SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "L773:1098 5522 srt2:(2010-2014)"

Sökning: L773:1098 5522 > (2010-2014)

  • Resultat 1-10 av 75
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Bauza, Karolis, et al. (författare)
  • Efficacy of a Plasmodium vivax Malaria Vaccine Using ChAd63 and Modified Vaccinia Ankara Expressing Thrombospondin-Related Anonymous Protein as Assessed with Transgenic Plasmodium berghei Parasites
  • 2014
  • Ingår i: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 82:3, s. 1277-1286
  • Tidskriftsartikel (refereegranskat)abstract
    • Plasmodium vivax is the world's most widely distributed malaria parasite and a potential cause of morbidity and mortality for approximately 2.85 billion people living mainly in Southeast Asia and Latin America. Despite this dramatic burden, very few vaccines have been assessed in humans. The clinically relevant vectors modified vaccinia virus Ankara (MVA) and the chimpanzee adenovirus ChAd63 are promising delivery systems for malaria vaccines due to their safety profiles and proven ability to induce protective immune responses against Plasmodium falciparum thrombospondin-related anonymous protein (TRAP) in clinical trials. Here, we describe the development of new recombinant ChAd63 and MVA vectors expressing P. vivax TRAP (PvTRAP) and show their ability to induce high antibody titers and T cell responses in mice. In addition, we report a novel way of assessing the efficacy of new candidate vaccines against P. vivax using a fully infectious transgenic Plasmodium berghei parasite expressing P. vivax TRAP to allow studies of vaccine efficacy and protective mechanisms in rodents. Using this model, we found that both CD8+ T cells and antibodies mediated protection against malaria using virus-vectored vaccines. Our data indicate that ChAd63 and MVA expressing PvTRAP are good preerythrocytic-stage vaccine candidates with potential for future clinical application.
  •  
2.
  • Belmonte, Rodrigo, et al. (författare)
  • Role of Pathogen-Derived Cell Wall Carbohydrates and Prostaglandin E-2 in Immune Response and Suppression of Fish Immunity by the Oomycete Saprolegnia parasitica
  • 2014
  • Ingår i: Infection and Immunity. - 0019-9567 .- 1098-5522. ; 82:11, s. 4518-4529
  • Tidskriftsartikel (refereegranskat)abstract
    • Saprolegnia parasitica is a freshwater oomycete that is capable of infecting several species of fin fish. Saprolegniosis, the disease caused by this microbe, has a substantial impact on Atlantic salmon aquaculture. No sustainable treatment against saprolegniosis is available, and little is known regarding the host response. In this study, we examined the immune response of Atlantic salmon to S. parasitica infection and to its cell wall carbohydrates. Saprolegnia triggers a strong inflammatory response in its host (i. e., induction of interleukin-1 beta(1) [IL-1 beta(1)], IL-6, and tumor necrosis factor alpha), while severely suppressing the expression of genes associated with adaptive immunity in fish, through downregulation of T-helper cell cytokines, antigen presentation machinery, and immunoglobulins. Oomycete cell wall carbohydrates were recognized by fish leukocytes, triggering upregulation of genes involved in the inflammatory response, similar to what is observed during infection. Our data suggest that S. parasitica is capable of producing prostaglanding E-2 (PGE(2)) in vitro, a metabolite not previously shown to be produced by oomycetes, and two proteins with homology to vertebrate enzymes known to play a role in prostaglandin biosynthesis have been identified in the oomycete genome. Exogenous PGE(2) was shown to increase the inflammatory response in fish leukocytes incubated with cell wall carbohydrates while suppressing genes involved in cellular immunity (gamma interferon [IFN-gamma] and the IFN-gamma-inducible protein [gamma-IP]). Inhibition of S. parasitica zoospore germination and mycelial growth by two cyclooxygenase inhibitors (aspirin and indomethacin) also suggests that prostaglandins may be involved in oomycete development.
  •  
3.
  • Bielig, H, et al. (författare)
  • NOD-like receptor activation by outer-membrane vesicles (OMVs) from non-O1 non-O139 Vibrio cholerae is modulated by the quorum sensing regulator HapR
  • 2011
  • Ingår i: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 79:4, s. 1418-1427
  • Tidskriftsartikel (refereegranskat)abstract
    • Vibrio cholerae is an inhabitant of aquatic systems and one of the causative agents of severe dehydrating diarrhea in humans. It has also emerged as an important cause of different kinds of inflammatory responses and in particular, V. cholerae strains of the non-O1 non-O139 serogroups (NOVC) have been associated with such infections in human. We analyzed the potential of outer membrane vesicles (OMVs) derived from the NOVC strain V:5/04 to induce inflammatory responses in human host cells. V:5/04 OMVs were taken up by human epithelial cells and induced inflammatory responses. siRNA-mediated gene knock-down revealed that the inflammatory potential of NOVC OMVs was partially mediated by the nucleotide-binding domain, leucine rich repeat containing family member NOD1. Physiochemical analysis of the content of these OMVs, in conjunction with NOD1 and NOD2 reporter assays in HEK293T cells, confirmed the presence of both NOD1 and NOD2 active peptidoglycan in the OMVs. Furthermore, we show that deletion of the quorum sensing regulator HapR which mimics an infective life style, specifically reduced the inflammatory potential of the V:5/04 OMVs and their ability to activate NOD1 and NOD2. In conclusion, our study shows that NOVC OMVs elicit immune responses mediated by NOD1 and NOD2 in mammalian host cells. Moreover, we provide evidence that the quorum sensing machinery plays an important regulatory role in this process by attenuating the inflammatory potential of OMVs in infective conditions. This work thus identified a new facet of how Vibrio affects host immune responses and defines a role for the quorum sensing machinery in this process.
  •  
4.
  • Birchenough, George M. H., et al. (författare)
  • Altered Innate Defenses in the Neonatal Gastrointestinal Tract in Response to Colonization by Neuropathogenic Escherichia coli
  • 2013
  • Ingår i: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 81:9, s. 3264-3275
  • Tidskriftsartikel (refereegranskat)abstract
    • Two-day-old (P2), but not 9-day-old (P9), rat pups are susceptible to systemic infection following gastrointestinal colonization by Escherichia coli K1. Age dependency reflects the capacity of colonizing K1 to translocate from gastrointestinal (GI) tract to blood. A complex GI microbiota developed by P2, showed little variation over P2 to P9, and did not prevent stable K1 colonization. Substantial developmental expression was observed over P2 to P9, including upregulation of genes encoding components of the small intestinal (alpha-defensins Defa24 and Defa-rs1) and colonic (trefoil factor Tff2) mucus barrier. K1 colonization modulated expression of these peptides: developmental expression of Tff2 was dysregulated in P2 tissues and was accompanied by a decrease in mucin Muc2. Conversely, alpha-defensin genes were upregulated in P9 tissues. We propose that incomplete development of the mucus barrier during early neonatal life and the capacity of colonizing K1 to interfere with mucus barrier maturation provide opportunities for neuropathogen translocation into the bloodstream.
  •  
5.
  • Birchenough, George M. H., et al. (författare)
  • Altered innate defenses in the neonatal gastrointestinal tract in response to colonization by neuropathogenic Escherichia coli.
  • 2013
  • Ingår i: Infection and immunity. - 1098-5522. ; 81:9, s. 3264-75
  • Tidskriftsartikel (refereegranskat)abstract
    • Two-day-old (P2), but not 9-day-old (P9), rat pups are susceptible to systemic infection following gastrointestinal colonization by Escherichia coli K1. Age dependency reflects the capacity of colonizing K1 to translocate from gastrointestinal (GI) tract to blood. A complex GI microbiota developed by P2, showed little variation over P2 to P9, and did not prevent stable K1 colonization. Substantial developmental expression was observed over P2 to P9, including upregulation of genes encoding components of the small intestinal (α-defensins Defa24 and Defa-rs1) and colonic (trefoil factor Tff2) mucus barrier. K1 colonization modulated expression of these peptides: developmental expression of Tff2 was dysregulated in P2 tissues and was accompanied by a decrease in mucin Muc2. Conversely, α-defensin genes were upregulated in P9 tissues. We propose that incomplete development of the mucus barrier during early neonatal life and the capacity of colonizing K1 to interfere with mucus barrier maturation provide opportunities for neuropathogen translocation into the bloodstream.
  •  
6.
  • Bröms, Jeanette E, 1974-, et al. (författare)
  • IglG and IglI of the Francisella pathogenicity island are important virulence determinants of Francisella tularensis LVS
  • 2011
  • Ingår i: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 79:9, s. 3683-3696
  • Tidskriftsartikel (refereegranskat)abstract
    • The Gram-negative bacterium Francisella tularensis is the causative agent of tularemia, a disease intimately associated with the multiplication of the bacterium within host macrophages. This in turn requires the expression of Francisella pathogenicity island (FPI) genes, believed to encode a type VI secretion system. While the exact functions of many of the components have yet to be revealed, some have been found to contribute to the ability of Francisella to cause systemic infection in mice as well as to prevent phagolysosomal fusion and facilitate escape into the host cytosol. Upon reaching this compartment, the bacterium rapidly multiplies, inhibits activation of the inflammasome, and ultimately causes apoptosis of the host cell. In this study, we analyzed the contribution of the FPI-encoded proteins IglG, IglI, and PdpE to the aforementioned processes in F. tularensis LVS. The ΔpdpE mutant behaved similarly to the parental strain in all investigated assays. In contrast, ΔiglG and ΔiglI mutants, although they were efficiently replicating in J774A.1 cells, both exhibited delayed phagosomal escape, conferred a delayed activation of the inflammasome, and exhibited reduced cytopathogenicity as well as marked attenuation in the mouse model. Thus, IglG and IglI play key roles for modulation of the intracellular host response and also for the virulence of F. tularensis.
  •  
7.
  • Börgeson, Emma, et al. (författare)
  • Lipoxin A(4) inhibits porphyromonas gingivalis-induced aggregation and reactive oxygen species production by modulating neutrophil-platelet interaction and CD11b expression
  • 2011
  • Ingår i: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 79:4, s. 1489-1497
  • Tidskriftsartikel (refereegranskat)abstract
    • Porphyromonas gingivalis is an etiological agent that is strongly associated with periodontal disease, and it correlates with numerous inflammatory disorders, such as cardiovascular disease. Circulating bacteria may contribute to atherogenesis by promoting CD11b/CD18-mediated interactions between neutrophils and platelets, causing reactive oxygen species (ROS) production and aggregation. Lipoxin A(4) (LXA(4)) is an endogenous anti-inflammatory and proresolving mediator that is protective of inflammatory disorders. The aim of this study was to investigate the effect of LXA(4) on the P. gingivalis-induced activation of neutrophils and platelets and the possible involvement of Rho GTPases and CD11b/CD18 integrins. Platelet/leukocyte aggregation and ROS production was examined by lumiaggregometry and fluorescence microscopy. Integrin activity was studied by flow cytometry, detecting the surface expression of CD11b/CD18 as well as the exposure of the high-affinity integrin epitope, whereas the activation of Rac2/Cdc42 was examined using a glutathione S-transferase pulldown assay. The study shows that P. gingivalis activates Rac2 and Cdc42 and upregulates CD11b/CD18 and its high-affinity epitope on neutrophils, and that these effects are diminished by LXA(4). Furthermore, we found that LXA(4) significantly inhibits P. gingivalis-induced aggregation and ROS generation in whole blood. However, in platelet-depleted blood and in isolated neutrophils and platelets, LXA(4) was unable to inhibit either aggregation or ROS production, respectively. In conclusion, this study suggests that LXA(4) antagonizes P. gingivalis-induced cell activation in a manner that is dependent on leukocyte-platelet interaction, likely via the inhibition of Rho GTPase signaling and the downregulation of CD11b/CD18. These findings may contribute to new strategies in the prevention and treatment of periodontitis-induced inflammatory disorders, such as atherosclerosis.
  •  
8.
  • Calderon Toledo, Carla, et al. (författare)
  • Cross-Reactive Protection against Enterohemorrhagic Escherichia coli Infection by Enteropathogenic Escherichia coli in a Mouse Model.
  • 2011
  • Ingår i: Infection and Immunity. - 1098-5522. ; 79:6, s. 2224-2233
  • Tidskriftsartikel (refereegranskat)abstract
    • Enteropathogenic (EPEC) and enterohemorrhagic (EHEC) Escherichia coli are related attaching and effacing (A/E) pathogens. The genes responsible for the A/E pathology are encoded in a chromosomal pathogenicity island termed the locus of enterocyte effacement (LEE). Both pathogens share a high degree of homology in the LEE and additional 'O' islands. EHEC prevalence is much lower in EPEC endemic areas. This may be due to the development of antibodies against common EPEC and EHEC antigens. This study investigated the hypothesis that EPEC infections may protect against EHEC infections. We used a mouse model to inoculate BALB/c mice intragastrically, first with EPEC, followed by EHEC (E. coli O157:H7). Four control groups received either a non-pathogenic E. coli (NPEC) strain followed by EHEC (NPEC/EHEC), alternatively PBS/EHEC, EPEC/PBS or PBS/PBS. Mice were monitored for weight loss and symptoms. EPEC colonized the intestine after challenge and mice developed serum antibodies to intimin and E. coli secreted protein B (encoded in the LEE). Prechallenge with an EPEC strain had a protective effect after EHEC infection as few mice developed mild symptoms, from which they recovered. These mice had an increase in body weight similar to control animals and tissue morphology exhibited mild intestinal changes and normal renal histology. All mice that were not pre-challenged with the EPEC strain developed mild to severe symptoms after EHEC infection, weight loss as well as intestinal and renal histopathological changes. These data suggest that EPEC may protect against EHEC infection in this mouse model.
  •  
9.
  • Cerveny, Lukas, et al. (författare)
  • Tetratricopeptide Repeat Motifs in the World of Bacterial Pathogens : Role in Virulence Mechanisms
  • 2013
  • Ingår i: Infection and Immunity. - : American Society for Microbiology. - 0019-9567 .- 1098-5522. ; 81:3, s. 629-635
  • Forskningsöversikt (refereegranskat)abstract
    • The tetratricopeptide repeat (TPR) structural motif is known to occur in a wide variety of proteins present in prokaryotic and eukaryotic organisms. The TPR motif represents an elegant module for the assembly of various multiprotein complexes, and thus, TPR-containing proteins often play roles in vital cell processes. As the TPR profile is well defined, the complete TPR protein repertoire of a bacterium with a known genomic sequence can be predicted. This provides a tremendous opportunity for investigators to identify new TPR-containing proteins and study them in detail. In the past decade, TPR-containing proteins of bacterial pathogens have been reported to be directly related to virulence-associated functions. In this minireview, we summarize the current knowledge of the TPR-containing proteins involved in virulence mechanisms of bacterial pathogens while high-lighting the importance of TPR motifs for the proper functioning of class II chaperones of a type III secretion system in the pathogenesis of Yersinia, Pseudomonas, and Shigella.
  •  
10.
  • Chowdhury, Fahima, et al. (författare)
  • Concomitant enterotoxigenic Escherichia coli infection induces increased immune responses to Vibrio cholerae O1 antigens in patients with cholera in Bangladesh.
  • 2010
  • Ingår i: Infection and immunity. - 1098-5522. ; 78:5, s. 2117-24
  • Tidskriftsartikel (refereegranskat)abstract
    • Vibrio cholerae O1 and enterotoxigenic Escherichia coli (ETEC) are major bacterial pathogens that cause dehydrating disease requiring hospitalization of children and adults. The cholera toxin (CT) produced by V. cholerae O1 and the heat-labile toxin (LT) and/or heat-stable toxin (ST) of ETEC are responsible for secretory diarrhea. We have observed that about 13% of hospitalized diarrheal patients are concomitantly infected with V. cholerae O1 and ETEC. In order to understand the outcome of such dual infections on the clinical and immunological responses in cholera patients, we studied patients infected with V. cholerae O1 (group VC; n = 25), those infected with both V. cholerae O1 and ETEC (group VCET; n = 25), and those infected with ETEC only (group ET; n = 25). The VCET group showed more severe dehydration and had a higher intake of intravenous fluid and more vomiting than the ETEC group (P = 0.01 to 0.003). The VCET patients showed higher vibriocidal responses and increased antibody titers to cholera toxin and lipopolysaccharide (LPS) in plasma than did the V. cholerae O1 patients (P = 0.02 to <0.001). All responses in the V. cholerae O1 and in the VCET groups were more robust than those seen in the group infected with ETEC only (P = 0.01 to <0.001). We thus show that concomitant colonization with ETEC induces immune responses to V. cholerae antigens that are more robust than those seen with V. cholerae O1 infection alone. It is possible that LT or other factors expressed by ETEC may play a role as a mucosal adjuvant in enhancing the immune responses to V. cholerae O1.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 75
Typ av publikation
tidskriftsartikel (74)
forskningsöversikt (1)
Typ av innehåll
refereegranskat (75)
Författare/redaktör
Mörgelin, Matthias (4)
Wai, Sun Nyunt (4)
Thorlacius, Henrik (3)
Raghavan, Sukanya, 1 ... (3)
Henriques-Normark, B (3)
Fernandez, Carmen (2)
visa fler...
Singh, Birendra (2)
Riesbeck, Kristian (2)
Jeppsson, Bengt (2)
Rahman, Milladur (2)
Urban, Constantin F (2)
Pejler, Gunnar (2)
Holmgren, Jan, 1944 (2)
Herwald, Heiko (2)
Engstrand, L (1)
Contreras, J. (1)
Sinisalo, J. (1)
Borén, Thomas (1)
Uhlin, Bernt Eric (1)
Bergman, P. (1)
Baumgarten, Maria (1)
Nayeri, Fariba (1)
Cava, Felipe (1)
Normark, S (1)
Potempa, Jan (1)
Sundell, Kristina, 1 ... (1)
Sundh, Henrik, 1976 (1)
Björkman, Anders (1)
Jin, Chunsheng (1)
Bylund, Johan, 1975 (1)
Karlsson, Niclas G., ... (1)
Röhm, Marc (1)
Syk, Ingvar (1)
Sjökvist Ottsjö, Lou ... (1)
Lundin, Samuel B, 19 ... (1)
Svennerholm, Ann-Mar ... (1)
Anar, Burcu (1)
Billker, Oliver (1)
Kallenius, G (1)
Deloron, Philippe (1)
Massougbodji, Achill ... (1)
Buhlin, K (1)
Adlerberth, Ingegerd ... (1)
Wold, Agnes E, 1955 (1)
Svanborg, Catharina (1)
Lindberg, Erika, 197 ... (1)
Nilsson, Kristina (1)
Blom, Anna (1)
Lindmark, B. (1)
Magnusson, Karl-Eric (1)
visa färre...
Lärosäte
Umeå universitet (22)
Göteborgs universitet (13)
Karolinska Institutet (13)
Lunds universitet (12)
Stockholms universitet (8)
Örebro universitet (4)
visa fler...
Sveriges Lantbruksuniversitet (4)
Kungliga Tekniska Högskolan (2)
Linköpings universitet (2)
Linnéuniversitetet (2)
Uppsala universitet (1)
visa färre...
Språk
Engelska (75)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (46)
Naturvetenskap (17)
Lantbruksvetenskap (4)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy