Bethe and colleagues reported that PrtA is a highly conserved vir

Bethe and colleagues reported that PrtA is a highly conserved virulence factor of Streptococcus pneumoniae, and might be a promising candidate for a protein-based

vaccine [21]. (ii) Autolysin, the autolysin encoded by cwh is also a reported virulence-associated factor in SS2 [22]. Most bacteria possess several autolysins that are able to degrade their cell walls, and are implicated in various biological functions Ilomastat including cell separation, cell wall turnover, restructuring of cell walls, and bacterial autolysis. In addition, certain autolysins have also been reported to contribute to the pathogenicity of gram-positive bacteria. For example, an intact autolytic function is required for the full virulence of Streptococcus pneumoniae [23]. (iii) protein TRAG, TRAG is a component of the type IV secretion system (T4SS), a virulence-associated pathway of SS2 [22]. The bacterial T4SS, which is widely distributed among the gram-negative and -positive bacteria and is ancestrally related to bacterial conjugation machines (which mediate protein and gene transfer), contributes to pathogenicity [24]. Analysis of the in vivo gene expression profiles Strain ZY05719 was selected for real-time PCR analysis because it is one of the strains isolated from the 2005 SS2 outbreak in China; ZY05719 was also used for constructing the genomic library. Of the 48 putative

IVI genes, 10 (ss-1616, trag, nlpa, srt, cwh, hprk, ysirk, ss-1955, Belnacasan cost sdh, ss-1298) were selected for further analysis of gene expression by real-time PCR. We selected these genes based on their putative functions, such as involvement in cell structure, metabolism, regulation, and transport, in order to maximize the variety of genes

chosen for further analysis. The in vitro expression of these 10 putative IVI genes was observed in early lag phase, log phase, late log phase, and check details stationary phase of growth, with the highest level of expression occurring at late log phase (data not shown). Before comparing the expression of these 10 putative IVI genes under the in vitro condition, they were first tested under in vivo conditions (expression after challenge with bacterial cells via intravenous inoculation measured at 12, 24, and 36 h pi). All of the putative IVI genes were expressed in vivo under the conditions tested (data not shown). With the exception of ysirk and ss-1955, which were expressed at 12 h pi but not at 24 and 36 h pi, and ss-1298, which was expressed until 36 h, the remaining 7 IVI genes were expressed at 12, 24 and 36 h post-inoculation in vivo. The aim of this study was to identify the genes whose expressions are upregulated in vivo; therefore, we determined the in vivo gene expression relative to the highest level of expression in vitro.

The analysis revealed that most differences in protein expression

The analysis revealed that most differences in protein expression patterns were genetically encoded rather than induced by antibiotic exposure. Over-expression of stress proteins

was expected, as they represent a common non-specific Ro 61-8048 response by bacteria when stimulated by different shock conditions. Positive transcription regulators were found to be over-expressed in rifampicin resistance, suggesting that bacteria could activate compensatory mechanisms to assist the transcription process in the presence of RNA polymerase inhibitors. Other differences in expression profiles were related to proteins involved in central metabolism; these modifications suggest metabolic disadvantages of resistant mutants compared to sensitive ones. Of particular interest are the proteins involved in the cell division site. The altered proteins can affect the integrity of the Z ring at various stages. In the same way, it was hypothesized that the Z ring assembly could be both coordinated with the cell cycle and rendered responsive to cellular and environmental stresses. The analysis of the protein differentially expressed may suggest the intricate series of events occurring in these strains. In this light, the growth results may be partially explained by a decrease Selleckchem MM-102 expression of proteins such as

the cell division protein and the septum site-determining protein MinD. Conclusions Our findings reveal that we need a deeper understanding of the interplay between antibiotic resistance, biological fitness and virulence. Although our results are not sufficient to establish an unequivocal association between the differential protein expression and the resistant phenotype, they may be considered a starting point in understanding the decreased invasion capacity of N. meningitidis rifampicin resistant strains. In fact, they support the hypothesis that the presence of more than one protein differentially expressed, having a role in the metabolism, influences

Protein kinase N1 the ability to infect and to spread in the population. Different reports have described and discussed how a drug resistant pathogen shows a high biological cost for survival [24, 25] and that may also explain why, for some pathogens, the rate of resistant organisms is relatively low considering the widespread use of a particular drug. This seems the case of rifampicin resistant meningococci. Only the combination gained from different experimental methods and clinical data reporting will enable to model the adaptation response of such strains in their physiological network. Our aim was to improve knowledge of the microbial physiology of resistant meningococci and understand why, despite widespread use of rifampicin in prophylactic treatment, the resistant isolates continue to be so rare. Ethical approval Not required.

5 (Biometris, The Nerherlands) Results Geochemical properties in

5 (Biometris, The Nerherlands). Results Geochemical properties in sampling sites Soil characteristics of these six sampling P505-15 price sites are summarized in Table 1. pH in all those sites was neutral or close to alkali, and they were rich in organic carbon (C) and nitrogen (N), ranging from 91.99 g/kg to 209.19 g/kg and 1.50 g/kg to 15.50 g/kg, respectively. It was noted that C/N ratios displayed a decreasing trend as the elevation increased. For example, sample SJY-GH with the lowest elevation (3400 m) had the highest value of C/N ratio, whereas

sample SJY-YS with the highest elevation (4813 m) had the lowest C/N ratio. In addition, sample SJY-GH had higher total C, N, P and K contents buy GF120918 than the other samples. Overview of functional gene diversity and structure of soil microbial

communities The examined microbial communities showed high diversity, as judged by the number of detected genes, overlapping genes between samples, unique genes and diversity indices (Table 2). The total number of detected genes ranged from 1,732 to 3,746 among the six study sites (Table 2). For instance, twice as many genes were detected in sample SJY-GH as in sample SJY-CD, SJY-ZD or SJY-YS. These samples had different community compositions, as shown by the unique and overlapped genes (Table 2). Sample SJY-GH and sample SJY-DR had the most overlapped genes (2029, 42.94%), while sample SJY-GH and sample SJY-YS had the fewest overlapped genes (1178, 27.22%). Simpson’s reciprocal diversity index (1/D) was the highest in sample SJY-GH

and the lowest many in sample SJY-CD (3716 and 1723, respectively). Similar results were obtained with Shannon-Weaver index (Table 2). Table 2 Total detected gene number, gene overlap, unique, diversity indices of soil sample a Unique and overlap genes SJY-GH SJY-DR SJY-QML SJY-CD SJY-ZD SJY-YS SJY-GH 1044(27.87%) 2029(42.94%) 1655(37.26%) 1264(30.00%) 1261(29.84%) 1178(27.22%) SJY-DR   617(20.51%) 1485(38.33%) 1171(32.81%) 1163(32.43%) 1107(30.24%) SJY-QML     403(17.14%) 1049(34.57%) 1062(35.05%) 973(31.01%) SJY-CD       242(13.97%) 916(35.82%) 840(31.67%) SJY-ZD         248(14.24%) 816(30.39%) SJY-YS           321(18.24%) Total no. of genes detected 3746 3008 2351 1732 1741 1760 Shannon weaver index 8.22 8.01 7.76 7.45 7.46 7.47 Simpson’s reciprocal diversity index (1/D) 3716 2988 2340 1723 1733 1752 a Values in parentheses are percentages. Italicized values indicate the number of overlapping genes between samples, boldface values indicate the number of unique genes in each sample. According to the phylogenetic analysis, the Proteobacteria group is the most dominant bacteria in all six samples, which account for over 56% (over 23% belong to α-proteobacteria, 13% belong to β-proteobacteria, 14% belong to γ-portecobacteria) among all the detected genes (Additional file 1: Table S1). The Actinobacteria (over 9.

5 months Conclusions FOLFIRI appears an effective and safe treat

5 months. Conclusions FOLFIRI appears an effective and safe treatment option for pretreated metastatic gastric cancer patients. However, second-line chemotherapy comparative trials are needed to better define the role of FOLFIRI in gastric cancer (e.g. versus monochemotherapy). Acknowledgements We thank Tania Merlino for technical assistance. References 1. 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. Cervantes A, Roda D, Tarazona N, Roselló

S, Pérez-Fidalgo JA: Current questions for the treatment of advanced gastric cancer. Cancer Treat Rev 2013, 39:60–67.PubMedCrossRef 3. Glimelius B, Ekström K, Hoffman K, Graf W, Sjödén PO, Haglund U, Svensson C, Enander LK, Linné T, Sellström H, Heuman R: Randomized comparison between LY3039478 cell line chemotherapy plus best supportive care with best supportive care in advanced gastric cancer. Ann Oncol 1997, 8:163–168.PubMedCrossRef 4. Murad AM, Santiago FF, Petroianu A, Rocha PR, Rodrigues MA, Rausch M: Modified therapy with 5-fluorouracil,

doxorubicin, and methotrexate in advanced gastric cancer. Cancer 1993, 72:37–41.PubMedCrossRef 5. Pyrhönen S, Kuitunen T, Nyandoto P, Kouri M: Randomised comparison of fluorouracil, epidoxorubicin and methotrexate (FEMTX) plus supportive care with supportive care learn more alone in patients with non-resectable gastric cancer. Br J Cancer 1995, 71:587–591.PubMedCrossRef 6. Van Cutsem E, Moiseyenko VM, Tjulandin S, Majlis A, Constenla M, Boni C, Rodrigues A, Fodor M, Chao Y, Voznyi E, Risse ML, Ajani JA: V325 Study Group. Phase III study of docetaxel and cisplatin plus fluorouracil compared with cisplatin and fluorouracil as first-line therapy for advanced gastric cancer: a report of the V325 Study Group. Reverse transcriptase J Clin Oncol 2006, 24:4991–4997.PubMedCrossRef 7. Koizumi W, Narahara H, Hara T, Takagane A, Akiya T, Takagi M, Miyashita K, Nishizaki T, Kobayashi

O, Takiyama W, Toh Y, Nagaie T, Takagi S, Yamamura Y, Yanaoka K, Orita H, Takeuchi M: S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol 2008, 9:215–221.PubMedCrossRef 8. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T, Aprile G, Kulikov E, Hill J, Lehle M, Rüschoff J, Kang YK, ToGA Trial Investigators: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010, 376:687–697.PubMedCrossRef 9.

2 +++ 100 0 +++ 52 7   5 +++ 100 0 +++ 78 7 +++ 100 0 +++ 100 0 T

2 +++ 100.0 +++ 52.7   5 +++ 100.0 +++ 78.7 +++ 100.0 +++ 100.0 Tylosin 80 +++ 100.0 +++ 100.0 +++ 100.0 +++ 79.4   40 +++ 100.0 +++ 100.0 +++ 100.0 +++ 92.2   5 +++ 100.0 +++ 94.5 +++ 100.0 +++ 100.0 Note: LIC-S2 and SIC-S2 mean inoculum from the first sub-culture of the large intestinal digesta or small intestinal digesta, respectively. + means slight growth; ++ moderate growth; +++ vigorous growth Figure 2 Flow chart showing the

process of selection for chicken intestinal bacteria with the ability to transform DON . *Selection criteria used in each step of the selection. Numbers in the parentheses indicate particular steps in the selection. The previously Alpelisib order selected cultures were diluted 10-fold in series, inoculated in the AIM+CecExt medium, incubated for 72 hr, and then examined for DON-transforming activity (Step 4 in Fig. 2). Among the serially diluted cultures (from 10-1 to 10-5), the diluted cultures in 10-1, 10-2, or 10-3

all completely transformed DON to DOM-1 in the medium. However, the diluted cultures in 10-4 and 10-5 demonstrated a partial activity of DON transformation with 44 and 24% of DON transformed to DOM-1, respectively. The process was repeated until the cultures had their cell density reduced Apoptosis inhibitor to 103 CFU ml-1, but still retained full activity of DON transformation prior to single colony isolation on L10 agar. Sixty eight and 128 single colonies were isolated from the diluted SIC and LIC cultures, respectively, and ten isolates (representing approximately 5% of the colonies examined) were found to be capable of transforming DON to DOM-1 (Fig. 3). One of the isolates was from the small intestine and the remaining from the large intestine. Figure 3 LC-MS chromatograms showing the biotransformation

of DON to DOM-1 . A) DON (100 μg ml-1) in L10 broth without any bacterial inoculum after 72 hr incubation. Selected ion monitoring at m/z 231, 249, 267, 279, and 297. B) Transformation of DON (100 μg ml-1) to DOM-1 in L10 broth inoculated with isolate LS100 after 72 hr incubation. Selected ion monitoring at m/z 215, 233, 245, 251, 263, and 281. PCR-DGGE bacterial profiles were used to guide the selection for DON-transforming bacteria in this study. Fig. 4 displays examples to show the effectiveness of PCR-DGGE bacterial profiles in guiding the bacterial selection. The large intestinal digesta sample (Panel A – Lane Janus kinase (JAK) 1) had many more DNA bands than the start culture (Lane 2) that was a subculture from the digesta, indicating the selective effect of subculturing. It was described above that tylosin had no detrimental effect on either DON transformation or bacterial growth of the start cultures at all tested concentrations. However, the treatment showed little influence over the richness of bacterial populations, as indicated by the similarity of PCR-DGGE bacterial profiles before and after tylosin treatment (Panel A – Lanes 2, 5, and 6). Thus no further experiments were pursued with the resulting cultures.

Biol Chem 2006, 387:1175–1187 PubMedCrossRef 8 Fritz WA, Lin TM,

Biol Chem 2006, 387:1175–1187.PubMedCrossRef 8. Fritz WA, Lin TM, Safe S, Moore RW, Peterson RE: The selective aryl hydrocarbon receptor modulator 6-methyl-1,3,8-trichlorodibenzofuran inhibits prostate tumor metastasis in TRAMP mice. Biochem Pharmacol 2009, 77:1151–1160.PubMedCrossRef 9. Peng TL, Chen J, Mao W, Liu X, Tao Y, Chen LZ, Chen MH: Potential therapeutic

significance of increased expression of aryl hydrocarbon receptor in human gastric cancer. World J Gastroenterol 2009, 15:1719–1729.PubMedCrossRef 10. Barouki R, Coumoul X, Fernandez-Salguero PM: The aryl hydrocarbon receptor, more than a xenobiotic-interacting protein. FEBS Lett 2007, 581:3608–3615.PubMedCrossRef 11. Cole P, Trichopoulos D, Pastides H, Starr T, Mandel JS: Dioxin and cancer: a critical review. Regul Toxicol Pharmacol 2003, 3-MA purchase 38:378–388.PubMedCrossRef 12. Bradfield CA, Bjeldanes LF: Structure-activity relationships of dietary indoles: a proposed mechanism of action as modifiers of xenobiotic metabolism. J Toxicol Environ Health 1987, 21:311–323.PubMedCrossRef 13. Chen I, Safe S, Bjeldanes L: Indole-3-carbinol and diindolylmethane as aryl hydrocarbon www.selleckchem.com/products/blu-285.html (Ah) receptor agonists and antagonists in T47D human breast cancer

cells. Biochem Pharmacol 1996, 51:1069–1076.PubMedCrossRef 14. Kim EJ, Park SY, Shin HK, Kwon DY, Surh YJ, Park JH: Activation of caspase-8 contributes to 3,3′-Diindolylmethane-induced apoptosis in colon cancer cells. J Nutr 2007, 137:31–36.PubMed 15. Koliopanos A, Kleeff J, Xiao Y, Safe S, Zimmermann A,

Büchler MW, Friess H: Increased aryl hydrocarbon receptor expression offers a potential therapeutic target in pancreatic cancer. Oncogene 2002, 21:6059–6070.PubMedCrossRef 16. Ciolino HP, Daschner PJ, Yeh GC: Resveratrol inhibits transcription of CYP1A1 in vitro by Ketotifen preventing activation of the aryl hydrocarbon receptor. Cancer Res 1998, 58:5707–5712.PubMed 17. Revel A, Raanani H, Younglai E, Xu J, Rogers I, Han R, Savouret JF, Casper RF: Resveratrol, a natural aryl hydrocarbon receptor antagonist, protects lung from DNA damage and apoptosis caused by benzo[a]pyrene. J Appl Toxicol 2003, 23:255–261.PubMedCrossRef 18. Mandal PK: Dioxin: a review of its environmental effects and its aryl hydrocarbon receptor biology. J Comp Physiol B 2005, 175:221–230.PubMedCrossRef 19. Safe S, McDougal A: Mechanism of action and development of selective aryl hydrocarbon receptor modulators for treatment of hormone-dependent cancers (Review). Int J Oncol 2002, 20:1123–1128.PubMed 20. Sugihara K, Okayama T, Kitamura S, Yamashita K, Yasuda M, Miyairi S, Minobe Y, Ohta S: Comparative study of aryl hydrocarbon receptor ligand activities of six chemicals in vitro and in vivo. Arch Toxicol 2008, 82:5–11.PubMedCrossRef 21. Chen I, McDougal A, Wang F, Safe S: Aryl hydrocarbon receptor-mediated antiestrogenic and antitumorigenic activity of diindolylmethane.

Biochim Biophys Acta 1972, 261:284–289 PubMed 39 Tsai CM, Frasch

Biochim Biophys Acta 1972, 261:284–289.PubMed 39. Tsai CM, Frasch CE: A sensitive silver stain for detecting lipopolysaccharides in polyacrylamide gels. Anal Biochem 1982, 119:115–119.PubMedCrossRef Competing interests The authors declare that they have no competing interests. Authors’ contributions LP has given an important contribution

to the elaboration of paper. CdL, SB, AL, LODL and MRC gave important contributions in the order to design of the paper and to draft of manuscript. GG and AlL have cooperated for technical assistance. GDR and MM have studied histopathology features. FR and LR conceived the study participating to its scientific design. C188-9 supplier All authors read and approved the final manuscript.”
“Background Mycoplasma synoviae is

an economically important pathogen of poultry, causing synovitis, chronic respiratory tract disease, and retarded growth in chickens and turkeys [1, 2]. M. synoviae is a member of the genus Mycoplasma of the class Mollicutes, a group of wall-less Gram-positive bacteria with genomes ranging from 1358 kb to as little as 580 kb [3]. The genome sequence of M. synoviae strain WVU 1853 has been determined and comparative analysis with M. gallisepticum, another major avian pathogen, provided evidence 17DMAG for horizontal gene transfer between the two species, though belonging to two distinct phylogenetic groups [4, 5]. Among the genes that could have arisen by horizontal gene transfer are those encoding for haemagglutinins. In avian mycoplasmas, genes encoding for these immunogenic and surface exposed proteins are the subject of considerable antigenic variability [6]. By alternating the composition of their surface proteins, mycoplasmas are thought to colonize more efficiently mucosal surfaces and become more virulent [7,

8]. Haemagglutinins account among the most important surface proteins involved in Wilson disease protein colonization and virulence of avian mycoplasmas [6, 9]. In M. synoviae, haemagglutinins are encoded by related sequences of a multigene family referred to as vlhA genes [10–12]. The haemagglutinins of M. gallisepticum (pMGA) and M. imitans are also encoded by multigene families related to vlhA [13, 14]. Both organization and control of expression of vlhA genes are quite different between M. gallisepticum and M. synoviae. In the former species, vlhA genes are located in five distinct genomic regions and each gene appears to be translationally competent [14, 15]. By contrast, in M. synoviae, all vlhA sequences are confined to a restricted genomic region with a unique copy being expressed in a single strain [16, 17] The uniquely expressed vlhA gene of M. synoviae yields a product that is cleaved post-translationally into a N-terminal lipoprotein (MSPB) and a C-terminal haemagglutinin protein (MSPA) [11]. Cleavage was found to occur immediately after amino acid residue 344 [17].

This temperature is commonly used for culture of S agalactiae fr

This temperature is commonly used for culture of S. agalactiae from fish [26]. Isolates were checked for Gram reaction and morphology and tested in a group B-specific latex agglutination test (Slidex Strepto Plus B; bioMérieux, Marcy L’Étoile, France). Single colonies were transferred to Brain Heart Infusion (BHI) broth (Oxoid, Basingstoke, United Kingdom) and incubated with gentle shaking at 28°C for 12h (ß-haemolytic strains, fast growing) or 48h (non-haemolytic strains, slow growing). AZD1390 Species identity of S. agalactiae was confirmed by polymerase chain reaction (PCR), using forward primer STRA-AgI (5′-AAGGAAACCTGCCATTTG-′3) and reverse primer STRA-AgII (5′-TTAACCTAGTTTCTTTAAAACTAGAA-3′),

which target the 16S to 23S rRNA intergenic spacer region [27]. Broth cultures were also used for PFGE as described below. Comparative typing: PFGE Bacterial cells were pelleted by centrifugation of 1 ml of incubated BHI, re-suspended in

0.5 ml of TE buffer (10 mM Tris-HCl, 1mM EDTA), warmed to 56°C and mixed with 0.5 ml of 2% (weight/vol) low-melting point agarose (Incert agarose; Lonza, Slough, United Kingdom) in TE buffer. The mixture was then pipetted into reusable plug moulds (Catalogue number 170-3622; BioRad Laboratories, Hemel Hempstead, United Kingdom) producing 20 × 9 × 1.2 mm3 agarose blocks. Each solidified plug was placed into 2 ml of TE buffer containing 4 mg of lysozyme (Sigma Aldrich, Poole, United Kingdom) (2 mg ml-1) and incubated overnight at 37°C with gentle shaking. The buffer was then Lumacaftor mw replaced with 2 ml of ES buffer (0.5 M EDTA–1% BMN 673 solubility dmso (weight/vol) N-lauroyl sarcosine [pH 8.0 to 9.3]) supplemented with 4 mg of proteinase K (Promega,

Southampton, United Kingdom) (2 mg ml-1) and incubated at 56°C for a minimum of 48 hr. Plugs were washed 6 times for 1 hr in TE buffer at room temperature and with gentle shaking. A slice (4 × 4 × 1.2 mm3) from each plug was exposed to digestion with restriction endonuclease SmaI (20 U in 100 μl of fresh reaction buffer; New England Biolabs, Hitchin, United Kingdom) at 25°C overnight. PFGE was performed with a CHEF-mapper system (BioRad Laboratories) in 0.5 × TBE using a 1% (weight/vol) agarose gel (Pulsed Field Certified Agarose, BioRad Laboratories), a run time of 24 hr and switch time of 3-55 s (linear ramp) at 14°C. Patterns were observed by UV transillumination after SYBR Gold staining (Invitrogen, Paisley, United Kingdom). Computer-assisted data analysis and dendogram construction were performed with Phoretix 1D Pro software (TotalLab Ltd, Newcastle upon Tyne, United Kingdom). Similarities between PFGE patterns were also assessed visually using standard criteria [10]. Housekeeping genes: multilocus sequence typing MLST consisted of the amplification by PCR and sequencing of seven housekeeping genes, namely adhP, atr, glcK, glnA, pheS, sdhA, and tkt[13].

In mammalian cells, PLK-1 is primarily localized in the centrosom

In mammalian cells, PLK-1 is primarily localized in the centrosome, where it is responsible for centrosome separation and maturation. PLK-1-specific antibodies introduced into HeLa cells by microinjection prevent centrosome separation and reduce γ-tubulin accumulation, suggesting that PLK-1 functions

selleck in regulating centrosome function [8]. PLK-1 is also a target of the G2 DNA damage checkpoint, where it undergoes ubiquitin-dependent proteolysis mediated by the checkpoint protein Chfr, implicating the loss of Plk-1 function as an important response to DNA damage during the G2 phase of the cell cycle [9]. Correspondingly, the elevation of PLK-1 expression occurs in a broad range of human tumors [10, 11], and a close correlation has been documented between mammalian PLK-1 expression and progression of endometrial and ovarian cancers [12, 13]. Therefore, PLK-1 is implicated as a critical candidate target for understanding find more the progression of cervical carcinoma and improving chemotherapy. However, little is known about the importance of PLK-1 in the development and management of cervical carcinoma. To address this issue, we investigated the expression and distribution of PLK-1 in cervical carcinoma tissues. Furthermore, in order to determine the importance of PLK-1 in tumor progression, we investigated the effects of PLK-1 knockdown on the biological characteristics of HeLa

cells by taking advantage of small interference RNA (siRNA) against PLK-1. Our results elucidate the pathogenesis of cervical carcinoma and may help to develop a novel strategy to improve the efficiency of chemotherapy delivered to patients with cervical carcinoma. Materials and methods Immunohistochemical staining

For immunohistochemical staining, thirty-six surgically resected human cervical carcinoma tissue samples were collected from the Department BCKDHB of Obstetrics and Gynecology, Wuhan Union Hospital. The study was approved by the institutional review boards. Immunohistochemical staining was performed according to our previous protocol [14]. Briefly, human tumor tissues were embedded in paraffin and cut into 5-μm sections that were placed onto glass slides. After antigen retrieval, sections were stained for the expression of PLK-1 (BD Biosciences, San Diego, CA) (1:100)detected by streptavidin-biotin-horseradish peroxidase complex formation. Tumor sections stained for IgG instead of primary antibodies were used as the negative control. The immunoactivities of PLK-1 were ranked according to the percentage of positive tumor cells: score 3 (> 75%), score 2 (25-75%), score 1 (< 25%), and score 0 (negative). Cell culture, transient transfection, RNA interference, and cisplatin treatment HeLa cells were cultured in RPMI 1640 supplemented with 10% fetal calf serum (FCS) (Invitrogen, Carlsbad, CA,). Plasmid construction and transfection were performed as previously described [4]. Briefly, PLK-1 cDNA was cloned into the pcDNA3.

J Biogeogr 36:2165–2175CrossRef Gullison RE, Frumhoff PC, Canadel

J Biogeogr 36:2165–2175CrossRef Gullison RE, Frumhoff PC, Canadell JG, Field CB, Nepstad DC, Hayhoe K, Avissar R, Curran LM, Friedlingstein P, Jones CD, Nobre C (2007) Tropical forests and climate policy. Science 316:985–986CrossRefPubMed Hedges S, Tyson MJ, Sitompul AF, Kinnaird MF, Gunaryadi D, Aslan (2005) Distribution, status, and conservation

needs of Asian elephants (Elephas maximus) in Lampung Province, Sumatra, Indonesia. Biol Conserv 124:35–48CrossRef Jepson P, Jarvie JK, MacKinnon K, Monk KA (2001) The end for Indonesia’s lowland forests? Science 292:859–861CrossRefPubMed Kanninen M, Murdiyarso D, Seymour F, Angelsen A, Wunder S, German L (2007) Do trees grow on money?: the implications of deforestation research for policies to promote REDD. For Perspect 4:61 Kinnaird MF, Sanderson EW, O’Brien TG, Wibisono HT, Woolmer G (2003) Deforestation trends in a tropical landscape RO4929097 ic50 and implications for endangered large mammals. Conserv Biol 17:245–257CrossRef Laumonier Y, Uryu Y, Stüwe M, Budiman A, Setiabudi B, Hadian O (submitted) Eco-floristic sectors and deforestation threats in Sumatra: buy C188-9 identifying new conservation area network priorities for ecosystem-based land use planning. Biodivers Conserv Laurance WF, Albernaz AKM, Schroth G, Fearnside

PM, Bergen S, Venticinque EM, da Costa C (2002) Predictors of deforestation in the Brazilian Amazon. J Biogeogr 29:737–748CrossRef Laurance WF, Goosem M, Laurance SGW (2009) Impacts of roads and linear clearings on tropical forests. Trends Ecol Evol 24:659–669CrossRefPubMed Leader-Williams N, Albon SD (1988) Allocation of resources

for conservation. Nature 336:533–535CrossRef Leader-Williams N, Albon SD, Berry PSM (1990) Illegal exploitation of black rhinoceros and elephant populations—patterns of decline, law-enforcement and patrol effort in Luangwa Valley, Zambia. J Appl Ecol 27:1055–1087CrossRef Linkie M, Smith RJ (2009) Measuring the effectiveness of conservation spending. In: Sodhi N, Ehrlich PR (eds) Conservation biology for all. Oxford University Adenosine Press, Oxford Linkie M, Smith RJ, Leader-Williams N (2004) Mapping and predicting deforestation patterns in the lowlands of Sumatra. Biodivers Conserv 13:1809–1818CrossRef Linkie M, Chapron G, Martyr DJ, Holden J, Leader-Williams N (2006) Assessing the viability of tiger subpopulations in a fragmented landscape. J Appl Ecol 43:576–586CrossRef Linkie M, Smith RJ, Zhu Y, Martyr DJ, Suedmeyer E, Pramono J, Leader-Williams N (2008) Evaluating biodiversity conservation around a large Sumatran protected area. Conserv Biol 22:683–690CrossRefPubMed Liu J, Linderman M, Ouyang Z, An L, Yang J, Zhang H (2001) Ecological degradation in protected areas: the case of Wolong Nature Reserve for giant pandas.