Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 196(4): 1105-1110, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34726258

RESUMEN

Transfusion of storage-damaged red blood cells (RBCs) increases non-transferrin-bound iron (NTBI) levels in humans. This can potentially enhance virulence of microorganisms. In this study, Pseudomonas aeruginosa replication and biofilm production in vitro correlated with NTBI levels of transfused subjects (R2 = 0·80; P < 0·0001). Transfusion of stored RBCs into catheterized mice enhanced P. aeruginosa virulence and mortality in vivo, while pre-administration of apotransferrin reduced NTBI levels improving survival (69% vs 27% mortality; P < 0·05). These results suggest that longer RBC storage, by modulating the bioavailability of iron, may increase the risk of P. aeruginosa biofilm-related infections in transfused patients.


Asunto(s)
Transfusión de Eritrocitos/métodos , Eritrocitos/metabolismo , Hierro/sangre , Animales , Biopelículas , Transfusión de Eritrocitos/mortalidad , Voluntarios Sanos , Humanos , Masculino , Ratones , Pseudomonas aeruginosa , Análisis de Supervivencia
2.
J Shoulder Elbow Surg ; 29(7): 1412-1424, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32014357

RESUMEN

BACKGROUND: Animal models are used to guide management of periprosthetic implant infections. No adequate model exists for periprosthetic shoulder infections, and clinicians thus have no preclinical tools to assess potential therapeutics. We hypothesize that it is possible to establish a mouse model of shoulder implant infection (SII) that allows noninvasive, longitudinal tracking of biofilm and host response through in vivo optical imaging. The model may then be employed to validate a targeting probe (1D9-680) with clinical translation potential for diagnosing infection and image-guided débridement. METHODS: A surgical implant was press-fit into the proximal humerus of c57BL/6J mice and inoculated with 2 µL of 1 × 103 (e3), or 1 × 104 (e4), colony-forming units (CFUs) of bioluminescent Staphylococcus aureus Xen-36. The control group received 2 µL sterile saline. Bacterial activity was monitored in vivo over 42 days, directly (bioluminescence) and indirectly (targeting probe). Weekly radiographs assessed implant loosening. CFU harvests, confocal microscopy, and histology were performed. RESULTS: Both inoculated groups established chronic infections. CFUs on postoperative day (POD) 42 were increased in the infected groups compared with the sterile group (P < .001). By POD 14, osteolysis was visualized in both infected groups. The e4 group developed catastrophic bone destruction by POD 42. The e3 group maintained a congruent shoulder joint. Targeting probes helped to visualize low-grade infections via fluorescence. DISCUSSION: Given bone destruction in the e4 group, a longitudinal, noninvasive mouse model of SII and chronic osteolysis was produced using e3 of S aureus Xen-36, mimicking clinical presentations of chronic SII. CONCLUSION: The development of this model provides a foundation to study new therapeutics, interventions, and host modifications.


Asunto(s)
Complicaciones Posoperatorias/microbiología , Infecciones Relacionadas con Prótesis/etiología , Articulación del Hombro , Prótesis de Hombro/efectos adversos , Infecciones Estafilocócicas/microbiología , Animales , Biopelículas , Desbridamiento , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Staphylococcus aureus
3.
Methods ; 127: 62-68, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28522324

RESUMEN

In contrast to two-dimensional bioluminescence imaging, three dimensional diffuse light imaging tomography with integrated micro-computed tomography (DLIT-µCT) has the potential to realise spatial variations in infection patterns when imaging experimental animals dosed with derivatives of virulent bacteria carrying bioluminescent reporter genes such as the lux operon from the bacterium Photorhabdus luminescens. The method provides an opportunity to precisely localise the bacterial infection sites within the animal and enables the generation of four-dimensional movies of the infection cycle. Here, we describe the use of the PerkinElmer IVIS SpectrumCT in vivo imaging system to investigate progression of lethal systemic infection in neonatal rats following colonisation of the gastrointestinal tract with the neonatal pathogen Escherichia coli K1. We confirm previous observations that these bacteria stably colonize the colon and small intestine following feeding of the infectious dose from a micropipette; invading bacteria migrate across the gut epithelium into the blood circulation and establish foci of infection in major organs, including the brain. DLIT-µCT revealed novel multiple sites of colonisation within the alimentary canal, including the tongue, oesophagus and stomach, with penetration of the non-keratinised oesophageal epithelial surface, providing strong evidence of a further major site for bacterial dissemination. We highlight technical issues associated with imaging of infections in new born rat pups and show that the whole-body and organ bioburden correlates with disease severity.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Escherichia coli/patología , Imagenología Tridimensional/métodos , Sepsis/patología , Factores de Edad , Animales , Animales Recién Nacidos , Progresión de la Enfermedad , Genes Reporteros , Mediciones Luminiscentes/métodos , Microorganismos Modificados Genéticamente/genética , Ratas , Tomografía Óptica/métodos , Microtomografía por Rayos X/métodos
4.
Antimicrob Agents Chemother ; 58(4): 2377-86, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24514089

RESUMEN

The incidence of infections related to cardiac devices (such as permanent pacemakers) has been increasing out of proportion to implantation rates. As management of device infections typically requires explantation of the device, optimal prophylactic strategies are needed. Cefazolin and vancomycin are widely used as single agents for surgical prophylaxis against cardiac device-related infections. However, combination antibiotic prophylaxis may further reduce infectious complications. To model a localized subcutaneous implant-related infection, a bioluminescent strain of Staphylococcus epidermidis was inoculated onto a medical-procedure-grade titanium disc, which was placed into a subcutaneous pocket in the backs of mice. In vivo bioluminescence imaging, quantification of ex vivo CFU from the capsules and implants, variable-pressure scanning electron microscopy (VP-SEM), and neutrophil enhanced green fluorescent protein (EGFP) fluorescence in LysEGFP mice were employed to monitor the infection. This model was used to evaluate the efficacies of low- and high-dose cefazolin (50 and 200 mg/kg of body weight) and vancomycin (10 and 110 mg/kg) intravenous prophylaxis with or without rifampin (25 mg/kg). High-dose cefazolin and high-dose vancomycin treatment resulted in almost complete bacterial clearance, whereas both low-dose cefazolin and low-dose vancomycin reduced the in vivo and ex vivo bacterial burden only moderately. The addition of rifampin to low-dose cefazolin and vancomycin was highly effective in further reducing the CFU harvested from the implants. However, vancomycin-rifampin was more effective than cefazolin-rifampin in further reducing the CFU harvested from the surrounding tissue capsules. Future studies in humans will be required to determine whether the addition of rifampin has improved efficacy in preventing device-related infections in clinical practice.


Asunto(s)
Cefazolina/farmacología , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Rifampin/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus epidermidis/efectos de los fármacos , Vancomicina/farmacología , Vancomicina/uso terapéutico , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Cefazolina/uso terapéutico , Masculino , Ratones
5.
Appl Environ Microbiol ; 80(10): 3007-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24610844

RESUMEN

In recent years, a greater appreciation for the microbes inhabiting human body sites has emerged. In the female mammary gland, milk has been shown to contain bacterial species, ostensibly reaching the ducts from the skin. We decided to investigate whether there is a microbiome within the mammary tissue. Using 16S rRNA sequencing and culture, we analyzed breast tissue from 81 women with and without cancer in Canada and Ireland. A diverse population of bacteria was detected within tissue collected from sites all around the breast in women aged 18 to 90, not all of whom had a history of lactation. The principal phylum was Proteobacteria. The most abundant taxa in the Canadian samples were Bacillus (11.4%), Acinetobacter (10.0%), Enterobacteriaceae (8.3%), Pseudomonas (6.5%), Staphylococcus (6.5%), Propionibacterium (5.8%), Comamonadaceae (5.7%), Gammaproteobacteria (5.0%), and Prevotella (5.0%). In the Irish samples the most abundant taxa were Enterobacteriaceae (30.8%), Staphylococcus (12.7%), Listeria welshimeri (12.1%), Propionibacterium (10.1%), and Pseudomonas (5.3%). None of the subjects had signs or symptoms of infection, but the presence of viable bacteria was confirmed in some samples by culture. The extent to which these organisms play a role in health or disease remains to be determined.


Asunto(s)
Bacterias/aislamiento & purificación , Mama/microbiología , Microbiota , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Bacterias/clasificación , Bacterias/genética , Biodiversidad , Canadá , Femenino , Humanos , Persona de Mediana Edad , Adulto Joven
6.
Transfusion ; 54(11): 2842-51, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24840185

RESUMEN

BACKGROUND: Although human red blood cell (RBC) units may be refrigerator stored for up to 42 days, transfusion of older RBCs acutely delivers a large bolus of iron to mononuclear phagocytes. Similarly, iron dextran circulates in plasma for hours to days and is progressively cleared by mononuclear phagocytes, which return iron to plasma. Finally, malaria infection continuously delivers iron to macrophages by intra- and extravascular hemolysis. Studies suggest that iron administration increases infectious risk. STUDY DESIGN AND METHODS: To assess the effects of increased iron availability on susceptibility to infection, we infected mice with model Gram-negative intracellular or extracellular pathogens (Salmonella typhimurium or Escherichia coli, respectively), accompanied by RBC transfusion, iron dextran administration, or malarial coinfection. RESULTS: In our mouse models, transfusion of older RBCs exacerbates infection with both Gram-negative pathogens. Although iron dextran exacerbates E. coli infection to a similar extent as transfusion of corresponding amounts of iron, higher iron doses are required to produce comparable effects with S. typhimurium. Coinfection of mice with Plasmodium yoelii and S. typhimurium produces overwhelming Salmonella sepsis. Finally, treating mice with antibiotics abrogates the enhancing effect on E. coli infection of both older RBC transfusion and iron dextran administration. CONCLUSIONS: Transfusion of older RBCs exacerbates Gram-negative infection to a similar extent as malaria coinfection or iron dextran administration. Appropriate antibiotic therapy abrogates the effect of older RBC transfusions on infection with E. coli. Iron delivery to macrophages may be an underappreciated mechanism mediating, at least some, adverse effects of RBC transfusions.


Asunto(s)
Conservación de la Sangre/efectos adversos , Transfusión de Eritrocitos , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Hierro/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Sepsis/inmunología , Animales , Modelos Animales de Enfermedad , Escherichia coli/metabolismo , Infecciones por Escherichia coli/etiología , Infecciones por Escherichia coli/metabolismo , Hemólisis , Humanos , Hierro/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Malaria/inmunología , Malaria/metabolismo , Masculino , Ratones , Plasmodium yoelii/inmunología , Plasmodium yoelii/metabolismo , Infecciones por Salmonella/etiología , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/metabolismo , Sepsis/etiología , Sepsis/metabolismo
7.
Pharm Res ; 31(11): 3073-84, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24831312

RESUMEN

PURPOSE: The objective of this study was to formulate nanoparticles of D-luciferin (Nano-Luc), DiR (Nano-DiR) and dual functional nanoparticles with DiR and luciferin (Nano-LucDiR) for in-vivo imaging as well as tracking of the nanoparticles in tumors. METHODS: Nano-Luc and Nano-LucDiR were prepared using different lipids, and subsequently characterized for loading and entrapment efficiency, physical properties, release profile, toxicity and stability. We utilized Response Surface Methodology (RSM) to optimize the nanoparticles using design of experiment (DOE Vr.8.0). Nano-Luc was evaluated against free luciferin to establish its pharmacokinetic parameters in mice. In-vivo imaging of tumors and tracking of nanoparticles was carried out with an IVIS® Spectrum-CT (Caliper) using xenograft, orthotopic and metastatic tumor models in BALB/c nude mice with different cell lines and different routes of nanoparticle administration (subcutaneous, intraperitoneal and intravenous). RESULTS: Particle size of both Nano-Luc and Nano-LucDiR were found to be <200 nm. Nano-Luc formulation showed a slow and controlled release upto 72 h (90%) in vitro. The optimized Nano-Luc had loading efficiency of 5.0 mg/ml with 99% encapsulation efficiency. Nano-Luc and Nano-LucDiR formulations had good shelf stability. Nano-Luc and Nano-LucDiR enhanced plasma half-life of luciferin compared to free luciferin thus providing longer circulation of luciferin in plasma enabling imaging of tumors for more than 24 h. Nano-LucDiR allowed simultaneous bioluminescent and fluorescent imaging to be conducted, with three-dimensional reconstruct of tumors without losing either signal during the acquisition time. CONCLUSION: Nano-Luc and Nano-LucDiR allowed prolonged reproducible in-vivo imaging of tumors, especially during multimodality 3D imaging.


Asunto(s)
Benzotiazoles , Nanopartículas , Imagen Óptica/métodos , Animales , Línea Celular , Línea Celular Tumoral , Química Farmacéutica/métodos , Femenino , Semivida , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula
8.
J Immunol ; 188(5): 2316-27, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22279102

RESUMEN

Glutathione reductase (Gsr) catalyzes the reduction of glutathione disulfide to glutathione, which plays an important role in the bactericidal function of phagocytes. Because Gsr has been implicated in the oxidative burst in human neutrophils and is abundantly expressed in the lymphoid system, we hypothesized that Gsr-deficient mice would exhibit marked defects during the immune response against bacterial challenge. We report in this study that Gsr-null mice exhibited enhanced susceptibility to Escherichia coli challenge, indicated by dramatically increased bacterial burden, cytokine storm, striking histological abnormalities, and substantially elevated mortality. Additionally, Gsr-null mice exhibited elevated sensitivity to Staphylococcus aureus. Examination of the bactericidal functions of the neutrophils from Gsr-deficient mice in vitro revealed impaired phagocytosis and defective bacterial killing activities. Although Gsr catalyzes the regeneration of glutathione, a major cellular antioxidant, Gsr-deficient neutrophils paradoxically produced far less reactive oxygen species upon activation both ex vivo and in vivo. Unlike wild-type neutrophils that exhibited a sustained oxidative burst upon stimulation with phorbol ester and fMLP, Gsr-deficient neutrophils displayed a very transient oxidative burst that abruptly ceased shortly after stimulation. Likewise, Gsr-deficient neutrophils also exhibited an attenuated oxidative burst upon encountering E. coli. Biochemical analysis revealed that the hexose monophosphate shunt was compromised in Gsr-deficient neutrophils. Moreover, Gsr-deficient neutrophils displayed a marked impairment in the formation of neutrophil extracellular traps, a bactericidal mechanism that operates after neutrophil death. Thus, Gsr-mediated redox regulation is crucial for bacterial clearance during host defense against massive bacterial challenge.


Asunto(s)
Infecciones por Escherichia coli/prevención & control , Espacio Extracelular/inmunología , Glutatión Reductasa/fisiología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estrés Oxidativo/inmunología , Fagocitosis/inmunología , Infecciones Estafilocócicas/prevención & control , Animales , Escherichia coli/inmunología , Infecciones por Escherichia coli/enzimología , Infecciones por Escherichia coli/inmunología , Espacio Extracelular/genética , Espacio Extracelular/metabolismo , Glutatión Reductasa/deficiencia , Glutatión Reductasa/genética , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Neutrófilos/microbiología , Estrés Oxidativo/genética , Fagocitosis/genética , Infecciones Estafilocócicas/enzimología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
9.
Nanomedicine ; 10(5): 1053-63, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24355163

RESUMEN

The drugs/strategies to selectively inhibit tumor blood supply have generated interest in recent years for enhancement of cancer therapeutics. The objective of this study was to formulate tumor homing PEGylated CREKA peptide conjugated theranostic nanoparticles of DIM-C-pPhC6H5 (DIM-P) and investigate in vivo antitumor activity as well as evaluate the targeted efficiency to lung tumors using imaging techniques. DIM-P loaded Nanoparticles (NCs-D) were prepared using lipids, and DOGS-NTA-Ni and the surface of NCs-D were modified with PEGylated CREKA peptide (PCNCs-D). PCNCs-D showed 3 fold higher binding to clotted plasma proteins in tumor vasculature compared to NCs-D. PCNCs-D showed 26%±4% and 22%±5% increase in tumor reduction compared to NCs-D in metastatic and orthotopic models respectively. In-vivo imaging studies showed ~40 folds higher migration of PCNCs-Di in tumor vasculature than NCs-Di. Our studies demonstrate the role of PCNCs-D as theranostic tumor homing drug delivery and imaging systems for lung cancer diagnosis and treatment. FROM THE CLINICAL EDITOR: This study demonstrates a very efficient delivery system to address lung cancer growth through blood supply inhibition.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Femenino , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Desnudos , Oligopéptidos/química , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Antimicrob Agents Chemother ; 57(10): 5080-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23917317

RESUMEN

Treatment of prosthetic joint infections often involves a two-stage exchange, with implant removal and antibiotic spacer placement followed by systemic antibiotic therapy and delayed reimplantation. However, if antibiotic therapy can be improved, one-stage exchange or implant retention may be more feasible, thereby decreasing morbidity and preserving function. In this study, a mouse model of prosthetic joint infection was used in which Staphylococcus aureus was inoculated into a knee joint containing a surgically placed metallic implant extending from the femur. This model was used to evaluate whether combination therapy of vancomycin plus rifampin has increased efficacy compared with vancomycin alone against these infections. On postoperative day 7, vancomycin with or without rifampin was administered for 6 weeks with implant retention. In vivo bioluminescence imaging, ex vivo CFU enumeration, X-ray imaging, and histologic analysis were carried out. We found that there was a marked therapeutic benefit when vancomycin was combined with rifampin compared with vancomycin alone. Taken together, our results suggest that the mouse model used could serve as a valuable in vivo preclinical model system to evaluate and compare efficacies of antibiotics and combinatory therapy for prosthetic joint infections before more extensive studies are carried out in human subjects.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Rifampin/uso terapéutico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/patogenicidad , Vancomicina/uso terapéutico , Animales , Antibacterianos/administración & dosificación , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos C57BL , Rifampin/administración & dosificación , Staphylococcus aureus/efectos de los fármacos , Vancomicina/administración & dosificación
11.
Antimicrob Agents Chemother ; 56(5): 2590-7, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22371896

RESUMEN

Vancomycin is widely used for intravenous prophylaxis against surgical implant infections. However, it is unclear whether alternative antibiotics used to treat methicillin-resistant Staphylococcus aureus (MRSA) infections are effective as prophylactic agents. The aim of this study was to compare the efficacies of vancomycin, daptomycin, and tigecycline as prophylactic therapy against a methicillin-sensitive S. aureus (MSSA) or MRSA surgical implant infection in mice. MSSA or MRSA was inoculated into the knee joints of mice in the presence of a surgically placed medical-grade metallic implant. The efficacies of low- versus high-dose vancomycin (10 versus 110 mg/kg), daptomycin (1 versus 10 mg/kg), and tigecycline (1 versus 10 mg/kg) intravenous prophylaxis were compared using in vivo bioluminescence imaging, ex vivo bacterial counts, and biofilm formation. High-dose vancomycin, daptomycin, and tigecycline resulted in similar reductions in bacterial burden and biofilm formation. In contrast, low-dose daptomycin and tigecycline were more effective than low-dose vancomycin against the implant infection. In this mouse model of surgical implant MSSA or MRSA infection, daptomycin and tigecycline prophylaxis were effective over a broader dosage range than vancomycin. Future studies in humans will be required to determine whether these broader effective dose ranges for daptomycin and tigecycline in mice translate to improved efficacy in preventing surgical implant infections in clinical practice.


Asunto(s)
Daptomicina/administración & dosificación , Prótesis de la Rodilla/microbiología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Minociclina/análogos & derivados , Infecciones Relacionadas con Prótesis/prevención & control , Infecciones Estafilocócicas/prevención & control , Animales , Antibacterianos/administración & dosificación , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Recuento de Colonia Microbiana , Esquema de Medicación , Humanos , Inyecciones Intravenosas , Masculino , Staphylococcus aureus Resistente a Meticilina/fisiología , Ratones , Ratones Endogámicos C57BL , Minociclina/administración & dosificación , Modelos Animales , Imagen Molecular , Infecciones Relacionadas con Prótesis/microbiología , Infecciones Estafilocócicas/microbiología , Tigeciclina , Vancomicina/administración & dosificación
12.
Microbiology (Reading) ; 158(Pt 11): 2826-2834, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22902730

RESUMEN

Citrobacter rodentium, which colonizes the gut mucosa via formation of attaching and effacing (A/E) lesions, causes transmissible colonic hyperplasia. The aim of this study was to evaluate whether prophylactic treatment with Bifidobacterium breve UCC2003 can improve the outcome of C. rodentium infection. Six-week-old albino C57BL/6 mice were pre-treated for 3 days with B. breve, challenged with bioluminescent C. rodentium and administered B. breve or PBS-C for 8 days post-infection; control mice were either administered B. breve and mock-infected with PBS, or mock-treated with PBS-C and mock-infected with PBS. C. rodentium colonization was monitored by bacterial enumeration from faeces and by a combination of both 2D bioluminescence imaging (BLI) and composite 3D diffuse light imaging tomography with µCT imaging (DLIT-µCT). At day 8 post-infection, colons were removed and assessed for crypt hyperplasia, histology by light microscopy, bacterial colonization by immunofluorescence, and A/E lesion formation by electron microscopy. Prophylactic administration of B. breve did not prevent C. rodentium colonization or A/E lesion formation. However, this treatment did alter C. rodentium distribution within the large intestine and significantly reduced colonic crypt hyperplasia at the peak of bacterial infection. These results show that B. breve could not competitively exclude C. rodentium, but reduced pathogen-induced colonic inflammation.


Asunto(s)
Bifidobacterium/fisiología , Citrobacter rodentium/fisiología , Enfermedades del Colon/prevención & control , Infecciones por Enterobacteriaceae/prevención & control , Probióticos/administración & dosificación , Animales , Citrobacter rodentium/inmunología , Colon/inmunología , Colon/microbiología , Colon/patología , Enfermedades del Colon/inmunología , Enfermedades del Colon/microbiología , Enfermedades del Colon/patología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos
13.
Blood ; 115(21): 4284-92, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20299509

RESUMEN

Although red blood cell (RBC) transfusions can be lifesaving, they are not without risk. In critically ill patients, RBC transfusions are associated with increased morbidity and mortality, which may increase with prolonged RBC storage before transfusion. The mechanisms responsible remain unknown. We hypothesized that acute clearance of a subset of damaged, stored RBCs delivers large amounts of iron to the monocyte/macrophage system, inducing inflammation. To test this in a well-controlled setting, we used a murine RBC storage and transfusion model to show that the transfusion of stored RBCs, or washed stored RBCs, increases plasma nontransferrin bound iron (NTBI), produces acute tissue iron deposition, and initiates inflammation. In contrast, the transfusion of fresh RBCs, or the infusion of stored RBC-derived supernatant, ghosts, or stroma-free lysate, does not produce these effects. Furthermore, the insult induced by transfusion of stored RBC synergizes with subclinical endotoxinemia producing clinically overt signs and symptoms. The increased plasma NTBI also enhances bacterial growth in vitro. Taken together, these results suggest that, in a mouse model, the cellular component of leukoreduced, stored RBC units contributes to the harmful effects of RBC transfusion that occur after prolonged storage. Nonetheless, these findings must be confirmed by prospective human studies.


Asunto(s)
Conservación de la Sangre/efectos adversos , Transfusión de Eritrocitos/efectos adversos , Inflamación/etiología , Hierro/sangre , Reacción de Fase Aguda/sangre , Reacción de Fase Aguda/etiología , Animales , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Endotoxemia/sangre , Endotoxemia/etiología , Endotoxinas/sangre , Envejecimiento Eritrocítico , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/etiología , Hemoglobinas/metabolismo , Humanos , Inflamación/sangre , Inflamación/prevención & control , Mediadores de Inflamación/sangre , Hierro/metabolismo , Quelantes del Hierro/farmacología , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Bazo/metabolismo , Factores de Tiempo
14.
Proc Natl Acad Sci U S A ; 106(35): 15002-6, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19706444

RESUMEN

Prions are infectious proteins that cause fatal neurodegenerative diseases. Because astrocytic gliosis marked by the deposition of fibrils composed of GFAP is a prominent feature of prion disease, we asked whether GFAP might be used as a surrogate marker for prions. To interrogate this posit, we inoculated prions into transgenic (Tg) mice expressing luciferase (luc) under the GFAP gene (Gfap) promoter, denoted Tg(Gfap-luc) mice. Weekly noninvasive, bioluminescence imaging (BLI) detected an increase in light emitted from the brains of Tg(Gfap-luc) mice at approximately 55 d after inoculation and approximately 62 d before neurologic deficits appeared. To determine whether BLI could be used as a proxy bioassay for prion infectivity, we performed endpoint titrations of prions in Tg(Gfap-luc) mice. BLI bioassays were as or more sensitive than those determined by the onset of neurological dysfunction, and were completed in approximately half the time. Our studies argue that BLI is likely to be a suitable surrogate for measuring prion infectivity, and might be useful in the study of Tg mouse models for other neurodegenerative illnesses.


Asunto(s)
Mediciones Luminiscentes/métodos , Enfermedades por Prión/metabolismo , Priones/análisis , Animales , Modelos Animales de Enfermedad , Genes Reporteros , Inmunohistoquímica , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Transgénicos , Enfermedades por Prión/patología , Priones/metabolismo
15.
J Biomed Mater Res B Appl Biomater ; 110(8): 1932-1941, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35352867

RESUMEN

Preclinical in vitro and in vivo methods to study bacterial interactions with dermal fillers and infection pathogenesis are lacking. In this work, first in vitro methods to assess protein biofouling and effective pore size of commercial dermal fillers, including degradable hyaluronic acid (HA)-based fillers and other semi-degradable or permanent fillers (non-HA), were developed. The results were then related to Staphylococcus aureus (S. aureus) adhesion rates in vitro. HA fillers had less protein sorption than non-HA fillers and overall had smaller effective pore sizes. The properties correlated with levels of bacterial adhesion, where the control glass surface had the most rapid increase in bacterial cell adhesion, with a slope of 0.29 cm-2  min-1 , three unique non-HA fillers had intermediate adhesion with slopes of 0.11 and 0.06 cm-2  min-1 , and three unique HA fillers had the least adhesion with slopes of 0.02, 0.02, and 0.01 cm-2  min-1 . S. aureus had greater motility on the HA fillers than on non-HA fillers. Next, a mouse model for dermal filler biofilm and infection was developed. Mice were inoculated with a controlled amount of bioluminescent bacteria (Xen36 S. aureus) and polyacrylamide hydrogels of different stiffness were injected. In vivo bioluminescence was monitored longitudinally for 35 days to ensure that lasting colonization was established. The inoculum was optimized to achieve adequate bioluminescent signal, and bacterial bioburden over time and inter-animal variability in bioburden were determined. These in vitro and in vivo approaches can be used for future studies of antimicrobial interventions for dermal fillers.


Asunto(s)
Rellenos Dérmicos , Animales , Ácido Hialurónico/farmacología , Hidrogeles/farmacología , Ratones , Staphylococcus aureus
16.
Viruses ; 14(3)2022 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-35336942

RESUMEN

Animal models recapitulating COVID-19 are critical to enhance our understanding of SARS-CoV-2 pathogenesis. Intranasally inoculated transgenic mice expressing human angiotensin-converting enzyme 2 under the cytokeratin 18 promoter (K18-hACE2) represent a lethal model of SARS-CoV-2 infection. We evaluated the clinical and virological dynamics of SARS-CoV-2 using two intranasal doses (104 and 106 PFUs), with a detailed spatiotemporal pathologic analysis of the 106 dose cohort. Despite generally mild-to-moderate pneumonia, clinical decline resulting in euthanasia or death was commonly associated with hypothermia and viral neurodissemination independent of inoculation dose. Neuroinvasion was first observed at 4 days post-infection, initially restricted to the olfactory bulb suggesting axonal transport via the olfactory neuroepithelium as the earliest portal of entry. Absence of viremia suggests neuroinvasion occurs independently of transport across the blood-brain barrier. SARS-CoV-2 tropism was neither restricted to ACE2-expressing cells (e.g., AT1 pneumocytes), nor inclusive of some ACE2-positive cell lineages (e.g., bronchiolar epithelium and brain vasculature). Absence of detectable ACE2 protein expression in neurons but overexpression in neuroepithelium suggest this as the most likely portal of neuroinvasion, with subsequent ACE2 independent lethal neurodissemination. A paucity of epidemiological data and contradicting evidence for neuroinvasion and neurodissemination in humans call into question the translational relevance of this model.


Asunto(s)
COVID-19 , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2 , Animales , Humanos , Queratina-18 , Melfalán , Ratones , Ratones Transgénicos , SARS-CoV-2/genética , Tropismo Viral , gammaglobulinas
17.
Cell Rep ; 39(3): 110714, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35421379

RESUMEN

The human immunological mechanisms defining the clinical outcome of SARS-CoV-2 infection remain elusive. This knowledge gap is mostly driven by the lack of appropriate experimental platforms recapitulating human immune responses in a controlled human lung environment. Here, we report a mouse model (i.e., HNFL mice) co-engrafted with human fetal lung xenografts (fLX) and a myeloid-enhanced human immune system to identify cellular and molecular correlates of lung protection during SARS-CoV-2 infection. Unlike mice solely engrafted with human fLX, HNFL mice are protected against infection, severe inflammation, and histopathological phenotypes. Lung tissue protection from infection and severe histopathology associates with macrophage infiltration and differentiation and the upregulation of a macrophage-enriched signature composed of 11 specific genes mainly associated with the type I interferon signaling pathway. Our work highlights the HNFL model as a transformative platform to investigate, in controlled experimental settings, human myeloid immune mechanisms governing lung tissue protection during SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Animales , COVID-19/genética , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Pulmón/patología , Macrófagos , Ratones , SARS-CoV-2
18.
Sci Transl Med ; 14(662): eabj2381, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36103517

RESUMEN

Drug-resistant Gram-positive bacterial infections are still a substantial burden on the public health system, with two bacteria (Staphylococcus aureus and Streptococcus pneumoniae) accounting for over 1.5 million drug-resistant infections in the United States alone in 2017. In 2019, 250,000 deaths were attributed to these pathogens globally. We have developed a preclinical glycopeptide antibiotic, MCC5145, that has excellent potency (MIC90 ≤ 0.06 µg/ml) against hundreds of isolates of methicillin-resistant S. aureus (MRSA) and other Gram-positive bacteria, with a greater than 1000-fold margin over mammalian cell cytotoxicity values. The antibiotic has therapeutic in vivo efficacy when dosed subcutaneously in multiple murine models of established bacterial infections, including thigh infection with MRSA and blood septicemia with S. pneumoniae, as well as when dosed orally in an antibiotic-induced Clostridioides difficile infection model. MCC5145 exhibited reduced nephrotoxicity at microbiologically active doses in mice compared to vancomycin. MCC5145 also showed improved activity against biofilms compared to vancomycin, both in vitro and in vivo, and a low propensity to select for drug resistance. Characterization of drug action using a transposon library bioinformatic platform showed a mechanistic distinction from other glycopeptide antibiotics.


Asunto(s)
Antiinfecciosos , Infecciones por Bacterias Grampositivas , Staphylococcus aureus Resistente a Meticilina , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , Biopelículas , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Lipoglucopéptidos/uso terapéutico , Mamíferos , Ratones , Pruebas de Sensibilidad Microbiana , Streptococcus pneumoniae , Vancomicina/farmacología , Vancomicina/uso terapéutico
19.
Nat Commun ; 12(1): 2680, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976191

RESUMEN

Bioluminescent imaging (BLI) is one of the most powerful and widely used preclinical imaging modalities. However, the current technology relies on the use of transgenic luciferase-expressing cells and animals and therefore can only be applied to a limited number of existing animal models of human disease. Here, we report the development of a "portable bioluminescent" (PBL) technology that overcomes most of the major limitations of traditional BLI. We demonstrate that the PBL method is capable of noninvasive measuring the activity of both extracellular (e.g., dipeptidyl peptidase 4) and intracellular (e.g., cytochrome P450) enzymes in vivo in non-luciferase-expressing mice. Moreover, we successfully utilize PBL technology in dogs and human cadaver, paving the way for the translation of functional BLI to the noninvasive quantification of biological processes in large animals. The PBL methodology can be easily adapted for the noninvasive monitoring of a plethora of diseases across multiple species.


Asunto(s)
Fenómenos Biológicos , Diagnóstico por Imagen/métodos , Mediciones Luminiscentes/métodos , Modelos Animales , Animales , Animales Modificados Genéticamente , Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Dipeptidil Peptidasa 4/química , Dipeptidil Peptidasa 4/metabolismo , Perros , Luciferina de Luciérnaga/química , Luciferina de Luciérnaga/metabolismo , Humanos , Luciferasas/química , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/instrumentación , Estructura Molecular , Reproducibilidad de los Resultados
20.
bioRxiv ; 2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-33469581

RESUMEN

Animal models recapitulating distinctive features of severe COVID-19 are critical to enhance our understanding of SARS-CoV-2 pathogenesis. Transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) under the cytokeratin 18 promoter (K18-hACE2) represent a lethal model of SARS-CoV-2 infection. The precise mechanisms of lethality in this mouse model remain unclear. Here, we evaluated the spatiotemporal dynamics of SARS-CoV-2 infection for up to 14 days post-infection. Despite infection and moderate pneumonia, rapid clinical decline or death of mice was invariably associated with viral neuroinvasion and direct neuronal injury (including brain and spinal neurons). Neuroinvasion was observed as early as 4 dpi, with virus initially restricted to the olfactory bulb supporting axonal transport via the olfactory neuroepithelium as the earliest portal of entry. No evidence of viremia was detected suggesting neuroinvasion occurs independently of entry across the blood brain barrier. SARS-CoV-2 tropism was not restricted to ACE2-expressing cells (e.g., AT1 pneumocytes), and some ACE2-positive lineages were not associated with the presence of viral antigen (e.g., bronchiolar epithelium and brain capillaries). Detectable ACE2 expression was not observed in neurons, supporting overexpression of ACE2 in the nasal passages and neuroepithelium as more likely determinants of neuroinvasion in the K18-hACE2 model. Although our work incites caution in the utility of the K18-hACE2 model to study global aspects of SARS-CoV-2 pathogenesis, it underscores this model as a unique platform for exploring the mechanisms of SARS-CoV-2 neuropathogenesis that may have clinical relevance acknowledging the growing body of evidence that suggests COVID-19 may result in long-standing neurologic consequences.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA