This bacterium readily acquires antibiotic resistance traits and therefore, new treatment alternatives are urgently needed. The virulence of A. baumannii linked to iron acquisition suggests a potential for new anti-infectives that target its iron acquisition. DIBI, a 3-hydroxypyridin-4-one chelator, is a purpose-designed, iron-sequestering antimicrobial that has shown promise for treating microbial infection. DIBI was investigated for its in vitro and in vivo activities against clinical A. baumannii isolates. DIBI was inhibitory for all isolates tested with very low MICs (2 µg/ml, equivalent to 0.2 µM), i.e., at or below the typical antibiotic MICs reported for antibiotic-sensitive strains. DIBI inhibition is Fe specific, and it caused an iron-restricted bacterial physiology that led to enhanced antibiotic killing by several discrete antibiotics. DIBI also strongly suppressed recovery growth of the surviving population following antibiotic exposure. A low intranasal dose (11 µmol/kg) of DIBI after intranasal challenge with hypervirulent ciprofloxacin (CIP)-resistant A. baumannii LAC-4 significantly reduced bacterial burdens in mice, and DIBI also suppressed the spread of the infection to the spleen. Treatment of infected mice with CIP alone (20 mg/kg, equivalent to 60 µmol/kg) was ineffective given LAC-4's CIP resistance, but if combined with DIBI, the treatment efficacy improved significantly. Our evidence suggests that DIBI restricts host iron availability to A. baumannii growing in the respiratory tract, bolstering the host innate iron restriction mechanisms. DIBI has potential as a sole anti-infective or in combination with conventional antibiotics for the treatment of A. baumannii pneumonia.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Hierro/metabolismo , Neumonía/tratamiento farmacológico , Neumonía/microbiología , Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/patogenicidad , Animales , Quimiocinas/metabolismo , Ciprofloxacina/farmacología , Ciprofloxacina/uso terapéutico , Citocinas/metabolismo , Farmacorresistencia Bacteriana Múltiple , Femenino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Neumonía/metabolismo , Virulencia
8.
BMC Infect Dis ; 16: 194, 2016 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-27146090

RESUMEN

BACKGROUND: Antibiotic resistant strains of Acinetobacter baumannii have been responsible for an increasing number of nosocomial infections including bacteremia and ventilator-associated pneumonia. In this study, we analyzed 38 isolates of A. baumannii obtained from two hospital outbreaks in Los Angeles County for the molecular epidemiology, antimicrobial susceptibility and resistance determinants. METHODS: Pulsed field gel electrophoresis, tri-locus multiplex PCR and multi-locus sequence typing (Pasteur scheme) were used to examine clonal relationships of the outbreak isolates. Broth microdilution method was used to determine antimicrobial susceptibility of these isolates. PCR and subsequent DNA sequencing were employed to characterize antibiotic resistance genetic determinants. RESULTS: Trilocus multiplex PCR showed these isolates belong to Global Clones I and II, which were confirmed to ST1 and ST2, respectively, by multi-locus sequence typing. Pulsed field gel electrophoresis analysis identified two clonal clusters, one with 20 isolates (Global Clone I) and the other with nine (Global Clone II), which dominated the two outbreaks. Antimicrobial susceptibility testing using 14 antibiotics indicated that all isolates were resistant to antibiotics belonging to four or more categories of antimicrobial agents. In particular, over three fourth of 38 isolates were found to be resistant to both imipenem and meropenem. Additionally, all isolates were found to be resistant to piperacillin, four cephalosporin antibiotics, ciprofloxacin and levofloxacin. Resistance phenotypes of these strains to fluoroquinolones were correlated with point mutations in gyrA and parC genes that render reduced affinity to target proteins. ISAba1 was detected immediately upstream of the bla OXA-23 gene present in those isolates that were found to be resistant to both carbapenems. Class 1 integron-associated resistance gene cassettes appear to contribute to resistance to aminoglycoside antibiotics. CONCLUSION: The two outbreaks were found to be dominated by two clonal clusters of A. baumannii belonging to MLST ST1 and ST2. All isolates were resistant to antibiotics of at least four categories of antimicrobial agents, and their antimicrobial susceptibility profiles correlate well with genetic determinants. The results of this study will facilitate our understanding of the molecular epidemiology, antimicrobial susceptibility and mechanisms of resistance of A. baumannii obtained from Los Angeles hospitals.


Asunto(s)
Infecciones por Acinetobacter/epidemiología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/genética , Antibacterianos/farmacología , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/aislamiento & purificación , Proteínas Bacterianas/genética , California , Infección Hospitalaria/epidemiología , Infección Hospitalaria/microbiología , Girasa de ADN/genética , Topoisomerasa de ADN IV , Brotes de Enfermedades , Farmacorresistencia Bacteriana Múltiple/genética , Electroforesis en Gel de Campo Pulsado , Humanos , Integrones , Unidades de Cuidados Intensivos , Pruebas de Sensibilidad Microbiana , Tipificación de Secuencias Multilocus , beta-Lactamasas/genética
9.
J Infect Dis ; 211(8): 1296-305, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378635

RESUMEN

BACKGROUND: Acinetobacter baumannii is one of the most antibiotic-resistant pathogens. Defining mechanisms driving pathogenesis is critical to enable new therapeutic approaches. METHODS: We studied virulence differences across a diverse panel of A. baumannii clinical isolates during murine bacteremia to elucidate host-microbe interactions that drive outcome. RESULTS: We identified hypervirulent strains that were lethal at low intravenous inocula and achieved very high early, and persistent, blood bacterial densities. Virulent strains were nonlethal at low inocula but lethal at 2.5-fold higher inocula. Finally, relatively avirulent (hypovirulent) strains were nonlethal at 20-fold higher inocula and were efficiently cleared by early time points. In vivo virulence correlated with in vitro resistance to complement and macrophage uptake. Depletion of complement, macrophages, and neutrophils each independently increased bacterial density of the hypovirulent strain but insufficiently to change lethality. However, disruption of all 3 effector mechanisms enabled early bacterial densities similar to hypervirulent strains, rendering infection 100% fatal. CONCLUSIONS: The lethality of A. baumannii strains depends on distinct stages. Strains resistant to early innate effectors are able to establish very high early bacterial blood density, and subsequent sustained bacteremia leads to Toll-like receptor 4-mediated hyperinflammation and lethality. These results have important implications for translational efforts to develop therapies that modulate host-microbe interactions.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Acinetobacter baumannii/inmunología , Bacteriemia/inmunología , Inmunidad Innata/inmunología , Interacciones Microbianas/inmunología , Infecciones por Acinetobacter/microbiología , Animales , Antibacterianos/inmunología , Bacteriemia/microbiología , Farmacorresistencia Bacteriana Múltiple/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C3H , Neutrófilos/inmunología , Neutrófilos/microbiología , Virulencia/inmunología , Factores de Virulencia/inmunología
10.
Antimicrob Agents Chemother ; 59(9): 5654-63, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26149990

RESUMEN

We previously demonstrated that ginsenosides Rg1 and Re enhanced the immune response in C3H/HeB mice but not in C3H/HeJ mice carrying a mutation in the Tlr4 gene. The results of the present study showed that both Rg1 and Re inhibited mRNA expression and production of proinflammatory mediators that included tumor necrosis factor α, interleukin-1ß, interleukin-6, cyclooxygenase-2, and inducible nitric oxide synthase from lipopolysaccharide (LPS)-stimulated macrophages. Rg1 was found to be distributed both extracellularly and intracellularly but Re was located only extracellularly to compete with LPS for binding to Toll-like receptor 4. Preinjection of Rg1 and Re into rats suppressed LPS-induced increases in body temperature, white blood cell counts, and levels of serum proinflammatory mediators. Preinjection of Rg1 and Re into mice prevented the LPS-induced decreases in total white blood cell counts and neutrophil counts, inhibited excessive expression of multiple proinflammatory mediators, and successfully rescued 100% of the mice from sepsis-associated death. More significantly, when administered after lethal LPS inoculation, Rg1, but not Re, still showed a potent antisepsis effect and protected 90% of the mice from death. The better protection efficacy of Rg1 could result from its intracellular distribution, suggesting that Rg1 may be an ideal antisepsis agent.


Asunto(s)
Ginsenósidos/metabolismo , Lipopolisacáridos/toxicidad , Sepsis/inducido químicamente , Sepsis/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Ciclooxigenasa 2/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Int J Med Microbiol ; 304(3-4): 360-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24440358

RESUMEN

Bacteremia caused by Acinetobacter baumannii is a highly lethal complication of hospital-acquired pneumonia. In the present study, we investigated the serum resistance, gallium nitrate tolerance and heme consumption of A. baumannii strain LAC-4 which was recently reported to display high virulence in a mouse pneumonia model with extrapulmonary dissemination leading to fatal bacteremia. This strain showed enhanced growth in mouse and fetal bovine serum that was independent of complement and was not observed with regular growth media. The LAC-4 strain was found to possess a high tolerance to gallium nitrate (GaN), whereas serum synergized with GaN in inhibiting A. baumannii strain ATCC 17978. We found that LAC-4 contains a heme oxygenase gene and expresses a highly efficient heme consumption system. This system can be fully blocked in vitro and in vivo by gallium protoporphyrin IX (GaPPIX). Inhibition of heme consumption by GaPPIX completely abrogated the growth advantage of LAC-4 in serum as well as its tolerance to GaN. More importantly, GaPPIX treatment of mice intranasally infected with LAC-4 prevented extrapulmonary dissemination and death. Thus, we propose that heme provides an additional source of iron for LAC-4 to bypass iron restriction caused by serum transferrin, lactoferrin or free gallium salts. Heme consumption systems in A. baumannii may constitute major virulence factors for lethal bacteremic isolates.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/metabolismo , Bacteriemia/microbiología , Actividad Bactericida de la Sangre , Galio/toxicidad , Hemo/metabolismo , Neumonía Bacteriana/microbiología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/inmunología , Acinetobacter baumannii/aislamiento & purificación , Animales , Bovinos , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Femenino , Humanos , Ratones Endogámicos C57BL , Neumonía Bacteriana/complicaciones , Suero/microbiología
12.
Front Microbiol ; 15: 1230997, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690360

RESUMEN

A rapid increase in antimicrobial resistant bacterial infections around the world is causing a global health crisis. The Gram-negative bacterium Acinetobacter baumannii is categorized as a Priority 1 pathogen for research and development of new antimicrobials by the World Health Organization due to its numerous intrinsic antibiotic resistance mechanisms and ability to quickly acquire new resistance determinants. Specialized phage enzymes, called depolymerases, degrade the bacterial capsule polysaccharide layer and show therapeutic potential by sensitizing the bacterium to phages, select antibiotics, and serum killing. The functional domains responsible for the capsule degradation activity are often found in the tail fibers of select A. baumannii phages. To further explore the functional domains associated with depolymerase activity, tail-associated proteins of 71 sequenced and fully characterized phages were identified from published literature and analyzed for functional domains using InterProScan. Multisequence alignments and phylogenetic analyses were conducted on the domain groups and assessed in the context of noted halo formation or depolymerase characterization. Proteins derived from phages noted to have halo formation or a functional depolymerase, but no functional domain hits, were modeled with AlphaFold2 Multimer, and compared to other protein models using the DALI server. The domains associated with depolymerase function were pectin lyase-like (SSF51126), tailspike binding (cd20481), (Trans)glycosidases (SSF51445), and potentially SGNH hydrolases. These findings expand our knowledge on phage depolymerases, enabling researchers to better exploit these enzymes for therapeutic use in combating the antimicrobial resistance crisis.

13.
Antimicrob Agents Chemother ; 57(8): 3601-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23689726

RESUMEN

Acinetobacter baumannii is an important emerging pathogen in health care-acquired infections and is responsible for severe nosocomial and community-acquired pneumonia. Currently available mouse models of A. baumannii pneumonia show poor colonization with little to no extrapulmonary dissemination. Here, we describe a mouse model of A. baumannii pneumonia using a clinical isolate (LAC-4 strain) that reliably reproduces the most relevant features of human pulmonary A. baumannii infection and pathology. Using this model, we have shown that LAC-4 infection induced rapid bacterial replication in the lungs, significant extrapulmonary dissemination, and severe bacteremia by 24 h postintranasal inoculation. Infected mice showed severe bronchopneumonia and dilatation and inflammatory cell infiltration in the perivascular space. More significantly, 100% of C57BL/6 and BALB/c mice succumbed to 10(8) CFU of LAC-4 inoculation within 48 h. When this model was used to assess the efficacy of antimicrobials, all mice treated with imipenem and tigecycline survived a lethal intranasal challenge, with minimal clinical signs and body weight loss. Moreover, intranasal immunization of mice with formalin-fixed LAC-4 protected 40% of mice from a lethal (100× 100% lethal dose) intraperitoneal challenge. Thus, this model offers a reproducible acute course of A. baumannii pneumonia without requiring additional manipulation of host immune status, which will facilitate the development of therapeutic agents and vaccines against A. baumannii pneumonia in humans.


Asunto(s)
Acinetobacter baumannii/inmunología , Vacunas Bacterianas/inmunología , Modelos Animales de Enfermedad , Neumonía Bacteriana/prevención & control , Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/prevención & control , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/patogenicidad , Animales , Antibacterianos/farmacología , Bacteriemia/inmunología , Bacteriemia/prevención & control , Vacunas Bacterianas/administración & dosificación , Peso Corporal/efectos de los fármacos , Bronconeumonía/inmunología , Bronconeumonía/microbiología , Femenino , Imipenem/farmacología , Inmunización/métodos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Minociclina/análogos & derivados , Minociclina/farmacología , Neumonía Bacteriana/inmunología , Reproducibilidad de los Resultados , Tigeciclina , Factores de Tiempo
14.
Nanomedicine (Lond) ; 18(5): 471-484, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-37170884

RESUMEN

Translation of antibacterial nanoparticles into nanomedicine requires a deep understanding of the dynamic nature of nanoparticles and the ways they overcome immunological and biological barriers. Nanomedicines need prolonged serum stability by proper stealth coating or forming beneficial protein corona, to avoid rapid clearance by the mononuclear phagocytic system. A preferred nanoparticle formulation may include nonimmunogenic carbohydrates, which act both as a stealth coating and ligands of specific endothelium receptors to facilitate nanomedicines crossing the vascular barrier. This may lead to more rapid delivery and accumulation of nanomedicine at the infection site and provide broader and faster clinical responses than targeting specific bacterial surface receptors. Ideally, antibacterial nanomedicines should be able to penetrate biofilms through fusion and/or diffusion for targeted delivery.


Asunto(s)
Nanopartículas , Corona de Proteínas , Nanomedicina , Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Nanopartículas/metabolismo , Sistemas de Liberación de Medicamentos
15.
Emerg Microbes Infect ; 12(2): 2272656, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37855122

RESUMEN

Pneumococcal disease is a major threat to public health globally, impacting individuals across all age groups, particularly infants and elderly individuals. The use of current vaccines has led to unintended consequences, including serotype replacement, leading to a need for a new approach to combat pneumococcal disease. A promising solution is the development of a broad-spectrum pneumococcal vaccine. In this study, we present the development of a broad-spectrum protein-based pneumococcal vaccine that contains three pneumococcal virulence factors: rlipo-PsaA (lipidated form), rPspAΔC (truncated form), and rPspCΔC (truncated form). Intranasal immunization with rlipo-PsaA, rPspAΔC, and rPspCΔC (LAAC) resulted in significantly higher IgG titres than those induced by administration of nonlipidated rPsaA, rPspAΔC, and rPspCΔC (AAC). Furthermore, LAAC immunization induced the production of higher IgA titres in vaginal washes, feces, and sera in mice, indicating that LAAC can induce systemic mucosal immunity. In addition, administration of LAAC also induced Th1/Th17-biased immune responses and promoted opsonic phagocytosis of Streptococcus pneumoniae strains of various serotypes, implying that the immunogenicity of LAAC immunization provides a protective effect against pneumococcal infection. Importantly, challenge data showed that the LAAC-immunized mice had a reduced bacterial load not only for several serotypes of the 13-valent conjugate pneumococcal vaccine (PCV13) but also for selected non-PCV13 serotypes. Consistently, LAAC immunization increased the survival rate of mice after bacterial challenge with both PCV13 and non-PCV13 serotypes. In conclusion, our protein-based pneumococcal vaccine provides protective effects against a broad spectrum of Streptococcus pneumoniae serotypes.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Humanos , Lactante , Femenino , Ratones , Animales , Anciano , Inmunidad Mucosa , Vacunas Neumococicas , Infecciones Neumocócicas/microbiología , Inmunización , Anticuerpos Antibacterianos
16.
Viruses ; 15(3)2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36992448

RESUMEN

The world is currently facing a global health crisis due to the rapid increase in antimicrobial-resistant bacterial infections. One of the most concerning pathogens is Acinetobacter baumannii, which is listed as a Priority 1 pathogen by the World Health Organization. This Gram-negative bacterium has many intrinsic antibiotic resistance mechanisms and the ability to quickly acquire new resistance determinants from its environment. A limited number of effective antibiotics against this pathogen complicates the treatment of A. baumannii infections. A potential treatment option that is rapidly gaining interest is "phage therapy", or the clinical application of bacteriophages to selectively kill bacteria. The myoviruses DLP1 and DLP2 (vB_AbaM-DLP_1 and vB_AbaM-DLP_2, respectively) were isolated from sewage samples using a capsule minus variant of A. baumannii strain AB5075. Host range analysis of these phages against 107 A. baumannii strains shows a limited host range, infecting 15 and 21 for phages DLP1 and DLP2, respectively. Phage DLP1 has a large burst size of 239 PFU/cell, a latency period of 20 min, and virulence index of 0.93. In contrast, DLP2 has a smaller burst size of 24 PFU/cell, a latency period of 20 min, and virulence index of 0.86. Both phages show potential for use as therapeutics to combat A. baumannii infections.


Asunto(s)
Acinetobacter baumannii , Bacteriófagos , Bacteriófagos/genética , Especificidad del Huésped , Antibacterianos
17.
Front Med (Lausanne) ; 10: 1134912, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37359001

RESUMEN

Background: Biofilm formation is a major clinical challenge contributing to treatment failure of periprosthetic joint infection (PJI). Lytic bacteriophages (phages) can target biofilm associated bacteria at localized sites of infection. The aim of this study is to investigate whether combination therapy of phage and vancomycin is capable of clearing Staphylococcus aureus biofilm-like aggregates formed in human synovial fluid. Methods: In this study, S. aureus BP043, a PJI clinical isolate was utilized. This strain is a methicillin-resistant S. aureus (MRSA) biofilm-former. Phage Remus, known to infect S. aureus, was selected for the treatment protocol. BP043 was grown as aggregates in human synovial fluid. The characterization of S. aureus aggregates was assessed for structure and size using scanning electron microscopy (SEM) and flow cytometry, respectively. Moreover, the formed aggregates were subsequently treated in vitro with: (a) phage Remus [∼108 plaque-forming units (PFU)/ml], (b) vancomycin (500 µg/ml), or (c) phage Remus (∼108 PFU/ml) followed by vancomycin (500 µg/ml), for 48 h. Bacterial survival was quantified by enumeration [colony-forming units (CFU)/ml]. The efficacy of phage and vancomycin against BP043 aggregates was assessed in vivo as individual treatments and in combination. The in vivo model utilized Galleria mellonella larvae which were infected with BP043 aggregates pre-formed in synovial fluid. Results: Scanning electron microscopy (SEM) images and flow cytometry data demonstrated the ability of human synovial fluid to promote formation of S. aureus aggregates. Treatment with Remus resulted in significant reduction in viable S. aureus residing within the synovial fluid aggregates compared to the aggregates that did not receive Remus (p < 0.0001). Remus was more efficient in eliminating viable bacteria within the aggregates compared to vancomycin (p < 0.0001). Combination treatment of Remus followed by vancomycin was more efficacious in reducing bacterial load compared to using either Remus or vancomycin alone (p = 0.0023, p < 0.0001, respectively). When tested in vivo, this combination treatment also resulted in the highest survival rate (37%) 96 h post-treatment, compared to untreated larvae (3%; p < 0.0001). Conclusion: We demonstrate that combining phage Remus and vancomycin led to synergistic interaction against MRSA biofilm-like aggregates in vitro and in vivo.

18.
Emerg Microbes Infect ; 12(1): 2192821, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36927227

RESUMEN

Influenza and Respiratory Syncytial virus (RSV) infections together contribute significantly to the burden of acute lower respiratory tract infections. Despite the disease burden, no approved RSV vaccine is available. While approved vaccines are available for influenza, seasonal vaccination is required to maintain protection. In addition to both being respiratory viruses, they follow a common seasonality, which warrants the necessity for a concerted vaccination approach. Here, we designed bivalent vaccines by utilizing highly conserved sequences, targeting both influenza A and RSV, as either a chimeric antigen or individual antigens separated by a ribosome skipping sequence. These vaccines were found to be effective in protecting the animals from challenge by either virus, with mechanisms of protection being substantially interrogated in this communication.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones por Virus Sincitial Respiratorio , Ratones , Animales , Humanos , Virus Sincitiales Respiratorios/genética , Vacunas Combinadas , Anticuerpos Antivirales , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra la Influenza/genética , Anticuerpos Neutralizantes
19.
Front Immunol ; 14: 1020134, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37006299

RESUMEN

Introduction: The incidence of Lyme disease (LD) in Canada and the United States has risen over the last decade, nearing 480,000 cases each year. Borrelia burgdorferi sensu lato, the causative agent of LD, is transmitted to humans through the bite of an infected tick, resulting in flu-like symptoms and often a characteristic bull's-eye rash. In more severe cases, disseminated bacterial infection can cause arthritis, carditis and neurological impairments. Currently, no vaccine is available for the prevention of LD in humans. Methods: In this study, we developed a lipid nanoparticle (LNP)-encapsulated DNA vaccine encoding outer surface protein C type A (OspC-type A) of B. burgdorferi. Results: Vaccination of C3H/HeN mice with two doses of the candidate vaccine induced significant OspC-type A-specific antibody titres and borreliacidal activity. Analysis of the bacterial burden following needle challenge with B. burgdorferi (OspC-type A) revealed that the candidate vaccine afforded effective protection against homologous infection across a range of susceptible tissues. Notably, vaccinated mice were protected against carditis and lymphadenopathy associated with Lyme borreliosis. Discussion: Overall, the results of this study provide support for the use of a DNA-LNP platform for the development of LD vaccines.


Asunto(s)
Borrelia burgdorferi , Enfermedad de Lyme , Miocarditis , Vacunas de ADN , Humanos , Ratones , Animales , Vacunas Bacterianas , Ratones Endogámicos C3H , ADN
20.
Infect Immun ; 80(10): 3521-32, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22851750

RESUMEN

We show in this study that toxin production in Clostridium difficile is altered in cells which can no longer form flagellar filaments. The impact of inactivation of fliC, CD0240, fliF, fliG, fliM, and flhB-fliR flagellar genes upon toxin levels in culture supernatants was assessed using cell-based cytotoxicity assay, proteomics, immunoassay, and immunoblotting approaches. Each of these showed that toxin levels in supernatants were significantly increased in a fliC mutant compared to that in the C. difficile 630 parent strain. In contrast, the toxin levels in supernatants secreted from other flagellar mutants were significantly reduced compared with that in the parental C. difficile 630 strain. Transcriptional analysis of the pathogenicity locus genes (tcdR, tcdB, tcdE, and tcdA) revealed a significant increase of all four genes in the fliC mutant strain, while transcription of all four genes was significantly reduced in fliM, fliF, fliG, and flhB-fliR mutants. These results demonstrate that toxin transcription in C. difficile is modulated by the flagellar regulon. More significantly, mutant strains showed a corresponding change in virulence compared to the 630 parent strain when tested in a hamster model of C. difficile infection. This is the first demonstration of differential flagellum-related transcriptional regulation of toxin production in C. difficile and provides evidence for elaborate regulatory networks for virulence genes in C. difficile.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Flagelos/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Factor sigma/metabolismo , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Femenino , Flagelos/genética , Mutación , Proteómica , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Factor sigma/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA