As expected, in Atg5−/− MEFs, LC3-II was never detected

w

As expected, in Atg5−/− MEFs, LC3-II was never detected

whatever the cell culture conditions because the presence of Atg5 is absolutely required for the LC3 recruitment onto autophagosome membrane [19]. In WT MEFs infected with B. abortus or with B. melitensis, the relative abundance of LC3-I and LC3-II at 18 h p.i. did not change when compared to non-infected MEFs (Figure 1B). Figure 1 Relative abundance of LC3B-I and LC3B-II in WT MEFs and in Atg5 click here −/− MEFs as determined by immunoblotting. A. Cells were maintained in DMEM/FCS (F), starved for 2 h in EBSS (S) or incubated for 5 h in the presence of 100 nM bafilomycin (Baf). B. Cells were infected with B. abortus (BA) or with B. melitensis (BM) for 18 h or left non infected (Ctl). Replication of B. abortus- and B. melitensis-mCherry in Atg5−/− fibroblasts We studied the contribution of the macroautophagic pathway on the replication of Brucellae using Atg5-deficient MEFs. First, we infected cells with B. abortus-mCherry (Figure 2A) or with B. melitensis-mCherry

(Figure 2B) for 1 h at a multiplicity of infection (MOI) of 300. After inoculation, the medium was removed and replaced by a medium containing gentamicin to kill extracellular bacteria. selleck kinase inhibitor As it can be seen on micrographs taken after increasing times postinfection, B. abortus-mCherry is able to enter, survive and replicate in MEFs, even in Atg5-deficient MEFs. In both cell lines, at 6 h p.i, there are only a few bacteria per infected cell but this number massively increases between 12 and 18 h p.i. and at 24 h p.i., the bacteria are so abundant that it is difficult to enumerate them. B. melitensis-mCherry is also able to replicate in both WT MEFs and Atg5−/− MEFs. However, it is clear that www.selleck.co.jp/products/cobimetinib-gdc-0973-rg7420.html the number of bacteria per infected cell at 24 h p.i. is lower compared to B. abortus-mCherry. Statistical analysis of these observations revealed that there is no significant difference in the number of B. abortus-mCherry per infected cell between the Atg5-deficient MEFs and the WT MEFs whatever the time postinfection (Figure 3A). In contrast, the number of B. melitensis-mCherry

per infected cell significantly increased in Atg5−/− MEFs when compared to WT MEFs at 9 h, 18 h and 24 h p.i. (Figure 3B). These data demonstrate that both Brucella strains can survive and replicate when the conventional Atg5-dependent macroautophagic pathway is impaired. Atg5-deficient cells seem to be even more permissive for B. melitensis replication than WT MEFs. Figure 2 Fluorescence microscopy analysis of WT MEFs and Atg5 −/− MEFs infected with B. abortus -mCherry (A) or with B. melitensis- mCherry (B). MEFs were infected for 1 h with Brucella-mCherry at an MOI of 300 and observed at 6 h, 12 h, 18 h and 24 h p.i. The nuclei were stained with DAPI. Figure 3 Quantification of the infection of WT MEFs and Atg5 −/− MEFs with B. abortus -mCherry (A) or with B. melitensis- mCherry (B). MEFs were infected for 1 h with Brucella-mCherry at an MOI of 300.

Group-I included 15 strains that did not enter cells, formed no p

Group-I included 15 strains that did not enter cells, formed no plaques and had no phospholipase activity. Group-II consisted of only one strain entering cells, forming no plaques and only expressing PI-PLC activity. Group-III comprised nine

strains entering cells, forming no plaques and only expressing PC-PLC activity. In this new analysis, the previously described Group-IV [7] has now been divided into 3 sub-Groups. The new Group-IV included nine strains forming plaques but fewer than virulent strains (mean 3 log versus 5). Three out of 9 strains were also characterized by a very low level of PC- and PI-PLC. The new Group-V comprised six strains also forming plaques but fewer than virulent strains ALK inhibitor and characterized by their very high PI-PLC activity. Finally, Group-VI contained three strains forming plaques within Sunitinib 48 h. In contrast the other strains formed plaques within 24 h, classic time necessary

to count the plaque number. Genotypic characterisation of the low-virulence strains Sequencing the prfA, plcA, plcB, inlA and inlB genes allowed us to observe that some phenotypes correlate with genotypic mutations which have been demonstrated to be the cause of the low virulence (Table 1) [7]. The sequences of the PrfA, InlA and ActA fragment were compared to those of the EGDe strain (serotype 1/2 – GenBank accession number AL591824) or F2365 strain (serotype 4 – GenBank accession number AE017262), according to the serotypes of the

strains. The phenotypic Group-I strains exhibited mutations in PrfA compared to the EGDe strain and were subdivised into 2 genotypic Groups: the PrfAK220T (genotypic Group-Ia) and the truncated PrfAΔ174-237 (genotypic below Group-Ib) previously described [8, 11]. One strain (NP26) exhibited a new putative causal mutation in prfA, K130Q, and is the only one of serotype 4b exhibiting a PrfA mutation (herein defined as genotypic Group-Ic). Two genotypic Groups were also identified for the phenotypic Group-III strains. One harbored exactly the same mutations in the plcA, inlA and inlB genes, characteristic of the previously genotypic Group-IIIa [8]. Only one strain (AF105) belonged to Group-IIIb and harbored a mutation at least in the inlA gene. No genotyping Group has been defined for the phenotypic Groups-II because this Group is formed by only one strain. The Group-IV, -V and –VI strains did not exhibit specific DNA sequence of the prfA, inlA and actA fragment genes, that allowed us to assign genotyping Groups. No causal mutations could have been displayed explaining the low virulence of these Groups. PFGE profiles To study the genetic relationships between the low-virulence strains, the 43 low-virulence strains were compared with 49 virulent strains (based on both the mouse s.c.

In: Mok DWS, Mok MC (eds) Cytokinins: chemistry, activity and fun

In: Mok DWS, Mok MC (eds) Cytokinins: chemistry, activity and function. CRC Press, Boca Raton, pp 179–195 Sathish P, Withana N, Biswas M, Bryant C, Templeton K, Al-Wahb M, Smith-Espinoza C, Roche JR, Elborough KM, Phillips JR (2007)

Transcriptome analysis reveals season-specific rbcS gene expression profiles in diploid perennial ryegrass (Lolium perenne L.). Plant Biotechnol J 5(1):146–161CrossRefPubMed ATM/ATR cancer Schmulling T, Schäfer S, Romanov G (1997) Cytokinins as regulators of gene expression. Physiol Plant 100:505–519CrossRef Soitama AJ, Piippo M, Allahverdiyea Y, Battchikova N, Aro EM (2008) Light has a specific role in modulating Arabidopsis gene expression at low temperature. BMC Plant Biol 8(1):13CrossRef Surpin M, Larkin RM, Chory J (2002) Signal transduction between the chloroplast and the nucleus. Plant Cell 14:S327–S328PubMed Synková H, Van Loven K, Pospišilová J, Valcke R (1999) Photosynthesis of transgenic Pssu-ipt tobacco. J Plant Physiol 155:173–182 Synková H, Pechova R, Valcke R (2003) Changes in chloropast ultrastructure

in Pssu-ipt tobacco during plant ontogeny. Photosynthetica 41:117–126CrossRef Synková H, Schnablová R, Polanská L, Hušák M, Šiffel P, Vácha F, Malbeck J, Macháchová I, Nebesářová J (2006) Three-dimensional reconstruction of anomalous chloroplasts in transgenic ipt tobacco. Planta 223(4):659–671CrossRefPubMed Thellin O, Zorzi W, Lakaye B, De Borman B, Coumand B, Hennen G, Grisar T, Igout A, Heinen E (1999) Housekeeping genes as internal standards: use and limits. J Biotechnol 75:291–295CrossRefPubMed Aloxistatin Ulvskov P, Nielsen T, Seiden P, Marcussen J (1992) Cytokinins and leaf development in sweet pepper (Capsicum annuum L.). Planta 188:70–77CrossRef Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, Speleman Astemizole F (2002) Accurate normalisation of real-time quantitative

RT-PCR data by geometric averaging of multiple internal control genes. Genome Biol 3(7):RESEARCH0034.1–0034.11 Volkov RA, Panchuk II, Schôffl F (2003) Heat-stress-dependency and developmental modulation of gene expression: the potential of house-keeping genes as internal standards in mRNA expression profiling using real-time RT-PCR. J Exp Bot 54(391):2343–2349CrossRefPubMed Werner T, Motyka V, Strnad M, Schmülling T (2001) Regulation of plant growth by cytokinin. Plant Biol 98(18):10487–10492 Werner T, Holst K, Pörs Y, Guivarc′h A, Mustroph A, Chrique D, Grimm B, Schmülling T (2008) Cytokinin deficiency causes distinct changes of sink and source parameters in tobacco shoots and roots. J Exp Bot 59:2659–2672. doi:10.​1093/​jxb/​ern134 Ya OZ, Selivankina SY, Yamburenko MV, Zubkova NK, Kulaeva ON, Kusnetsov VV (2005) Cytokinins activate transcription of chloroplast genes.

Haplotype properties differ between different antibiotic exposure

Haplotype properties differ between different antibiotic exposures Diversification of P. aeruginosa LESB58 in ASM cultured with and without the various antibiotics was observed only with respect to colony morphology, pyocyanin production and antibiotic susceptibilities (Table 1). The culture of LESB58 in ASM with sub-inhibitory concentrations of ceftazidime

and colistin led to diversity in antimicrobial susceptibilities, changes in colony morphology and a loss of pyocyanin production (Table 1). LESB58 cultured in the presence of these antibiotics, generated more isolates that were outside the normal range of the antibiotic sensitivity profiles of LESB58 controls (Figure 3). In addition, exposure to azithromycin and tobramycin promoted increased cross-resistance Selleck Saracatinib to other antibiotics (Table 1, Figure 3). There

was no variation in the auxotrophic phenotype in the isolates analysed in all experimental and control groups (LESB58 has an auxotrophic phenotype). The populations exposed to meropenem exhibited no clear phenotypic diversification (Table 1 and Figure 2). Figure 3 Variations in zones of inhibition within LESB58 populations. The 120 LESB58 isolates obtained from the triplicate ASM cultures were assessed for susceptibility to six commonly used antibiotics (ceftazidime, ciprofloxacin, click here colistin, meropenem, tazobactam/piperacillin and tobramycin). Boxplots showing the range in the diameter of the zones of inhibition to these antibiotics are presented. 1. LB (18 hours) 2. ASM 3. ASM with ceftazidime 4. ASM with colistin 5. ASM with meropenem 6. ASM with tobramycin 7. ASM with azithromycin 8. Normal range of LESB58 (Groups 1–8: n = 120). The red line represents the cut-off for the the sensitivity of P. aeruginosa to the antibiotics tested, in accordance with the guidelines of Andrews

and Howe [37]. The values above the red line denote a higher sensitivity to antibiotics and the values below the line denote a higher resistance. Table 1 Number of isolates in each group (total of 120) exhibiting each of the traits measured   Colony morphology Virulence Mutations Outside normal range of antimicrobials susceptibility Culture Green non-mucoid Straw non-mucoid Pyocyanin Hypermutability Ceftazidime Ciprofloxacin Tobramycin Meropenem Colistin Tazobactam/piperacillin ASM 120 0 117 0 3 0 19 0 2 8 ASM + CAZ 110 10 92 0 16 19 20 18 10 11 ASM + CT 113 7 84 0 17 37 29 15 7 9 ASM + AZT 120 0 120 0 0 16 34 0 4 4 ASM + MEM 120 0 118 0 1 8 4 0 0 1 ASM + TOBI 118 2 119 0 1 24 69 3 22 1 LB (18 hours) 120 0 120 1 0 0 0 0 0 0 Isolates that were characterized as being outside the normal range of antimicrobial susceptibility typically observed in LESB58, included isolates that had either an increased or reduced susceptibility to the antibiotic under test. ASM = Artificial Sputum Medium, LB = Luria Bertani, CAZ = Ceftazidime, CT = Colistin, AZT = Azithromycin, MEM = Meropenem and TOBI = Tobramycin.

Kremer N, Voronin D, Charif D, Mavingui P, Mollereau B, Vavre F:

Kremer N, Voronin D, Charif D, Mavingui P, Mollereau B, Vavre F: Wolbachia interferes with ferritin expression and iron metabolism in insects. PLoS Pathog 2009, 5:e1000630.PubMedCrossRef 15. Brennan LJ, Keddie BA, Braig HR, Harris HL: The endosymbiont Wolbachia pipientis induces the expression of host antioxidant proteins in an Aedes albopictus cell line. PLoS One 2008, 3:e2083.PubMedCrossRef 16. Molina-Cruz A, DeJong RJ, Charles B, Gupta L, Kumar S, Jaramillo-Gutierrez G, Barillas-Mury C: Reactive oxygen species modulate Anopheles gambiae immunity against bacteria and Plasmodium . J Biol Chem 2008, 283:3217–3223.PubMedCrossRef 17. Ryu J-H, Ha E-M, Lee W-J: Innate immunity and gut-microbe

mutualism in Drosophila . Dev Comp Immunol 2010, 34:369–376.PubMedCrossRef find more 18. Cabozantinib cost Teixeira L, Ferreira A, Ashburner M: The bacterial symbiont Wolbachia induces resistance to RNA viral infections in Drosophila melanogaster . PLoS Biol 2008, 6:e2.PubMedCrossRef 19. Osborne SE, Leong YS, O’Neill SL, Johnson KN: Variation in antiviral protection mediated by different Wolbachia strains in Drosophila simulans . PLoS Pathog 2009,

5:e1000656.PubMedCrossRef 20. Moreira LA, Iturbe-Ormaetxe I, Jeffery JA, Lu G, Pyke AT, Hedges LM, Rocha BC, Hall-Mendelin S, Day A, Riegler M, Hugo LE, Johnson KN, Kay BH, McGraw EA, van den Hurk AF, Ryan PA, O’Neill SL: A Wolbachia symbiont in Aedes aegypti limits infection with dengue, Chikungunya , and Plasmodium . Cell 2009, 139:1268–1278.PubMedCrossRef 21. Bian G, Xu Y, Lu P, Xie Y, Xi Z: The endosymbiotic bacterium Wolbachia induces resistance to dengue virus in Aedes aegypti . PLoS Pathog 2010, 6:e1000833.PubMedCrossRef 22. Iturbe-Ormaetxe I, O’Neill SL: Wolbachia -host interactions: connecting phenotype to genotype.

Curr Opin Microbiol 2007, 10:221–224.PubMedCrossRef 23. Xi Z, Gavotte L, Xie Y, Dobson SL: Genome-wide analysis of the interaction between the endosymbiotic bacterium Wolbachia and its Drosophila host. BMC Genomics 2008, 9:1.PubMedCrossRef 24. Ikeya T, Broughton S, Alic N, Grandison R, Partridge L: The endosymbiont Wolbachia increases insulin/IGF-like signalling in Drosophila . Proc Biol Sci 2009, 276:3799–37807.PubMedCrossRef 25. Dedeine F, Vavre F, Shoemaker DD, Boulétreau M: Intra-individual coexistence of a Wolbachia strain required for host oogenesis with two strains inducing cytoplasmic incompatibility enough in the wasp Asobara tabida . Evolution 2004, 58:2167–2174.PubMed 26. Zouache K, Voronin D, Tran-Van V, Mavingui P: Composition of bacterial communities associated with natural and laboratory populations of Asobara tabida infected with Wolbachia . Appl Environ Microbiol 2009, 75:3755–3764.PubMedCrossRef 27. David J: A new medium for rearing Drosophila in axenic conditions. Drosophila Info Serv 1962, 36:128. 28. Zhu YY, Machleder EM, Chenchik A, Li R, Siebert PD: Reverse transcriptase template switching: a SMART approach for full-length cDNA library construction. Biotechniques 2001, 30:892–897.PubMed 29.

Enteritidis PT4 P125109 chromosome and predicted as absent in the

Enteritidis PT4 P125109 chromosome and predicted as absent in the test strain. In red, genes absent in the S. Enteritidis PT4 P125109 chromosome and predicted as present in the test strain. In white, genes present or absent in both reference and test strains. Only those isolates for which any divergence is predicted are shown. S. Enteritidis PT4 P125109 results are shown as C646 molecular weight reference.

Detailed analysis of the genes within the DG showed that prophage-like elements constitute the major source of genetic variation distinguishing these S. Enteritidis isolates. However, this analysis also revealed some interesting differences in metabolic potential and in genes associated with restriction-modification systems (discussed below). S. Enteritidis variable prophage-like regions within the DG Of the annotated prophages from S. Enteritidis PT4 P125109 represented on the array one Kenyan and 4 Uruguayan isolates lacked ϕSE20 (Region 4 in our analysis), a ~41 kb phage similar to ϕST64B. Phage SE20 is thought to be intact

and a recent acquisition in S. Enteritidis PT4 P125109 and like ϕST64B, it carries fragments of the sopE and orgA genes, which have been implicated in Salmonella virulence [27, 29]. Two of the 4 Uruguayan isolates that lack ϕSE20 were isolated from human infections more than 5 years before the beginning of the epidemic in Uruguay (31/88 and 8/89), whereas the other 2 were from food samples, one from before (53/94) and the other from the middle (206/99) of the epidemic. Similarly, Porwollik and collaborators have reported that this phage https://www.selleckchem.com/products/Cyclopamine.html (called ϕST64B in their work) is absent in strains of S. Enteritidis isolated more than 50 years ago and suggested that acquisition of this phage may be related to the emergence of S. Enteritidis as being epidemic worldwide [21]. We corroborated the presence of ϕSE20 among the 29 Uruguayan isolates by PCR using two set of ϕSE20-specific primers that amplify fragments of sb9 and sb41 (SEN1935 and SEN1993 respectively). Only isolates 31/88, 8/89,

53/94 and 206/99 were negative validating IMP dehydrogenase the microarray results. We extended the PCR screening with sb41 primers to another 85 S. Enteritidis isolates from the original sample set, which included 28 isolates from human gastroenteritis, 30 isolates from invasive human disease and 27 isolates from non-human origin (including the 2 other pre-epidemic isolates that had not been included in the CGH analysis). Among them we found only 4 other isolates that lack sb41, i.e. 50/99 and 211/00 originating from food, 107/99 from enteric disease and 209/01 from invasive infection. In summary, we found that only 5 out of 108 isolates tested from the epidemic and post-epidemic periods lack ϕSE20, whereas 3 out of 6 pre-epidemic isolates lack this phage. This provides further support for the idea that the presence of ϕSE20 is a marker for the emergence of particular isolates as epidemic strains [21, 27]. It has been proposed that S.

: Determinants of the human infant intestinal microbiota after th

: Determinants of the human infant intestinal microbiota after the introduction of first complementary foods in infant samples from five European centres. Microbiology 2011,157(Pt JAK2 inhibitors clinical trials 5):1385–1392.PubMedCrossRef 27. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al.: A core gut microbiome

in obese and lean twins. Nature 2009,457(7228):480–484.PubMedCrossRef 28. Suau A, Bonnet R, Sutren M, Godon JJ, Gibson GR, Collins MD, Dore J: Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl Environ Microbiol 1999,65(11):4799–4807.PubMed 29. Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent LT, Ley RE: Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci USA 2011,108(Suppl

1):4578–4585.PubMedCrossRef 30. Favier CF, Vaughan EE, De Vos WM, Akkermans AD: Molecular monitoring of succession of bacterial communities in human neonates. Appl Environ Microbiol 2002,68(1):219–226.PubMedCrossRef 31. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO: Development of the human infant intestinal microbiota. PLoS Biol 2007,5(7):e177.PubMedCrossRef 32. Bager P, Wohlfahrt J, Westergaard T: Caesarean delivery and risk of atopy and allergic disease: meta-analyses. Clin Exp Allergy 2008,38(4):634–642.PubMedCrossRef 33. Kummeling I, Stelma FF, Dagnelie PC, Snijders BE, Penders J, Huber M, van Ree R, van den Brandt selleck chemicals PA, Thijs C: Early life exposure to antibiotics and the subsequent development of eczema, wheeze, and allergic sensitization

in the first 2 years of life: the KOALA Birth Cohort Study. Pediatrics 2007,119(1):e225–231.PubMedCrossRef 34. Lewis SA, Britton JR: Consistent effects of high socioeconomic status and low Non-specific serine/threonine protein kinase birth order, and the modifying effect of maternal smoking on the risk of allergic disease during childhood. Respir Med 1998,92(10):1237–1244.PubMedCrossRef 35. Kull I, Bohme M, Wahlgren CF, Nordvall L, Pershagen G, Wickman M: Breast-feeding reduces the risk for childhood eczema. J Allergy Clin Immunol 2005,116(3):657–661.PubMedCrossRef 36. Biasucci G, Rubini M, Riboni S, Morelli L, Bessi E, Retetangos C: Mode of delivery affects the bacterial community in the newborn gut. Early Hum Dev 2010,86(Suppl 1):13–15.PubMedCrossRef 37. Huurre A, Kalliomaki M, Rautava S, Rinne M, Salminen S, Isolauri E: Mode of delivery – effects on gut microbiota and humoral immunity. Neonatology 2008,93(4):236–240.PubMedCrossRef 38. Zhou X, Bent SJ, Schneider MG, Davis CC, Islam MR, Forney LJ: Characterization of vaginal microbial communities in adult healthy women using cultivation-independent methods. Microbiology (Reading, England) 2004,150(Pt 8):2565–2573.CrossRef 39.

Br J Nutr 2009, 101:1673–1678 PubMedCrossRef 104 Engels HJ, Kolo

Br J Nutr 2009, 101:1673–1678.PubMedCrossRef 104. Engels HJ, Kolokouri I, Cieslak TJ, Wirth JC: Effects of ginseng

supplementation on TAM Receptor inhibitor supramaximal exercise performance and short-term recovery. J Strength Cond Res 2001, 15:290–295.PubMed 105. Eschbach LF, Webster MJ, Boyd JC, McArthur PD, Evetovich TK: The effect of siberian ginseng (Eleutherococcus senticosus) on substrate utilization and performance. Int J Sport Nutr Exerc Metab 2000, 10:444–451.PubMed 106. Ferrando A, Vila L, Voces JA, Cabral AC, Alvarez AI, Prieto JG: Effects of ginseng extract on various haematological parameters during aerobic exercise in the rat. Planta Med 1999, 65:288–290.PubMedCrossRef 107. Ferrando A, Vila L, Voces JA, Cabral AC, Alvarez AI, Prieto JG: Effects of a standardized Panax ginseng extract on the skeletal muscle of the rat: a comparative study in animals at rest and under exercise. Planta Med 1999, 65:239–244.PubMedCrossRef 108. Ziemba AW, Chmura J, Kaciuba-Uscilko H, Nazar K, Wisnik P, Gawronski W: Ginseng treatment improves psychomotor performance at rest and during graded exercise in young athletes. Int J Sport selleck chemicals llc Nutr 1999, 9:371–377.PubMed 109. Allen JD, McLung J, Nelson AG, Welsch M: Ginseng supplementation does not enhance healthy young adults’ peak aerobic exercise performance. J Am Coll Nutr 1998, 17:462–466.PubMed

110. Engels HJ, Wirth JC: No ergogenic effects of ginseng (Panax ginseng C.A. Meyer) during graded maximal aerobic exercise. J Am Diet Assoc 1997, 97:1110–1115.PubMedCrossRef 111. Pieralisi G, Ripari P, Vecchiet L: Effects of a standardized ginseng extract combined with dimethylaminoethanol bitartrate, vitamins, minerals, DNA ligase and trace elements on physical performance during exercise. Clin Ther 1991, 13:373–382.PubMed 112. Karlic H, Lohninger A: Supplementation of L-carnitine in athletes: does it make sense? Nutrition 2004, 20:709–715.PubMedCrossRef 113. Pauly DF, Pepine CJ: D-Ribose as a supplement for cardiac energy metabolism. J Cardiovasc

Pharmacol Ther 2000, 5:249–258.PubMedCrossRef 114. Kerksick C, Rasmussen C, Bowden R, Leutholtz B, Harvey T, Earnest C, Greenwood M, Almada A, Kreider R: Effects of ribose supplementation prior to and during intense exercise on anaerobic capacity and metabolic markers. Int J Sport Nutr Exerc Metab 2005, 15:653–664.PubMed 115. Kreider RB, Melton C, Greenwood M, Rasmussen C, Lundberg J, Earnest C, Almada A: Effects of oral D-ribose supplementation on anaerobic capacity and selected metabolic markers in healthy males. Int J Sport Nutr Exerc Metab 2003, 13:76–86.PubMed 116. Berardi JM, Ziegenfuss TN: Effects of ribose supplementation on repeated sprint performance in men. J Strength Cond Res 2003, 17:47–52.PubMed 117. Dunne L, Worley S, Macknin M: Ribose versus dextrose supplementation, association with rowing performance: a double-blind study. Clin J Sport Med 2006, 16:68–71.PubMedCrossRef 118.

World J Gastroenterol 2008,14(16):2511–2516 CrossRef 23 Smits HH

World J Gastroenterol 2008,14(16):2511–2516.CrossRef 23. Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, van Capel TM, Zaat BA, Yazdanbakhsh M, Wierenga EA, van Kooyk Y, Kapsenberg ML: Selective probiotic bacteria induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. J Allergy Clin Immunol 2005,115(6):1260–1267.PubMedCrossRef 24. Kim SY, Kim JY, Kim SH,

Bae HJ, Yi H, Yoon SH, Koo BS, Kwon M, Cho JY, Lee CE, Hong S: Surfactin from Bacillus subtilis displays antiproliferative effect via apoptosis induction, cell cycle arrest and survival signaling suppression. Decitabine FEBS Lett 2007, 581:865–871.PubMedCrossRef 25. Koonin EV, Aravind L: Origin and evolution of eukaryotic apoptosis: the bacterial connection. Cell Death Differ 2002, 9:394–404.PubMedCrossRef 26. Hooper LV, Gordon JI: Commensal host-bacterial relationships in the gut. Science 2001, 292:1114–1118.CrossRef Authors’ contributions QG and JW participated in the design

of the experiment and its implementation, data analysis, and wrote the manuscript. LQ carried out this website bacteria culture, western blotting, real-time PCR and ELISA. TW was involved in the cell culture, SiRNA transient transfection, IL-10 neutralization, stimulation of cells, PI assay, Caspase-3 activity Thiamet G assay and DNA fragmentation analyses. All authors have read and approved the final manuscript. The authors declare no conflict of interest.”
“Background In recent years, coagulase-negative Staphylococcus epidermidis ( Se)

has become the leading cause of infections related to indwelling medical devices such as vascular catheters, prosthetic joints and artificial heart valves [1, 2]. Pathogenicity of Se is attributed to its formation of biofilm on the surface of medical devices, thereby enhancing Se resistance to antibiotics and host defenses in this setting [3, 4]. In general, Se biofilm formation is a two-step process, in which bacteria first adhere to the surface (initial attachment phase) and subsequently form cell–cell aggregates and a multilayered architecture (accumulative phase) [5, 6]. One autolysin protein, AtlE, facilitates bacterial attachment to the surface of medical devices and dictates pathogenesis for Se biofilm-associated infections in vivo [7, 8]. In the accumulative phase, the polysaccharide intercellular adhesin (PIA), a linear poly-Nacetyl-1,6-β-glucosamine (PNAG) encoded by the icaADBC locus, is the major pathogenic determinant for intercellular adhesion [9, 10].

PubMed 18 Gerner EW, Meyskens FL Jr: Polyamines and cancer: old

PubMed 18. Gerner EW, Meyskens FL Jr: Polyamines and cancer: old molecules, new understanding. Nat Rev Cancer 2004, 4:781–792.PubMed 19. Erdman SH, Ignatenko NA, Powell MB, Blohm-Mangone KA, Holubec H, Guillen-Rodriguez JM, Gerner EW: APC-dependent changes in expression of genes influencing polyamine metabolism, and consequences for gastrointestinal carcinogenesis, in the Min mouse. Carcinogenesis 1999, 20:1709–1713.PubMed 20. Becciolini A, Porciani S, Lanini A, Balzi M, Cionini L, Bandettini L: Polyamine levels in healthy and tumor

tissues of patients with colon adenocarcinoma. Dis Colon Rectum 1991, 34:167–173.PubMed 21. Canizares F, Salinas J, de las Heras M, Diaz J, Tovar I, Martinez P, Penafiel R: Prognostic value of ornithine decarboxylase and Alisertib chemical structure polyamines in human breast cancer: correlation with clinicopathologic parameters. Clin Cancer Res 1999, 5:2035–2041.PubMed 22. Radford DM, Nakai H, Eddy RL, Haley LL, Byers MG, Henry WM, Lawrence DD, Porter CW,

Shows TB: Two chromosomal locations for human ornithine decarboxylase gene sequences and elevated expression in colorectal neoplasia. Cancer Res 1990, 50:6146–6153.PubMed 23. Kingsnorth AN, Lumsden AB, Wallace HM: Polyamines in colorectal cancer. Br J Surg 1984, 71:791–794.PubMed 24. LaMuraglia GM, Lacaine F, Malt RA: High ornithine decarboxylase activity and polyamine levels in human colorectal neoplasia. Ann Surg 1986, 204:89–93.PubMed 25. Takenoshita S, Matsuzaki S, MAPK inhibitor almost Nakano G, Kimura H, Hoshi H, Shoda H, Nakamura T: Selective elevation of the N1-acetylspermidine level in human colorectal adenocarcinomas. Cancer Res 1984, 44:845–847.PubMed 26. Moulinoux JP, Quemener V, Khan NA, Delcros JG, Havouis R: Spermidine uptake by erythrocytes from normal and Lewis lung carcinoma (3LL) grafted mice: I. In vitro study. Anticancer Res 1989, 9:1057–1062.PubMed 27. Uda K, Tsujikawa T, Fujiyama Y, Bamba T: Rapid absorption of luminal polyamines in

a rat small intestine ex vivo model. J Gastroenterol Hepatol 2003, 18:554–559.PubMed 28. Bardocz S, Brown DS, Grant G, Pusztai A: Luminal and basolateral polyamine uptake by rat small intestine stimulated to grow by Phaseolus vulgaris lectin phytohaemagglutinin in vivo. Biochim Biophys Acta 1990, 1034:46–52.PubMed 29. Bardocz S, Grant G, Brown DS, Ralph A, Pusztai A: Polyamines in food–implications for growth and health. J Nutr Biochem 1993, 4:66–71. 30. Soda K, Kano Y, Sakuragi M, Takao K, Lefor A, Konishi F: Long-term oral polyamine intake increases blood polyamine concentrations. J Nutr Sci Vitaminol (Tokyo) 2009, 55:361–366. 31. Soda K, Dobashi Y, Kano Y, Tsujinaka S, Konishi F: Polyamine-rich food decreases age-associated pathology and mortality in aged mice. Exp Gerontol 2009, 44:727–732.PubMed 32. Brodal BP, Eliassen KA, Ronning H, Osmundsen H: Effects of dietary polyamines and clofibrate on metabolism of polyamines in the rat. J Nutr Biochem 1999, 10:700–708.PubMed 33.