Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biol Reprod ; 109(5): 618-634, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37665249

RESUMO

OBJECTIVES: The bacterium Listeria monocytogenes (Lm) is associated with adverse pregnancy outcomes. Infection occurs through consumption of contaminated food that is disseminated to the maternal-fetal interface. The influence on the gastrointestinal microbiome during Lm infection remains unexplored in pregnancy. The objective of this study was to determine the impact of listeriosis on the gut microbiota of pregnant macaques. METHODS: A non-human primate model of listeriosis in pregnancy has been previously described. Both pregnant and non-pregnant cynomolgus macaques were inoculated with Lm and bacteremia and fecal shedding were monitored for 14 days. Non-pregnant animal tissues were collected at necropsy to determine bacterial burden, and fecal samples from both pregnant and non-pregnant animals were evaluated by 16S rRNA next-generation sequencing. RESULTS: Unlike pregnant macaques, non-pregnant macaques did not exhibit bacteremia, fecal shedding, or tissue colonization by Lm. Dispersion of Lm during pregnancy was associated with a significant decrease in alpha diversity of the host gut microbiome, compared to non-pregnant counterparts. The combined effects of pregnancy and listeriosis were associated with a significant loss in microbial richness, although there were increases in some genera and decreases in others. CONCLUSIONS: Although pregnancy alone is not associated with gut microbiome disruption, we observed dysbiosis with listeriosis during pregnancy. The macaque model may provide an understanding of the roles that pregnancy and the gut microbiota play in the ability of Lm to establish intestinal infection and disseminate throughout the host, thereby contributing to adverse pregnancy outcomes and risk to the developing fetus.


Assuntos
Bacteriemia , Microbioma Gastrointestinal , Listeria monocytogenes , Listeriose , Gravidez , Animais , Feminino , RNA Ribossômico 16S/genética , Listeriose/veterinária , Listeriose/complicações , Listeriose/microbiologia , Macaca fascicularis , Bacteriemia/complicações
2.
Angew Chem Int Ed Engl ; : e202201798, 2022 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-35334139

RESUMO

A synthetic peptide was found to block cell-to-cell signalling, or quorum sensing, in bacteria and be highly bioavailable in mouse tissue. The controlled release of this agent from degradable polymeric microparticles strongly inhibited skin infection in a wound model at levels that far surpassed the potency of the peptide when delivered conventionally.

3.
Microb Pathog ; 122: 30-38, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29842898

RESUMO

Bacterial biofilms impair healing in 60% of chronic skin wounds. Various animal models (mice, rats, rabbits, and pigs) have been developed to replicate biofilm infected wounds in vivo. We developed a sustained wound infection model by applying preformed Pseudomonas aeruginosa biofilms on a wound dressing to full-thickness murine skin wounds. We bathed a commercially available wound dressing in P. aeruginosa for 48 h, allowing a biofilm to establish on the dressing prior to application to the wound. Dressings were removed from the wounds after 3 days at which time the wound beds contained ∼108 bacterial cells per gram tissue. Significant numbers of P. aeruginosa persisted within the skin wounds for up to 21 days. Un-inoculated wounds reached closure between 9 and 12 days. In contrast, biofilm-inoculated wounds achieved closure between 18 and 21 days. Histologic analysis confirmed decreased re-epithelialization and collagen deposition, coupled with increased inflammation, in the biofilm-inoculated wounds compared to un-inoculated controls. This novel model of delayed healing and persistent infection of full-thickness murine skin wounds may provide a robust in vivo system in which to test novel treatments to prevent wound infection by bacterial biofilms.


Assuntos
Biofilmes/crescimento & desenvolvimento , Modelos Animais de Doenças , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Cicatrização , Infecção dos Ferimentos/patologia , Animais , Bandagens , Histocitoquímica , Camundongos
4.
Curr Top Microbiol Immunol ; 396: 71-87, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26728064

RESUMO

Histophilus somni resides as part of the normal microflora in the upper respiratory tract of healthy cattle. From this site, the organism can make its way into the lower respiratory tract, where it is one of the important bacterial agents of the respiratory disease complex. If H. somni cells disseminate to the bloodstream, they frequently result in thrombus formation. A series of in vitro investigations have examined potential mechanisms that might contribute to such thrombus formation. Earlier work showed that H. somni can stimulate some bovine endothelial cells to undergo apoptosis. More recent studies indicate that H. somni stimulates endothelial cell tissue factor activity and disrupts intercellular junctions. The net effect is to enhance procoagulant activity on the endothelium surface and to make the endothelial monolayer more permeable to molecules, leukocytes, and perhaps H. somni cells. H. somni also activates bovine platelets, which also can enhance tissue factor activity on the endothelium surface. When exposed to H. somni, bovine neutrophils and mononuclear phagocytes form extracellular traps in vitro. Ongoing research is investigating how the interplay among endothelial cells, platelets, and leukocytes might contribute to the thrombus formation seen in infected cattle.


Assuntos
Interações Hospedeiro-Patógeno , Pasteurellaceae/patogenicidade , Animais , Permeabilidade Capilar , Bovinos , Armadilhas Extracelulares/fisiologia , Imunidade Inata , Trombose/etiologia
5.
Angew Chem Int Ed Engl ; 55(19): 5698-702, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27059788

RESUMO

Antimicrobial surfaces with covalently attached biocidal functionalities only kill microbes that come into direct contact with the surfaces (contact-killing surfaces). Herein, the activity of contact-killing surfaces is shown to be enhanced by using gradients in the concentration of soluble chemoattractants (CAs) to attract bacteria to the surfaces. Two natural and nonbiocidal CAs (aspartate and glucose) were used to attract bacteria to model surfaces decorated with quaternary ammonium groups (known to kill bacteria that come into contact with them). These results demonstrate the killing of Escherichia coli and Salmonella typhimurium, two common pathogens, at levels 10- to 20-times greater than that of the native surfaces alone. This approach is general and provides new strategies for the design of active or dynamic contact-killing surfaces with enhanced antimicrobial activities.


Assuntos
Anti-Infecciosos/farmacologia , Fatores Quimiotáticos/química , Escherichia coli/efeitos dos fármacos , Salmonella typhimurium/efeitos dos fármacos , Anti-Infecciosos/química , Ácido Aspártico/química , Fatores Quimiotáticos/farmacologia , Compostos de Organossilício/química , Compostos de Amônio Quaternário/química , Silício/química , Propriedades de Superfície
6.
Wound Repair Regen ; 23(6): 842-54, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26342168

RESUMO

Chronic nonhealing skin wounds often contain bacterial biofilms that prevent normal wound healing and closure and present challenges to the use of conventional wound dressings. We investigated inhibition of Pseudomonas aeruginosa biofilm formation, a common pathogen of chronic skin wounds, on a commercially available biological wound dressing. Building on prior reports, we examined whether the amino acid tryptophan would inhibit P. aeruginosa biofilm formation on the three-dimensional surface of the biological dressing. Bacterial biomass and biofilm polysaccharides were quantified using crystal violet staining or an enzyme linked lectin, respectively. Bacterial cells and biofilm matrix adherent to the wound dressing were visualized through scanning electron microscopy. D-/L-tryptophan inhibited P. aeruginosa biofilm formation on the wound dressing in a dose dependent manner and was not directly cytotoxic to immortalized human keratinocytes although there was some reduction in cellular metabolism or enzymatic activity. More importantly, D-/L-tryptophan did not impair wound healing in a splinted skin wound murine model. Furthermore, wound closure was improved when D-/L-tryptophan treated wound dressing with P. aeruginosa biofilms were compared with untreated dressings. These findings indicate that tryptophan may prove useful for integration into wound dressings to inhibit biofilm formation and promote wound healing.


Assuntos
Anti-Infecciosos Locais/farmacologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/efeitos dos fármacos , Lesões dos Tecidos Moles/patologia , Triptofano/farmacologia , Cicatrização , Infecção dos Ferimentos/patologia , Animais , Bandagens , Biofilmes/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Varredura , Infecções por Pseudomonas/microbiologia , Lesões dos Tecidos Moles/microbiologia , Infecção dos Ferimentos/microbiologia
7.
Anal Chem ; 86(8): 3764-72, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24559115

RESUMO

Accurate and rapid assessment of the healing status of a wound in a simple and noninvasive manner would enable clinicians to diagnose wounds in real time and promptly adjust treatments to hasten the resolution of nonhealing wounds. Histologic and biochemical characterization of biopsied wound tissue, which is currently the only reliable method for wound assessment, is invasive, complex to interpret, and slow. Here we demonstrate the use of Raman microspectroscopy coupled with multivariate spectral analysis as a simple, noninvasive method to biochemically characterize healing wounds in mice and to accurately identify different phases of healing of wounds at different time-points. Raman spectra were collected from "splinted" full thickness dermal wounds in mice at 4 time-points (0, 1, 5, and 7 days) corresponding to different phases of wound healing, as verified by histopathology. Spectra were deconvolved using multivariate factor analysis (MFA) into 3 "factor score spectra" (that act as spectral signatures for different stages of healing) that were successfully correlated with spectra of prominent pure wound bed constituents (i.e., collagen, lipids, fibrin, fibronectin, etc.) using non-negative least squares (NNLS) fitting. We show that the factor loadings (weights) of spectra that belonged to wounds at different time-points provide a quantitative measure of wound healing progress in terms of key parameters such as inflammation and granulation. Wounds at similar stages of healing were characterized by clusters of loading values and slowly healing wounds among them were successfully identified as "outliers". Overall, our results demonstrate that Raman spectroscopy can be used as a noninvasive technique to provide insight into the status of normally healing and slow-to-heal wounds and that it may find use as a complementary tool for real-time, in situ biochemical characterization in wound healing studies and clinical diagnosis.


Assuntos
Análise Espectral Raman/métodos , Cicatrização/fisiologia , Ferimentos e Lesões/classificação , Ferimentos e Lesões/patologia , Animais , Biópsia , Análise Fatorial , Tecido de Granulação/química , Inflamação/metabolismo , Análise dos Mínimos Quadrados , Camundongos , Camundongos Endogâmicos BALB C , Análise Multivariada , Pele/química
8.
Antimicrob Agents Chemother ; 57(4): 1921-5, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23318791

RESUMO

Biofilm formation by Pseudomonas aeruginosa has been implicated in the pathology of chronic wounds. Both the d and l isoforms of tryptophan inhibited P. aeruginosa biofilm formation on tissue culture plates, with an equimolar ratio of d and l isoforms producing the greatest inhibitory effect. Addition of d-/l-tryptophan to existing biofilms inhibited further biofilm growth and caused partial biofilm disassembly. Tryptophan significantly increased swimming motility, which may be responsible in part for diminished biofilm formation by P. aeruginosa.


Assuntos
Biofilmes/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , Triptofano/farmacologia
9.
Microb Pathog ; 56: 47-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23178254

RESUMO

In this study, we first assessed the effect of intragastric infection of pregnant mice with Listeria monocytogenes on relative expression of select genes associated with T cell subsets. Relative gene expression was moderately increased in placental tissues for IFNγ, IL-4, IL-17a, IL-22, CD3, and FoxP3. To assess the roles of IL-17a and IL-22 in resistance to listeriosis during pregnancy, we compared the severity of maternal and fetal infection in IL-17a((-/-)), IL-22((-/-)), and IL-17a((-/-))/IL-22((-/-)) mice with that of wild type C57BL/6 mice. Intragastric infection with modest numbers of bacterial cells (10(5) CFU) caused reproducible maternal and fetal infection in all four mouse strains. We recovered greater numbers of CFU from the bloodstream of pregnant IL-22((-/-)) mice than pregnant wild type mice. Otherwise we found no significant difference in bacterial load in maternal or fetal tissues (spleen, liver, fetoplacental units) from pregnant IL-17a((-/-)), IL-22((-/-)), or IL-17a((-/-))/IL-22((-/-)) or wild type mice. Nor did we observe histopathologic differences in severity of inflammation in maternal or fetal tissues from the various groups of mice. Although IL-17a and IL-22 are up-regulated in placental tissue, our study suggests that antibacterial resistance and the host inflammatory response are not dependent on IL-17a or IL-22 during infection of mice with L. monocytogenes at 10-14 days of gestation.


Assuntos
Carga Bacteriana , Feto/microbiologia , Interleucina-17/imunologia , Interleucinas/imunologia , Listeria monocytogenes/patogenicidade , Listeriose/patologia , Complicações Infecciosas na Gravidez/patologia , Estruturas Animais/microbiologia , Estruturas Animais/patologia , Animais , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Feminino , Feto/patologia , Perfilação da Expressão Gênica , Listeria monocytogenes/imunologia , Listeriose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placenta/microbiologia , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Interleucina 22
10.
Microb Pathog ; 54: 67-75, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23022668

RESUMO

Histophilus somni (formerly Haemophilus somnus) is a Gram-negative pleomorphic coccobacillus that causes respiratory, reproductive, cardiac and neuronal diseases in cattle. H. somni is a member of the bovine respiratory disease complex that causes severe bronchopneumonia in cattle. Previously, it has been reported that bovine neutrophils and macrophages have limited ability to phagocytose and kill H. somni. Recently, it was discovered that bovine neutrophils and macrophages produce extracellular traps in response to Mannheimia haemolytica, another member of the bovine respiratory disease complex. In this study, we demonstrate that H. somni also causes extracellular trap production by bovine neutrophils in a dose- and time-dependent manner, which did not coincide with the release of lactate dehydrogenase, a marker for necrosis. Neutrophil extracellular traps were produced in response to outer membrane vesicles, but not lipooligosacchride alone. Using scanning electron microscopy and confocal microscopy, we observed H. somni cells trapped within a web-like structure. Further analyses demonstrated that bovine neutrophils trapped and killed H. somni in a DNA-dependent manner. Treatment of DNA extracellular traps with DNase I freed H. somni cells and diminished bacterial death. Treatment of bovine monocyte-derived macrophages with H. somni cells also caused macrophage extracellular trap formation. These findings suggest that extracellular traps may play a role in the host response to H. somni infection in cattle.


Assuntos
Macrófagos/imunologia , Macrófagos/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Pasteurellaceae/imunologia , Animais , Bovinos , DNA/metabolismo , Viabilidade Microbiana , Microscopia Confocal , Microscopia Eletrônica de Varredura
11.
Immunopharmacol Immunotoxicol ; 35(1): 15-27, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22994841

RESUMO

α-Hemolysin (HLY) is an important virulence factor for uropathogenic Escherichia coli. HLY is a member of the RTX family of exotoxins secreted by a number of Gram-negative bacteria. Recently, it was reported that a related RTX toxin, the Mannheimia haemolytica leukotoxin, exhibits increased cytotoxicity following brief heat treatment. In this article, we show that brief heat treatment (1 min at 100°C) increases cytotoxicity of HLY for human bladder cells, kidney epithelial cells (A498) and neutrophils. Heat treatment also increased hemolysis of human red blood cells (RBCs). Furthermore, heat treatment of previously inactived HLY restored its cytotoxicity. Heat-activated and native HLY both required glycophorin A to lyse RBCs. Native and heat-activated HLY appeared to bind equally well to the surface of A498 cells; although, Western blot analyses demonstrated binding to different proteins on the surface. Confocal microscopy revealed that heat-activated HLY bound more extensively to internal structures of permeabilized A498 cells than did native HLY. Several lines of spectroscopic evidence demonstrate irreversible changes in the structure of heat activated compared to native HLY. We show changes in secondary structure, increased exposure of tryptophan residues to the aqueous environment, an increase in molecular dimension and an increase in hydrophobic surface area. These properties are among the most common characteristics described for the molten globule state, first identified as an intermediate in protein folding. We hypothesize that brief heat treatment of HLY causes a conformational change leading to significant differences in protein-protein interactions that result in increased cytotoxicity for target cells.


Assuntos
Escherichia coli/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Eritrócitos/metabolismo , Feminino , Glicoforinas/farmacologia , Hemólise/fisiologia , Temperatura Alta , Humanos , Simulação de Dinâmica Molecular , Neutrófilos/metabolismo , Dobramento de Proteína , Triptofano/metabolismo
12.
JDS Commun ; 4(2): 86-90, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36974218

RESUMO

Cattle persistently infected with bovine-adapted serotypes of Salmonella enterica are an important animal health and food safety issue. One possible mechanism by which infection is sustained in a dairy herd is by survival of Salmonella in sand used as bedding material. In this study we assessed the survival of 107 to 108 cfu bovine-associated serotypes of Salmonella enterica (sv. Cerro, Dublin, and Heidelberg) in sterile sand, recycled bedding sand, and gray water collected from a Wisconsin dairy farm. All 3 serotypes persisted at relatively high numbers (>106 cfu/g) for at least 28 d in sterile sand, with Salmonella sv. Dublin decreasing less than 1 log10 over 70 d. To our surprise, when low numbers of Salmonella sv. Dublin (103 cfu) were inoculated into sterile sand, the organism multiplied within 3 d to approximately 106 cfu/g sand and persisted at that level for 28 d. When we inoculated Salmonella sv. Dublin into recycled bedding sand or sand taken directly from cow pens, we observed a significant decrease in colony-forming units by d 7. In contrast, we observed a significant increase in colony-forming units when Salmonella sv. Dublin was inoculated into gray water from the sand recycling system. These data demonstrate that Salmonella can persist for extended periods of time in bedding sand, although this is limited to some extent by the native microbiota in recycled bedding sand.

13.
Infect Immun ; 80(5): 1923-33, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22354029

RESUMO

Human and bovine neutrophils release neutrophil extracellular traps (NETs), which are protein-studded DNA matrices capable of extracellular trapping and killing of pathogens. Recently, we reported that bovine neutrophils release NETs in response to the important respiratory pathogen Mannheimia haemolytica and its leukotoxin (LKT). Here, we demonstrate macrophage extracellular trap (MET) formation by bovine monocyte-derived macrophages exposed to M. haemolytica or its LKT. Both native fully active LKT and noncytolytic pro-LKT (produced by an lktC mutant of M. haemolytica) stimulated MET formation. Confocal and scanning electron microscopy revealed a network of DNA fibrils with colocalized histones in extracellular traps released from bovine macrophages. Formation of METs required NADPH oxidase activity, as previously demonstrated for NET formation. METs formed in response to LKT trapped and killed a portion of the M. haemolytica cells. Bovine alveolar macrophages, but not peripheral blood monocytes, also formed METs in response to M. haemolytica cells. MET formation was not restricted to bovine macrophages. We also observed MET formation by the mouse macrophage cell line RAW 264.7 and by human THP-1 cell-derived macrophages, in response to Escherichia coli hemolysin. The latter is a member of the repeats-in-toxin (RTX) toxin family related to the M. haemolytica leukotoxin. This study demonstrates that macrophages, like neutrophils, can form extracellular traps in response to bacterial pathogens and their exotoxins.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Exotoxinas/metabolismo , Espaço Extracelular/metabolismo , Macrófagos/metabolismo , Mannheimia haemolytica/fisiologia , Animais , Bovinos , Linhagem Celular , Escherichia coli/metabolismo , Exotoxinas/toxicidade , Proteínas Hemolisinas/metabolismo , Humanos , Camundongos , NADPH Oxidases/metabolismo
14.
Ann Surg ; 256(2): 371-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22609841

RESUMO

OBJECTIVE: To investigate the antibacterial effect of augmenting a biological dressing with polymer films containing silver nanoparticles. BACKGROUND: Biological dressings, such as Biobrane, are commonly used for treating partial-thickness wounds and burn injuries. Biological dressings have several advantages over traditional wound dressings. However, as many as 19% of wounds treated with Biobrane become infected, and, once infected, the Biobrane must be removed and a traditional dressing approach should be employed. Silver is a commonly used antimicrobial in wound care products, but current technology uses cytotoxic concentrations of silver in these dressings. We have developed a novel and facile technology that allows immobilization of bioactive molecules on the surfaces of soft materials, demonstrated here by augmentation of Biobrane with nanoparticulate silver. Surfaces modified with nanometer-thick polyelectrolyte multilayers (PEMs) impregnated with silver nanoparticles have been shown previously to result in in vitro antibacterial activity against Staphylococcus epidermidis at loadings of silver that are noncytotoxic. METHODS: We demonstrated that silver-impregnated PEMs can be nondestructively immobilized onto the surface of Biobrane (Biobrane-Ag) and determined the in vitro antibacterial activity of Biobrane-Ag with Staphylococcus aureus. In this study, we used an in vivo wound infection model in mice induced by topical inoculation of S aureus onto full-thickness 6-mm diameter wounds. After 72 hours, bacterial quantification was performed. RESULTS: Wounds treated with Biobrane-Ag had significantly (P < 0.001) fewer colony-forming units than wounds treated with unmodified Biobrane (more than 4 log10 difference). CONCLUSIONS: The results of our study indicate that immobilizing silver-impregnated PEMs on the wound-contact surface of Biobrane significantly reduces bacterial bioburden in full-thickness murine skin wounds. Further research will investigate whether this construct can be considered for human use.


Assuntos
Curativos Biológicos , Materiais Revestidos Biocompatíveis/uso terapêutico , Curativos Oclusivos , Engenharia Tecidual/métodos , Animais , Materiais Revestidos Biocompatíveis/química , Modelos Animais de Doenças , Nanopartículas Metálicas , Camundongos , Polímeros/química , Prata/química , Cicatrização
15.
PLoS Pathog ; 6(6): e1000966, 2010 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-20585572

RESUMO

Type I interferons (IFNs) are known to mediate viral control, and also promote survival and expansion of virus-specific CD8+ T cells. However, it is unclear whether signaling cascades involved in eliciting these diverse cellular effects are also distinct. One of the best-characterized anti-viral signaling mechanisms of Type I IFNs is mediated by the IFN-inducible dsRNA activated protein kinase, PKR. Here, we have investigated the role of PKR and Type I IFNs in regulating viral clearance and CD8+ T cell response during primary and secondary viral infections. Our studies demonstrate differential requirement for PKR, in viral control versus elicitation of CD8+ T cell responses during primary infection of mice with lymphocytic choriomeningitis virus (LCMV). PKR-deficient mice mounted potent CD8+ T cell responses, but failed to effectively control LCMV. The compromised LCMV control in the absence of PKR was multifactorial, and linked to less effective CD8+ T cell-mediated viral suppression, enhanced viral replication in cells, and lower steady state expression levels of IFN-responsive genes. Moreover, we show that despite normal expansion of memory CD8+ T cells and differentiation into effectors during a secondary response, effective clearance of LCMV but not vaccinia virus required PKR activity in infected cells. In the absence of Type I IFN signaling, secondary effector CD8+ T cells were ineffective in controlling both LCMV and vaccinia virus replication in vivo. These findings provide insight into cellular pathways of Type I IFN actions, and highlight the under-appreciated importance of innate immune mechanisms of viral control during secondary infections, despite the accelerated responses of memory CD8+ T cells. Additionally, the results presented here have furthered our understanding of the immune correlates of anti-viral protective immunity, which have implications in the rational design of vaccines.


Assuntos
Interferon Tipo I/fisiologia , Listeriose/virologia , Coriomeningite Linfocítica/virologia , Vacínia/virologia , eIF-2 Quinase/fisiologia , Animais , Western Blotting , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunofluorescência , Proteínas de Homeodomínio/fisiologia , Memória Imunológica , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Listeriose/patologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Linfócitos T Citotóxicos/virologia , Vacínia/imunologia , Vacínia/patologia , Vaccinia virus/fisiologia , Replicação Viral
16.
Appl Environ Microbiol ; 78(7): 2213-20, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22287000

RESUMO

Salmonella enterica serovar Enteritidis strain E40 filaments were developed under conditions of a reduced water activity (a(w)) of 0.95 in tryptic soy broth (TSB) or tryptic soy agar (TSA) supplemented with 8% or 7% NaCl, respectively. Filament formation was accompanied by an increase of biomass without an increase in CFU and was affected by incubation temperature and the physical milieu. The greatest amount of filaments was recovered from TSA with 7% NaCl and incubation at 30°C. Within 2 h of transfer to fresh TSB, filaments started to septate into normal-sized cells, resulting in a rapid increase in CFU. S. Enteritidis E40 filaments were not more tolerant of low- or high-temperature stresses than nonfilamented control cells. However, there was greater survival of filaments in 10% bile salts after 24 to 48 h of incubation, during pH 2.0 acid challenge for 10 min, and under desiccation on stainless steel surfaces at 25°C and 75.5% relative humidity for 7 days. S. Enteritidis E40 filaments invaded and multiplied within Caco-2 human intestinal epithelial cells to a similar degree as control cells when a comparable CFU of filaments and control cells was used. S. Enteritidis E40 filaments established a successful infection in mice via intragastric inoculation. The filaments colonized the gastrointestinal tract and disseminated to the spleen and liver at levels comparable to those attained by control cells, even when animals were inoculated with 10- to 100-fold fewer CFU. To our knowledge this is the first demonstration of virulence of stress-induced Salmonella filaments in vitro and in vivo. Formation of filaments by Salmonella in food products and food processing environments is significant to food safety, because detection and quantitation of the pathogen may be compromised. The finding that these filaments are virulent further enhances their potential public health impact.


Assuntos
Resposta ao Choque Térmico , Salmonella enteritidis/crescimento & desenvolvimento , Salmonella enteritidis/patogenicidade , Animais , Biomassa , Células CACO-2/microbiologia , Contagem de Colônia Microbiana , Meios de Cultura , Dessecação , Feminino , Trato Gastrointestinal/microbiologia , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Salmonelose Animal/microbiologia , Salmonella enteritidis/ultraestrutura , Cloreto de Sódio/farmacologia , Temperatura , Virulência , Água
17.
Appl Environ Microbiol ; 78(18): 6704-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22798362

RESUMO

Salmonella enterica forms aseptate filaments with multiple nucleoids when cultured in hyperosmotic conditions. These osmotic-induced filaments are viable and form single colonies on agar plates even though they contain multiple genomes and have the potential to divide into multiple daughter cells. Introducing filaments that are formed during osmotic stress into culture conditions without additional humectants results in the formation of septa and their division into individual cells, which could present challenges to retrospective analyses of infectious dose and risk assessments. We sought to characterize the underlying mechanisms of osmotic-induced filament formation. The concentration of proteins and chromosomal DNA in filaments and control cells was similar when standardized by biomass. Furthermore, penicillin-binding proteins in the membrane of salmonellae were active in vitro. The activity of penicillin-binding protein 2 was greater in filaments than in control cells, suggesting that it may have a role in osmotic-induced filament formation. Filaments contained more ATP than did control cells in standardized cell suspensions, though the levels of two F(0)F(1)-ATP synthase subunits were reduced. Furthermore, filaments could septate and divide within 8 h in 0.2 × Luria-Bertani broth at 23°C, while nonfilamentous control cells did not replicate. Based upon the ability of filaments to septate and divide in this diluted broth, a method was developed to enumerate by plate count the number of individual, viable cells within a population of filaments. This method could aid in retrospective analyses of infectious dose of filamented salmonellae.


Assuntos
Pressão Osmótica , Salmonella enterica/citologia , Salmonella enterica/fisiologia , Estresse Fisiológico , Trifosfato de Adenosina/análise , Proteínas de Bactérias/análise , Meios de Cultura/química , DNA Bacteriano/análise , Proteínas de Ligação às Penicilinas/análise , ATPases Translocadoras de Prótons/análise , Salmonella enterica/química , Salmonella enterica/crescimento & desenvolvimento , Temperatura
18.
Vet Microbiol ; 264: 109280, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34808430

RESUMO

Histophilus somni is a Gram-negative coccobacillus that causes diffuse vasculitis and intravascular thrombosis that can lead to multiple organ failure in cattle. Macrophages are important cellular mediators of fibrin deposition and removal at sites of inflammation. It has become evident that macrophages and other cells release microparticles (MPs) that have an array of biological activities, including pro-coagulant activity. We sought to determine whether monocyte-derived macrophages exposed to H. somni in vitro release MPs that activate the clotting cascade in a manner that could lead to thrombus formation. Bovine monocyte-derived macrophages were incubated with H. somni (at a 10:1 ratio) in RPMI with 10% heat inactivated fetal bovine serum for 6 h at 37 °C with 5 % CO2. Membrane-shed MPs were isolated from the conditioned media, washed twice with Ca2+ and Mg2+ free HBSS, and pro-coagulant activity assessed by a one-step plasma clotting assay. We observed greater pro-coagulant activity for MPs from H. somni stimulated macrophages than from unstimulated controls. Microparticle pro-coagulant activity was inhibited by addition of an anti-tissue factor antibody. We also observed co-localization of fluorescein-labeled H. somni cells and annexin V staining as evaluated by confocal microscopy. These results demonstrate that exposure to H. somni cells causes bovine monocyte-derived macrophages to release MPs that contain tissue factor, the first such report for bovine macrophages. We infer that if similar events occur in vivo they could amplify thrombus formation in bovine histophilosis.


Assuntos
Fibrina , Macrófagos , Pasteurellaceae , Trombose , Animais , Bovinos , Doenças dos Bovinos/imunologia , Fibrina/metabolismo , Técnicas In Vitro , Macrófagos/imunologia , Pasteurellaceae/imunologia , Trombose/veterinária
19.
Mol Pharmacol ; 79(4): 724-34, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21252291

RESUMO

Bone marrow (BM) hematopoietic cells are selectively sensitive to polycyclic aromatic hydrocarbons (PAH) in vivo. 7,12-Dimethylbenz(a)anthracene (DMBA), but not benzo(a)pyrene (BP), depletes BM hematopoietic cells in C57BL/6 mice. This difference is due to a BP-selective aryl hydrocarbon receptor (AhR)-mediated recovery. Colony-forming unit assays show suppression of lymphoid progenitors by each PAH within 6 h but a subsequent recovery, exclusively after BP treatment. Suppression of myeloid progenitors (6 h) occurs only for DMBA. Each progenitor responded equally to DMBA and BP in congenic mice expressing the PAH-resistant AhR (AhR(d)). AhR, therefore, mediates this BP recovery in each progenitor type. These PAH suppressions depend on Cyp1b1-mediated metabolism. Paradoxically, few genes responded to DMBA, whereas 12 times more responded to BP. Progenitor suppression by DMBA, therefore, occurs with minimal effects on the general BM population. Standard AhR-mediated stimulations (Cyp1a1, Cyp1b1, Ahrr) were similar for each PAH and for the specific agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin but were absent in AhR(d) mice. A group of 12 such AhR responses was sustained from 6 to 24 h. A second, larger set of BP responses (chemokines, cytokines, cyclooxygenase 2) differed in two respects; DMBA responses were low and BP responses declined extensively from 6 to 24 h. A third cluster exhibited BP-induced increases in protective genes (Nqo1, GST-mu) that appeared only after 12 h. Conversion of BP to quinones contributes oxidative signaling not seen with DMBA. We propose that genes in this second cluster, which share oxidative signaling and AhR activation, provide the AhR-dependent protection of hematopoietic progenitors seen for BP.


Assuntos
Benzo(a)pireno/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Células Cultivadas , Hematopoese/fisiologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Fatores de Tempo
20.
Adv Funct Mater ; 21(10): 1863-1873, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-25558188

RESUMO

We report the design of polyelectrolyte multilayers (PEMs) that can be prefabricated on an elastomeric stamp and mechanically transferred onto biomedically-relevant soft materials, including medical-grade silicone elastomers (E'~450-1500 kPa; E'-elastic modulus) and the dermis of cadaver-skin (E'~200-600 kPa). Whereas initial attempts to stamp PEMs formed from poly(allylamine hydrochloride) and poly(acrylic acid) resulted in minimal transfer onto soft materials, we report that integration of micrometer-sized beads into the PEMs (thicknesses of 6-160 nm) led to their quantitative transfer within 30 seconds of contact at a pressure of ~196 kPa. To demonstrate the utility of this approach, PEMs were impregnated with a range of loadings of silver-nanoparticles and stamped onto the dermis of human cadaver-skin (a wound-simulant) that was subsequently incubated with bacterial cultures. Skin-dermis stamped with PEMs that released 0.25±0.01 µg cm-2 of silver ions caused a 6 log10 reduction in colony forming units of Staphylococcus epidermidis and Pseudomonas aeruginosa within 12 h. Significantly, this level of silver release is below that which is cytotoxic to NIH 3T3 mouse fibroblast cells. Overall, this study describes a general and facile approach for the functionalization of biomaterial surfaces without subjecting them to potentially deleterious processing conditions.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa