Nature 2007, 445:106–110 PubMedCrossRef 7 Ponti D, Costa A, Zaff

Nature 2007, 445:106–110.PubMedCrossRef 7. Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, Pierotti MA, Daidone MG: Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 2005, 65:5506–5511.PubMedCrossRef 8. Sadlonova A, Novak Z, Johnson MR, Bowe DB, Gault SR, Page GP, Thottassery JV, Welch DR, Frost AR: Breast fibroblasts modulate selleck epithelial cell proliferation in three-dimensional in vitro co-culture. Breast Cancer Res 2005, 7:R46–59.PubMedCrossRef

9. Hu M, Yao J, Cai L, Bachman KE, van den Brule F, Velculescu V, Polyak K: Distinct epigenetic changes in the stromal cells of breast GSK1120212 cancers. Nat Genet 2005, 37:899–905.PubMedCrossRef 10. Allinen M, Beroukhim R, Cai L, Brennan C, Lahti-Domenici J, Huang H, Porter D, Hu M, Chin L, Richardson A, Schnitt S, Sellers WR, Polyak K: Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 2004, 6:17–32.PubMedCrossRef 11. Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A:

Involvement of chemokine receptors in breast cancer metastasis. Nature 2001, 410:50–56.PubMedCrossRef BVD-523 order 12. Salvucci O, Yao L, Villalba S, Sajewicz A, Pittaluga S, Tosato G: Regulation of endothelial cell branching morphogenesis by endogenous chemokine stromal-derived factor-1. Blood 2002, 99:2703–2711.PubMedCrossRef 13. Guo Y, Hangoc G, Bian H, Pelus LM, Broxmeyer HE: Sdf-1/cxcl12 enhances survival and chemotaxis of murine embryonic stem cells and production of primitive and definitive hematopoietic progenitor cells. Stem Cells 2005, 23:1324–1332.PubMedCrossRef 14. Chalasani SH, Sabol A, Xu H, Gyda MA, Rasband K, Granato M, Chien CB, Raper JA: Stromal cell-derived factor-1 antagonizes slit/robo signaling Florfenicol in vivo. J Neurosci 2007, 27:973–980.PubMedCrossRef 15. Chepko G, Slack R, Carbott D, Khan S, Steadman L, Dickson RB:

Differential alteration of stem and other cell populations in ducts and lobules of tgfalpha and c-myc transgenic mouse mammary epithelium. Tissue Cell 2005, 37:393–412.PubMedCrossRef 16. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, Oh EY, Gaber MW, Finklestein D, Allen M, Frank A, Bayazitov IT, Zakharenko SS, Gajjar A, Davidoff A, Gilbertson RJ: A perivascular niche for brain tumor stem cells. Cancer Cell 2007, 11:69–82.PubMedCrossRef 17. Huang MZ, Zhang FC, Zhang YY: [influence factors on the formation of mammospheres from breast cancer stem cells]. Beijing Da Xue Xue Bao 2008, 40:500–504.PubMed 18. Dontu G, Abdallah WM, Foley JM, Jackson KW, Clarke MF, Kawamura MJ, Wicha MS: In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells. Genes Dev 2003, 17:1253–1270.PubMedCrossRef 19. Beachy PA, Karhadkar SS, Berman DM: Tissue repair and stem cell renewal in carcinogenesis. Nature 2004, 432:324–331.PubMedCrossRef 20.

4 14 4 0 3 38 3 7 4 53 2 9 5 7 7 20 6 28 9 28 1 29 5 29 8 22 8 OR

This value corresponds approximately to the calculated number of mRNA IWR-1 manufacturer of this transcript per cell. b) buy Stattic standard deviation from the average mRNA, expressed in percentage. Table 3 Quantification of ICEclc core gene expression by dot-blot hybridization in strain B13 grown on different carbon TPCA-1 in vivo substrates.   Exponential phase After 24 h at stationary phase   3-chlorobenzoate succinate 3-chlorobenzoate succinate fructose glucose Probe number and probe mRNA a Std Dev b mRNA Std Dev mRNA Std Dev mRNA Std Dev mRNA Std Dev mRNA Std Dev     (%)   (%)   (%)   (%)   (%)   (%) 1) intB13 4.5 11.2 7.3 13.1 5.1 28.5 4.1 11.2 4.4 51.7 3.2 8.1 2) ORF52710 21.3 46.5 19.7 PRKACG 16.9 9.3 39.9 9.6 8 5.1 42.7 9.4 30.6 3) ORF53587 4.2 30.2 3.6 0.1 1.7 37.3 1.7 21.1 2 0.4 1.9 2.6 4) ORF59888 18.6 33 16.9 2.3 8.4 32.3 12.9 18.6 16.8 7.3 23.8 15.9 5) ORF65513 17.3 19.4 19.5

2.8 13.4 9.9 12.7 ‡ 5.3 13.8 7.6 13.8 11 6) ORF67800 16.6 2.7 16.6 5.5 8 12.9 12.7 18.3 11.6 33.7 17.9 38.6 7) ORF68987 2.1 4.3 2.1 11 0.8 12.9 0.8 0.2 1.3 13.8 1.1 11.7 8) ORF73029 2.5 20.8 2.9 12.6 2.6* 15 0.9 ‡ 18.2 1.4 6.7 1.1 4.2 9) ORF75419 7.5 18.1 7.3 6.8 11.1 32 3 ‡ 3.9 3.9 3.3 2.8 5.4 10) ORF81655 10.2 30.1 18.7 36.6 168* 24.5 6.3 2.7 45.7* 3.6 9.2 27 11) ORF83350 3.3 18.9 2.8 16.5 0.9 26.1 0.5 37.3 0.5 14.5 0.4 10.2 12) ORF84835 0.4 14.4 0.3 16.3 9.5* 7.7 0.3 25.8 1.7 16.1 0.3 1.5 13) ORF87986 5 1 5.2 0.1 64.5* 7.2 5.5 ‡ 0.4 14.2 26.9 5.5 2 14) ORF89746 12.9 34.1 24.4 19.8 2.2 41.2 2.1 17.3 2.1 36.7 0.5 15.4 15) ORF91884 3.3 11.7 4.5 3 3 32.4 1.6 ‡ 3.2 2.3 33.1 1.1 5.9 16) inrR 8.3 11.9 8.2 21.3 4.5 11.6 4 7.5 6.4 8.1 4.9 39.7 17) ORF96323 3 13 5.3 27.1 1 2.8 1.8 35.6 0.9 53.2 1.1 31.8 18) ORF98147 1.1 10.7 1.5 5.1 0.5 5.9 0.4 ‡ 7 0.4 3.7 0.4 1.7 19) ORF100033 30.6 4 40.4 20.2 12.3 16.7 17.6 18 22.9 6.4 22.2 30.2 20) ORF100952 1.4 13.2 2.2 22.2 1.8 § 3.1 0.9 1.7 1.9 7.9 0.9 29.4 21) ORF101284 3.7 18.9 3.2 6.9 1 23.1 1 ‡ 7.9 1.1 1.9 1.2 14.5 a) mRNA is the average calculated amount of mRNA copies × 108 per μg of total RNA from triplicate determinations.

Am J Clin Pathol 2001, 115:44–58 PubMedCrossRef 19 Krecicki T, Z

Am J Clin Pathol 2001, 115:44–58.selleck products PubMedCrossRef 19. Krecicki T, Zalesska-Krecicka M, Jelen M, Szkudlarek T, Horobiowska M: Expression of type IV collagen and matrix metalloproteinase-2 (type IV collagenase) in relation to nodal status in laryngeal cancer. Clin Otolaryngol Allied Sci 2001, 26:469–472.PubMedCrossRef 20. Santos-Garcia A, Abad-Hernandez

MM, Fonseca-Sanchez E, Julian-Gonzalez R, Galindo-Villardon P, Cruz-Hernandez JJ, Bullon-Sopelana A: E-cadherin, laminin and collagen IV expression in the evolution from dysplasia to oral squamous cell carcinoma. Med Oral Patol Oral Cir Bucal 2006, 11:E100-E105.PubMed 21. Bar JK, Grelewski P, Popiela A, Noga L, Rabczynski J: Type IV collagen and CD44v6 expression in benign, malignant primary and metastatic ovarian tumours: correlation with Ki-67 and p53 immunoreactivity. Doramapimod Gynecol Oncol 2004, 95:23–31.PubMedCrossRef 22. Ingber DE: Can cancer be reversed by engineering the tumour microenvironment? Semin Cancer Biol 2008, 18:356–364.PubMedCrossRef 23. Albini A, Sporn MB: The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer 2007, 7:139–147.PubMedCrossRef 24. Silzle T, Randolph

GJ, Kreutz M, Kunz-Schughart LA: The fibroblast: sentinel cell and local immune modulator in tumour tissue. Int J Cancer 2004, 108:173–180.PubMedCrossRef 25. Kalluri R, Zeisberg M: Fibroblasts in cancer. Nat Rev Cancer 2006, 6:392–401.PubMedCrossRef 26. Shimoda M, Mellody KT, Orimo TH-302 order A: Carcinoma-associated fibroblasts are a rate-limiting determinant

for tumour progression. Semin Cell Dev Biol 2010, 21:19–25.PubMedCrossRef 27. Qian BZ, Pollard JW: Macrophage diversity enhances tumour progression and metastasis. Cell 2010, 141:39–51.PubMedCrossRef 28. Mantovani A: La mala educacion of tumour-associated 4��8C macrophages: Diverse pathways and new players. Cancer Cell 2010, 17:111–112.PubMedCrossRef 29. Sobral LM, Bufalino A, Lopes MA, Graner E, Salo T, Coletta RD: Myofibroblasts in the stroma of oral cancer promote tumourigenesis via secretion of activin A. Oral Oncol 2011, 47:840–846.PubMedCrossRef 30. Kamat AA, Fletcher M, Gruman LM, Mueller P, Lopez A, Landen CN Jr, Han L, Gershenson DM, Sood AK: The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res 2006, 12:1707–1714.PubMedCrossRef 31. Ranogajec I, Jakic-Razumovic J, Puzovic V, Gabrilovac J: Prognostic value of matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9) and aminopeptidase N/CD13 in breast cancer patients. Med Oncol 2011, 29:561–569.PubMedCrossRef 32. Zhou CX, Gao Y, Johnson NW, Gao J: Immunoexpression of matrix metalloproteinase-2 and matrix metalloproteinase-9 in the metastasis of squamous cell carcinoma of the human tongue. Aust Dent J 2010, 55:385–389.PubMedCrossRef 33.

Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D: Global ca

Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D: Global cancer statistics. CA Cancer J Clin 2011, 61:69–90.PubMedCrossRef 2. Chiba T, Marusawa H,

Seno H, Watanabe N: Mechanism for gastric cancer development learn more by Helicobacter pylori infection. J Gastroenterol Hepatol 2008, 23:1175–1181.PubMedCrossRef 3. Meyer-ter-Vehn T, Covacci A, Kist M, Pahl HL: Helicobacter pylori activates mitogen-activated protein kinase cascades and induces expression of the proto-oncogenes c-fos and c-jun. J Biol Chem 2000, 275:16064–16072.PubMedCrossRef 4. Sohn SH, Lee YC: The genome-wide expression ACP-196 profile of gastric epithelial cells infected by naturally occurring cagA isogenic strains of Helicobacter pylori. Environ Toxicol Pharmacol 2011, 32:382–389.PubMedCrossRef 5. Calvino FM, Parra CT: H. pylori and mitochondrial ABT-737 mouse changes in epithelial cells. The role of oxidative stress. Rev Esp Enferm Dig 2010, 102:41–50. 6. Konturek PC, Konturek SJ, Brzozowski T: Helicobacter pylori infection in gastric cancerogenesis. J Physiol Pharmacol 2009, 60:3–21.PubMed 7. Pierzchalski P, Pytko-Polonczyk

J, Jaworek J, Konturek SJ, Gonciarz M: Only live Helicobacter pylori is capable of caspase-3 dependent apoptosis induction in gastric mucosa epithelial cells. J Physiol Pharmacol 2009, 60:119–128.PubMed 8. Matsumoto A, Isomoto H, Nakayama M, Hisatsune J, Nishi Y, Nakashima Y, et al.: Helicobacter pylori VacA reduces the cellular expression of STAT3 and pro-survival Bcl-2 family proteins, Bcl-2 and Bcl-XL, leading to apoptosis in gastric

epithelial cells. Dig Dis Sci 2011, 56:999–1006.PubMedCrossRef 9. Kusters JG, Van Vliet AH, Kuipers EJ: Pathogenesis of Helicobacter pylori infection. Clin Microbiol Rev 2006, 19:449–490.PubMedCrossRef 10. Blaser MJ, Atherton FER JC: Helicobacter pylori persistence: biology and disease. J Clin Invest 2004, 113:321–333.PubMed 11. Mimuro H, Suzuki T, Nagai S, Rieder G, Suzuki M, Nagai T, et al.: Helicobacter pylori dampens gut epithelial self-renewal by inhibiting apoptosis, a bacterial strategy to enhance colonization of the stomach. Cell Host Microbe 2007, 2:250–263.PubMedCrossRef 12. Buti L, Spooner E, Van der Veen AG, Rappuoli R, Covacci A, Ploegh HL: Helicobacter pylori cytotoxin-associated gene A (CagA) subverts the apoptosis-stimulating protein of p53 (ASPP2) tumor suppressor pathway of the host. Proc Natl Acad Sci USA 2011, 108:9238–9243.PubMedCrossRef 13. Tannaes T, Dekker N, Bukholm G, Bijlsma JJ, Appelmelk BJ: Phase variation in the Helicobacter pylori phospholipase A gene and its role in acid adaptation. Infect Immun 2001, 69:7334–7340.PubMedCrossRef 14. Bukholm G, Tannaes T, Nedenskov P, Esbensen Y, Grav HJ, Hovig T, et al.: Colony variation of Helicobacter pylori: pathogenic potential is correlated to cell wall lipid composition. Scand J Gastroenterol 1997, 32:445–454.PubMedCrossRef 15.

J Bacteriol 2006, 188:5731–5740 PubMedCrossRef 40 Payne SM, Mey

J Bacteriol 2006, 188:5731–5740.PubMedCrossRef 40. Payne SM, Mey AR: Pathogenic Escherichia coli , Shigella , and Salmonella . In Iron Transport in Bacteria. Edited by: Crosa JH, Mey AR, Payne SM. Washington, DC: American Society for Microbiology; 2004:199–218. 41. Diarra MS, Dolence JA, Dolence EK, Darwish I, Miller MJ, Malouin F, Jacques M: Growth of Actinobacillus pleuropneumoniae

is promoted by exogenous hydroxamate and catechol siderophores. Appl Environ Microbiol 1996, 62:853–859.PubMed 42. National Center for Biotechnology Information [http://​www.​ncbi.​nlm.​nih.​gov/​Genomes/​] 43. Wellcome Trust Sanger Institute [http://​www.​sanger.​ac.​uk] 44. Harrison LH, Simonsen V, Waldman EA: Emergence and disappearance of

a virulent clone of Haemophilus influenzae biogroup aegyptius, cause of Brazilian purpuric fever. Clin Microbiol DNA Damage inhibitor Rev 2008, 21:594–605.PubMedCrossRef 45. Musser JM, Barenkamp SJ, Granoff DM, Selander RK: Genetic relationships of serologically nontypable and serotype b strains of Haemophilus influenzae . Infect Immun 1986, 52:183–191.PubMed 46. Mikael LG, Srikumar R, Coulton JW, Jacques M: fhuA of Actinobacillus pleuropneumoniae encodes a ferrichrome receptor but is not regulated by iron. Infect Immun 2003, 71:2911–2915.PubMedCrossRef 47. Cope LD, Yogev R, Muller-Eberhard U, Hansen EJ: A gene cluster involved in the utilization of both free heme and heme:hemopexin by Haemophilus influenzae PRKD3 type b. J Bacteriol 1995, 177:2644–2653.PubMed SBI-0206965 manufacturer 48. Kidd SP, Jiang D, Jennings MP, McEwan AG: Glutathione-dependent alcohol dehydrogenase AdhC is required for defense against nitrosative stress in Haemophilus influenzae . Infect Immun 2007, 75:4506–4513.PubMedCrossRef 49. Whitby PW, VanWagoner TM, Seale TW, Morton DJ, Stull TL: Transcriptional profile

of Haemophilus influenzae : Effects of iron and heme. J Bacteriol 2006, 188:5640–5645.PubMedCrossRef 50. Whitby PW, Seale TW, VanWagoner TM, Morton DJ, Stull TL: The iron/heme regulated genes of Haemophilus influenzae : Comparative transcriptional profiling as a tool to define the species core modulon. BMC Genomics 2009, 10:6.PubMedCrossRef 51. Whitby PW, Sim KE, Morton DJ, Patel JA, Stull TL: Transcription of genes encoding iron and heme acquisition proteins of Haemophilus influenzae during acute otitis media. Infect Immun 1997, 65:4696–4700.PubMed 52. Speziali CD, Dale SE, Henderson JA, Vines ED, Heinrichs DE: Requirement of Staphylococcus aureus Belnacasan ic50 ATP-binding cassette-ATPase FhuC for iron-restricted growth and evidence that it functions with more than one iron transporter. J Bacteriol 2006, 188:2048–2055.PubMedCrossRef 53. Pramanik A, Braun V: Albomycin uptake via a ferric hydroxamate transport system of Streptococcus pneumoniae R6. J Bacteriol 2006, 188:3878–3886.PubMedCrossRef 54.

Such models allow independent testing of different experimental t

Such models allow independent testing of different experimental treatments on both gut microbiota

selleck inhibitor composition and metabolic activity within a single experimental period, using the same microbiota under controlled environmental conditions, which are designed to simulate the proximal, transverse and distal colon of healthy and infected subjects [9–14]. More recently, a three-stage in vitro colonic fermentation model of Salmonella infection in child colon was used to assess the effects of probiotic and prebiotic treatments on gut microbial behavior and on S. Typhimurium infection [15]. The activity of microcin B17-producing Escherichia coli L1000 wt [16] and bacteriocinogenic Bifidobacterium thermophilum RBL67, both exhibiting strong anti-Salmonella activity in simple in vitro tests [17, 18], as well as the microcin B17-negative mutant strain MccB17-, were tested in two three-stage models inoculated with the same fecal inoculum. When added to the colonic model, E. coli L1000 unexpectedly stimulated Salmonella growth in all reactors independently of the microcin B17-phenotype, partly due to a low colonization of the strain in the complex Selleckchem GF120918 intestinal environment. In contrast, thermophilicin RBL67-producing Bifidobacterium thermophilum RBL67 revealed

high competitiveness and colonized at high levels but did not reduce Salmonella counts, most likely a function of the presence of a very high Salmonella population in the in vitro model prior to probiotic addition. Tariquidar supplier Most data available on the mechanistic effects of probiotics on the host are derived from in vitro studies with

intestinal cells [19]. Such models have also been used to investigate bacterial interactions with the intestinal epithelium during enteric infection [20]. Salmonella Arachidonate 15-lipoxygenase pathogenesis, for example, has been studied in pure cultures using epithelial Caco-2 and HT-29 cell models [21, 22], both of which lack the ability to produce mucus. The mucus-secreting HT29-MTX cell line however, represents more accurate physiological conditions of the gastrointestinal tract for investigating pathogenic behavior during infection, as the presence of mucus has been shown to enhance pathogenicity of pathogens such as Campylobacter jejuni [23]. All interaction studies of pathogens and probiotics with intestinal cells have been performed with simple systems of either pure or mixed cultures. Microbe cell interactions are however different when tested in the presence of a complex gut microbiota [24, 25]. Gut metabolites such as SCFAs affect epithelial cell metabolism, turnover and apoptosis [26] but may also enhance virulence (e.g. S. Typhimurium), by inducing an acid tolerance response or increasing expression of porins [27]. To our knowledge, the effects of an infected gut microbiota, including its metabolites and probiotic treatment on intestinal cells has not been previously reported.

Chemistry-an Asian J 2010,5(10):2144–2153

Chemistry-an Asian J 2010,5(10):2144–2153.CrossRef 4. Sohn IY, Kim DJ, Jung JH:

Ja Yoon O, Thanh Tien N, Quang Trung T, Lee NE: pH Talazoparib in vivo sensing characteristics and biosensing application of solution-gated reduced graphene oxide field-effect transistors. Biosens Bioelectron 2013, 45:70–76.CrossRef 5. Kiani MJ, Ahmadi MT, Abadi HKF, Rahmani M, Hashim A: Analytical modelling of monolayer graphene-based ion-sensitive FET to pH changes. Nanoscale Res Lett 2013, 8:1–9.CrossRef 6. Dong X, Shi Y, Huang W, Chen P, Li L: Electrical detection of DNA hybridization with single base specificity using transistors based on CVD grown graphene sheets. Adv Mater 2010,22(14):1649–1653.CrossRef 7. Lee SJ, Youn BS, Park JW, Niazi JH, Kim YS: Gu MB: ssDNA aptamer-based surface plasmon resonance biosensor

for the detection of retinol binding protein 4 for the early diagnosis of type 2 diabetes. Anal Chem 2008,80(8):2867–2873.CrossRef 8. Liu AL, Zhong GX, Chen JY, Weng SH, Huang HN, Chen W, Lin LQ, Lei Y, Fu FH: Sun Zl: A sandwich-type DNA biosensor based on electrochemical VS-4718 molecular weight co-reduction synthesis of graphene-three dimensional nanostructure gold nanocomposite films. Anal Chimica Acta 2013, 767:50–8.CrossRef 9. Singh V, Joung D, Zhai L, Das S, Khondaker SI, Seal S: Graphene based materials: past, present and future. Prog Mater Sci 2011,56(8):1178–1271.CrossRef 10. Shao Y, Wang J, Wu H, Liu J, Aksay IA, Lin Y: Graphene based electrochemical sensors and biosensors: a review. Electroanal 2010,22(10):1027–1036.CrossRef 11. Zheng M, Jagota A, Semke ED, Diner BA, McLean RS, Lustig SR, Richardson RE, Tassi NG: DNA-assisted dispersion and separation of carbon nanotubes. Nat Mater 2003,2(5):338–342.CrossRef 12. Souteyrand E, Cloarec J, Martin J, Wilson C, Lawrence I, Mikkelsen S, Lawrence M: Direct detection of the hybridization of synthetic homo-oligomer DNA sequences by field effect. J Phys Chem B 1997,101(15):2980–2985.CrossRef 13. Fritz J, Cooper EB, Gaudet S, Sorger PK, Manalis SR: Electronic detection of DNA by its intrinsic molecular charge. Proc Nat Acad Sci 2002,99(22):14142–14146.CrossRef 14. Wei F, Sun B, Guo Y, Zhao XS: Monitoring DNA hybridization on

alkyl modified silicon surface through capacitance measurement. Biosens Bioelectron 2003,18(9):1157–1163.CrossRef 15. Abouzar MH, Poghossian A, Cherstvy AG, Pedraza AM, Ingebrandt S, Schoening MJ: Label-free electrical detection of DNA by Chlormezanone means of field-effect nanoplate capacitors: experiments and modeling. Physica Status Solidi a-Applications Mater Sci 2012,209(5):925–934.CrossRef 16. Kim DS, Jeong YT, Park HJ, Shin JK, Choi P, Lee JH, Lim G: An FET-type charge sensor for highly sensitive detection of DNA sequence. Biosens Bioelectron 2004, 20:69–74.CrossRef 17. Kim DS, Park HJ, Jung HM, Shin JK, Choi P, Lee JH, Lim G: Field effect transistor-based bimolecular sensor employing a Pt see more reference electrode for the detection of deoxyribonucleic acid sequence. Jpn J Appl Phys 2004,43(6B):3855–3859. [http://​jjap.​jsap.

XRD, TEM, Raman, and optical transmission techniques have been ut

XRD, TEM, Raman, and optical transmission techniques have been utilized to understand the microstructure characterization of nc-Si:H thin films. XPS results have confirmed that oxygen impurities on the surface of the nc-Si:H films have the dominant formation state of SiO2. The good agreement between the bonded hydrogen content and the volume fraction of grain boundary illustrates that as an important defect structure, the volume fraction of grain boundary in nc-Si:H films can be effectively regulated through hydrogen dilution. The inverse relationship between the RG-7388 integrated intensity of MSM and the oxygen content presents that the oxygen incursions due to

post-oxidation originate from the location of grain boundaries inside nc-Si:H films. The tuning mechanism of hydrogen on oxygen impurities Selleck MK5108 is that the hydrides corresponding Givinostat purchase to the MSM with a certain kind of bonding configuration are formed by the incorporation of H atoms and ions with the silicon dangling bonds located at grain boundaries, which can effectively prevent the oxygen incursions from residing along grain boundaries and further forming the Si-O/Si defects. Therefore, applying an extra negative bias on the substrate during the growth process is proposed

to reduce the probability of oxygen contamination, which can produce films with better light absorption properties in the solar cell application. Acknowledgements This work was supported by the National Major Basic Research Projects (2012CB934302) and Natural Science Foundation of China (11174202 and 61234005). References 1. Kitao J, Harada H, Yoshida NJ, Kitao H, Yoshidaa HN, Kasuya Y, Nishio M, Sakamoto T, Itoh T, Nonomura S, Nitta S: Absorption coefficient spectra of μc-Si in the low-energy region

0.4–1.2 eV. Sol Energy Mater Sol Cells 2001, 66:245–251.CrossRef 2. Zhang R, Chen XY, Zhang K, Shen WZ: Photocurrent response of hydrogenated nanocrystalline silicon thin films. J Appl Phys 2006, 100:104310–104315.CrossRef 3. Chen XY, Shen WZ, He YL: Enhancement of electron mobility in nanocrystalline silicon/crystalline silicon heterostructures. J Appl Phys 2005, 97:024305–5.CrossRef 4. Keppner H, Meier J, Torres P, Fischer D, Shah A: Microcrystalline silicon and micromorph tandem solar cells. Appl Phys A 1999, PAK6 69:169–177.CrossRef 5. Mai Y, Klein S, Geng X, Finger F: Structure adjustment during high-deposition-rate growth of microcrystalline silicon solar cells. Appl Phys Lett 2004, 85:2839–2841.CrossRef 6. Yang J, Yan B, Guha S: Amorphous and nanocrystalline silicon-based multi-junction solar cells. Thin Solid Films 2005, 487:162–169.CrossRef 7. Yamamoto K, Nakajima A, Yoshimi M, Sawada T, Fukuda S, Suezaki T, Ichikawa M, Koi Y, Goto M, Meguro T, Matsuda T, Kondo M, Sasaki T, Tawada Y: A thin-film silicon solar cell and module. Prog Photovolt Res Appl 2005, 13:489–494.

LV Shmeleva She made mathematical calculations, take part in the

LV Shmeleva. She made mathematical calculations, take part in the discussing of the results and conclusions. Both authors www.selleckchem.com/products/pf-03084014-pf-3084014.html read and approved the final manuscript.”
“Background ZnO semiconductor attracted considerable research attention in the last decades due to its excellent properties in a wide range of applications. ZnO is inherently an n-type semiconductor and has a wide bandgap of approximately 3.37 eV and a large exciton binding energy of approximately 60 meV at room temperature. As mentioned

above, ZnO is a promising semiconductor for various applications such as UV emitters and photodetectors, light-emitting diodes (LEDs), gas sensors, field-effect transistors, and solar cells [1–6]. Additionally, ZnO resists radiation, and hence, it is a suitable semiconductor for space technology applications. Recently, ZnO nanostructures have been used to produce short-wavelength optoelectronic devices due to their ideal optoelectronic, physical, and chemical properties that arise from a high surface-to-volume ratio and quantum confinement effect [6–8].

Among the ZnO nanostructures, ZnO nanorods showed excellent properties in different applications and acted as a main component for various nanodevices [1, 2, 9–11]. Stattic chemical structure Previous research showed that the optical and Vactosertib purchase structural properties of ZnO nanorods can be modified by doping with a suitable element to meet pre-determined needs [12, 13]. The most commonly investigated metallic dopants are Cu and Al [13–15]. Specifically, copper is known as a prominent luminescence activator, which can

enhance the green luminescence Y-27632 molecular weight band by creating localized states in the bandgap of ZnO [16–19]. Previous research showed that Cu has high ionization energy and low formation energy, which speedup the incorporation of Cu into the ZnO lattice [16, 20]. Experimentally, it was observed that the addition of Cu into ZnO-based systems has led to the appearance of two defective states at +0.45 eV (above the valence band maximum) and −0.17 eV (below the conduction band minimum) [21, 22]. Currently, a green emission band was observed for many Cu-doped ZnO nanostructures grown by different techniques [23, 24]. Moreover, Cu as a dopant gained more attention due to its room-temperature ferromagnetism, deep acceptor level, some similar properties to those of Zn, gas sensitivity, and enhanced green luminescence [15–17]. However, there are several points that have to be analyzed such as the effect of the copper source on the structural, morphological, and optical properties of Cu-doped ZnO. Moreover, the luminescence and the structural properties of Cu-doped ZnO nanorods are affected by different parameters such as growth conditions, growth mechanism, post growth treatments, and Cu concentration. Despite the promising properties, research on the influence of Cu precursors on Cu-doped ZnO nanorod properties remains low.

In sum, this work shows the value of DNA synthesis and standardiz

In sum, this work shows the value of DNA synthesis and standardization of functional modules for combining in a single genetic tool many valuable properties that are otherwise scattered in various vectors and rendered useless for the lack of fixed assembly formats. We anticipate pBAM1 to become one frame of reference

for the construction of a large number of vectors aimed at deployment of heavily engineered genetic and metabolic circuits. Methods Strains, plasmids and media The bacterial strains and plasmids used in this study are listed in Table 3. Bacteria were grown routinely in LB (10 g l-1 of tryptone, 5 g l-1 of yeast extract and 5 g l-1 of NaCl). E. coli cells were grown at 37°C while P. putida AZD4547 in vivo was cultured at 30°C. Selection of P. putida cells was made onto M9 selleck products minimal medium plates [55] CT99021 molecular weight with citrate (2 g l-1) as the

sole carbon source. Antibiotics, when needed, were added at the following final concentration: ampicillin (Ap) 150 μg ml-1 for E. coli and 500 μg ml-1 for P. putida, kanamycin (Km) 50 μg ml-1 and chloramphenicol (Cm) 30 μg ml-1 for both species. 5-bromo-4-chloro-3-indolyl- β-D-galactopyranoside (Xgal) was added when required at 40 μg ml-1. The Pu-lacZ fusion of P. putida MAD1 (Table 3) was induced by exposing cells to saturating m-xylene vapors. DNA techniques Standard procedures were employed for manipulation of DNA [55]. Plasmid DNA was prepared using Wizard Plus SV Minipreps (Promega) and PCR-amplified DNA purified with NucleoSpin Extract II (MN). Oligonucleotides were purchased CHIR-99021 chemical structure from SIGMA. For colony PCR a fresh single colony was picked from a plate and transferred directly into the PCR reaction tube. Transposon insertions were localized by arbitrary PCR of genomic DNA

[33]. Single colonies were used as the source of the DNA template for the first PCR round, which was programmed as follows: 5 minutes at 95°C, 6 cycles of 30 s at 95°C, 30 sec at 30°C, and 1 min and 30 s at 72°C; 30 cycles of 30 s at 95°C, 30 s at 30°C and 1 min and 30 s at 72°C. This was followed by an extra extension period of 4 min at 72°C. The primers used for the first round included ARB6 in combination with either ME-O-extF or ME-I-extR/GFP-extR (described in Table 2). 1 μl of the resulting product was then used as template for the second PCR round, using with the following conditions: 1 min at 95°C, 30 cycles of 30 s at 95°C, 30 sec at 52°C and 1 min and 30 sec at 72°C, followed by an extra extension period of 4 min at 72°C. The second round was performed with ARB2 and ME-O-intF or ME-I-intR/GFP-intR (Table 2). PCR reaction mixtures were purified and sequenced with either ME-O-intF or ME-I-intR/GFP-intR primers. DNA sequences were visually inspected for errors and analyzed using the Pseudomonas Genome Databasev2 (http://​www.​pseudomonas.​com) and blast (http://​blast.​ncbi.​nlm.​nih.​gov/​Blast.​cgi) to map the precise transposon insertion point.