Table 1 Plaque morphology upon infection with λcIII 67 Genotype o

Table 1 Plaque morphology upon infection with λcIII 67 Genotype of host E. coli cell Plaque morphology Wild Type Clear Wild Type + pQKC Turbid AK990 (ΔhflKC::Kan) Turbid Is it then possible that enhancement of lysogeny can occur through a different mechanism that does not involve the stabilization of CII? Increase in lambda lysogeny is invariably

linked to the stability of CII in all published reports to date. Can the two phenomena be delinked in some special case such as a ΔhflKC host? We tested this possibility by measuring the stability of cloned CII in wild type and ΔhflKC cells, both infected with λcIII 67 . A greater stabilization of CII BIBW2992 occurred in ΔhflKC cells (Figure 4). Therefore, an increase in the lysogenic frequency indeed requires the stabilization of CII. Figure 4 Effect of infection by cIII -mutant lambda on in vivo proteolysis of CII. The proteolysis of CII was visualized in wild type (open circles) or AK990 (diamonds) cells infected with λcIII 67 . The expression of CII was induced with IPTG, and the cells

were infected with the phage after 20 minutes. Protein synthesis was stopped 25 minutes later with spectinomycin. The relative amount of CII was measured at regular intervals by western blotting followed by quantification using densitometric analysis. This enhanced stabilization of CII is observed only under conditions of phage infection, even when CIII is nonfunctional. Therefore in addition selleck inhibitor to CIII, there could be another as yet unidentified factor in λ that increases the stability of CII and hence, promotes lysogeny (see Figure 5A). The presence of such a CII-stabilizing factor (CSF) can only be demonstrated in HflKC-deleted

cells. Therefore, the activities of CSF and HflKC must have some connections (Figure 5B). Likewise, CIII and HflKC are likely to be connected as well. The different outcomes for deletion or overexpression of hflKC on lysogeny as well as on the stability of CII under various conditions are summarized in Figure 5A. Figure 5 The effect of deletion or overexpression of hflKC on λ lysogeny and on the stability of CII: A summary of results and possible mechanisms. (A) A summary of results published previously as well as reported in this study is shown schematically. Some unanswered questions that remain Resminostat are highlighted in the boxes. (B) Mechanisms for the stability of CII and the lysogenic outcome under various conditions are shown. HflB acts upon CII to digest CII, as indicated by the arrow. This digestion is inhibited by HflKC, by CIII or by the postulated CII-stabilizing factor CSF. The levels of inhibition are denoted by the lengths of the blunt lines. Possible crosstalk between HflKC and CIII or CSF are indicated by curved arrows. Dashed arrows denote lack of crosstalk. HflKC, CIII or CSF inhibits the digestion of CII. In wild type E. coli cells, this inhibition is unable to sufficiently stabilize CII, leading to normal plaques (left panel).

Curiously, six proteins in the molecular mass range of 40–42 kDa

Curiously, six proteins in the molecular mass range of 40–42 kDa have also been shown to be over-expressed Selleckchem INCB28060 in C. perfringens ATCC13124 cells when grown

on CMM, using 2-DE profiling of whole cell proteins. These proteins varied in their observed pI values from 5.6 – 7.0 and are likely to migrate closely on a one dimensional SDS-PAGE. The results indicate that with reference to TPYG grown cells, some additional proteins expressed in vivo (in mouse experimental gangrene model) are also expressed when C. perfringens ATCC13124 cells are grown on CMM. Based on the results obtained in the present investigation, it will be highly speculative to suggest that CMM provides host simulated conditions for C. perfringens. In a pre-gangrenous infection, C. perfringens cells encounter live muscle and immune cells that will be responding and fighting to kill the bacterium. By comparison, cooked meat media (CMM) is processed, granulated and boiled muscle tissue. Further work using proteome from cells obtained from infected host and those from

CMM and TPYG grown cells may provide further clue in this direction. Most of the cell envelope and up-regulated proteins existed as multiple isoelectropherotypes and often differences in their observed Selleckchem LY2874455 and theoretical pI values were more pronounced, compared to those observed for molecular masses [see Additional file 1]. We cannot exclude a possibility that there are major post translational oxyclozanide events in these proteins resulting in pI value differences. Nevertheless, earlier observations have indicated that different isoelectropherotypes of polypeptides in 2-DE gels do not always arise from true post translational modifications, but also from the 2-DE procedure itself [31, 32]. The outer surface of bacteria is of great importance to the understanding of bacterial pathogenesis. Elements of the

surface are implicated in bacterial defense mechanisms and virulence related functions e.g. adhesion, invasion, direct injury, and induction of septic shock. There is no information available with respect to surface proteins of this medically important bacterium. In the present study, several of the surface proteins and those over-expressed in CMM grown cells were largely assigned putative function in amino acid transport and metabolism [see Additional file 1], suggesting that this organism is adapted to protein rich environment of host tissue. Together, these identified and predicted proteins could be useful targets for the development of improved vaccines against gangrenous infections. Two of the surface proteins of C. perfringens, ornithine carbamoyltransferase and phosphoglycerate kinase have also been identified as immunogenic proteins in the outer surface protein preparation of S. agalactiae and S. pyogenes [24, 25]. Curiously, sera directed against the two proteins were shown to protect neonatal animals from S.

Fusion allows the nerve impulse to be delivered across

th

Fusion allows the nerve impulse to be delivered across

the synaptic junction. Botulinum neurotoxin G (BoNT/G) is the least studied of the seven serotypes. BoNT/G-producing organisms were first isolated by Gimenez and Ciccarelli in 1969 from soil samples taken from a cornfield in the Mendoza Province of Argentina [4]. The investigators indicated that a novel strain of bacterium produced an antigenically specific, heat-labile botulinum-like toxin that was not neutralized by any of the known botulinum antisera. The antitoxin developed using this strain only neutralized its homologous toxin and showed no activity on any of the other known types of BoNT [4]. Overall, nine strains of type G producing organisms have been isolated, two from Argentina and seven from Switzerland; none of which have ever been clearly implicated Selleck Sotrastaurin as the cause of paralytic illness or death in humans or

animals [5]. Type G organisms are historically associated with the C. botulinum species, because of their ability to produce botulinum neurotoxin [3, 4]. However, it is well known that botulinal toxin production is a poor parameter on which to base species identification and that the C. botulinum species is a taxonomic collection of several distinct species [3, 5–7]. Type/G producing organisms are classified as Clostridium argentinense [5]. This species includes 12 strains of bacteria from the genus Clostridium: nine toxigenic strains and three Poziotinib non-toxigenic strains. These strains are genetically and phenotypically distinct from all other strains of C. botulinum and other clostridial species

[5]. Two of the three non-toxigenic strains were once classified this website as C. subterminale, and the third as C. hastiforme. These strains were often reported to cause serological cross-reactions with type/G producing organisms and the BoNT/G protein in ELISA and Fluorescence Resonance Energy Transfer (FRET) detection assays [5, 8, 9]. The C. argentinense species can be distinguished from other asaccharolytic, proteolytic clostridia by a biochemical test that detects the production of a unique derivative of indole [5]. However, to avoid confusion among the medical and scientific communities, C. argentinense type/G producing organisms are still referred to as C. botulinum type/G [7]. Type/G toxin is produced in culture as a relatively large protein complex (L complex ~500 kDa) consisting of a neurotoxin (BoNT) and three neurotoxin-associated proteins (NAPs): two hemagglutinins (HA17 and HA70) and a nontoxic-nonhemagglutinin (NTNH) component. In addition, there is a gene expression protein (P21) that is responsible for regulating the expression of the four complex proteins. P21, however, is not associated with the toxin complex itself [10, 11].

These numbers for richness are considerably lower than found in H

These numbers for richness are considerably lower than found in HF urine (Table 1 and Figure 3A). The number of OTUs at 3% difference for the individual samples for both IC and HF

are indicated in https://www.selleckchem.com/products/tpca-1.html box plots (Figure 3B) for both V1V2 and V6 analysis. In general, fewer number of OTU clusters were observed for IC individuals than that for HF individuals. Ecological diversity measured by Shannon and inverse Simpson indices also indicate lower diversity in IC urine in comparison to what was seen in urine from HF (Figure 3C and D). Specifically, a significant (p < 0.05) decrease in inverse Simpson index in IC patients compared to HF was found for the V6 analysis. Taken together, the results for both V1V2 and V6 support each other and confirm that the urine community is less diverse in IC patients than in HF individuals. However, the compound screening assay single IC outlier with high richness and diversity (Figure 3B-D) also clustered outside the IC group in the clustering analysis done using taxonomy data (Figure 2) showing that there is also potential for variation within the IC community. Figure 3 Comparison of richness and diversity estimations of urine from interstitial cystitis (IC) patients and healthy females (HF). A: Rarefaction curves depicting number of OTUs (at 3% genetic difference) as function of the total number of

sequences for the combined sequence pool datasets for IC urine V1V2 and V6 (red and orange) and HF urine V1V2 and V6 (dark and light blue). The curves show a decreased estimate of species richness in the IC urine microbiome compared to the HF urine microbiome. B, C, and D: Box plots showing richness and diversity of 16S rDNA sequences. Boxes contain 50% of Casein kinase 1 the data and have lines

at the lower quartile (red), median and upper quartile (green) values. Ends of the whiskers mark the lowest and highest value. The plots show the results of a combined assessment of the eight urine samples in each HF and IC microbiome and with normalized numbers of sequences for OTU and Shannon index values (B and C). B: Observed OTU counts (at 3% genetic difference) of all urine samples taken from HF and IC, for both V1V2 and V6 datasets. C and D: Shannon index and inverse Simpson index at 3% sequence dissimilarity calculated to estimate diversity for both V1V2 and V6 datasets. Asterisks (*) indicate significant differences (Wilcox rank sum test: * p < 0.05). Note that a single sample (P2) in the IC community is the only outlier with the highest values for both richness and diversity (for both V1V2 and V6 analysis). The IC and HF urine also showed a degree of community similarity at 3% sequence dissimilarity level – about 12% and 9.5% of the total OTUs for V1V2 and V6, respectively, were present in both groups (Additional file 4: Figure S1).

Though such studies are crucial for identifying stimulus specific

Though such studies are crucial for identifying stimulus specific effects, they are unable to account for the immunomodulatory effects of live bacteria, which frequently employ multiple survival strategies in parallel. Viable pathogenic bacteria secrete active components in the intercellular space and in the invaded cells in order to modulate the cellular response. In order to track the early events of gram-positive induced immune activation, we examined the total transcriptional response of isolated peripheral human CD14+/CD11b+ monocytes, infected with the viable

bacterial pathogens: Listeria monocytogenes, Staphylococcus aureus and Streptococcus pneumoniae (hereafter referred to as LM, SA Etomoxir concentration and SP respectively). All three pathogens belong to the

group of low GC content bacteria. SP and SA are leading pathogens in cases of gram-positive sepsis and LM is a cause of meningitis in immunocompromised patients and also sepsis in newborns. We designed and established a protocol enabling the detection of pathological changes early in the onset of infections with gram positive pathogens, before usual clinical parameters are upregulated, in an easily accessible cellular sample material. For these purposes, we focused our experimental analysis of naïve monocytes, which are easier to work with in ex vivo conditions than granulocytes, selleckchem even though they are represented in much lower numbers in vivo than the latter. Peripheral monocytes also are among the first members of the host immune system to encounter pathogens after injury and epithelial penetration. We limited the infection to a short interval of 1 hour in the attempt to mimic the in vivo early reaction of the cells after first encountering

the pathogen but before the onset of clinically manifested inflammation. Using microarray analysis, we were able to detect the transcriptional upregulation or repression of a robust minimal set of genes in infected cells compared to untreated controls in the short interval of one Aspartate hour. Despite donor specific gene variations and despite the different invasion strategies of the bacteria studied, we identified a common program of gene expression induced by all three bacterial pathogens. This program is characterized by the upregulation of a key cytokine – interleukin 23 (IL23). Results Global response pattern of peripheral monocytes to infection To assess the global response we performed clustering of the correlation coefficients of the entire gene expression matrix comprising the unchallenged and the infected monocytes with all three pathogens (Figure 1). This revealed an interesting pattern. As can be seen from the figure, there are three main clusters. Cluster A comprising the controls, Cluster B comprising infection with L. monocytogenes (LM) and S. aureus (SA), and Cluster C comprising infection with S. pneumoniae (SP).

They bind to DNA [3, 5] preferring AT-rich DNA-sequences [11] as

They bind to DNA [3, 5] preferring AT-rich DNA-sequences [11] as well as to laminin, hyaluronic acid, heparin, and chondroitin sulphate [5, 6, 12]. The data available so far portray Hlp as multi-faceted proteins, and accordingly a find more variety of possible functions have been ascribed to Hlp. Hlp were suggested to impact DNA packaging, protection of DNA from enzymatic and non-enzymatic strand breakage [11], gene regulation [1], nucleic acid metabolism, non-homologous-end-joining repair [13], adaptation to hypoxic conditions [2],

induction of dormancy [2], adaptation to cold shock [14], adhesion [6, 9, 12, 15–17], cell wall biogenesis [10] and regulation of growth rate [1, 5, 10]. A role in transition to the non-culturable state and in resuscitation from the non-culturable state was shown in M. smegmatis[18]. Whiteford et al. [19] investigated the growth characteristics of an M. smegmatis with a deletion of hlp. They found that the mutant showed less aggregation in broth cultures. Furthermore, they observed an increased sensitivity towards Isoniazid. The M. smegmatis mutant also was affected in UV-resistance and resistance towards freezing/thawing. Takatsuka et al. [20] have recently shown that Hlp has a similar activity to ferritin superfamily proteins and protects DNA by ferroxidase activity. It furthermore captures iron molecules and functions Selleck CDK inhibitor as iron storage protein. Approaches to elucidate the

functions of Hlp by mutagenesis did not always confirm the expected roles of Hlp [2, 15, 21]. Our own attempts to generate a MDP1 deletion mutant had failed. Furthermore and in line with our own experience, Sassetti et al. [22] had shown by high density mutagenesis that the gene Rv2986c from M. tuberculosis, which is homologous to MDP1 from BCG, is required for optimal growth of M. tuberculosis. We therefore followed the strategy Anidulafungin (LY303366) to analyse Hlp functions by down-regulation of Hlp expression by antisense-technique. Advantages of this technique are the possibility to analyse essential genes and to repress genes present in several copies. In mycobacteria the antisense-technique

has been applied to down-regulate ahpC from M. bovis[23], dnaA from M. smegmatis[24], FAP-P from M. avium subsp. paratuberculosis[25] or pknF from M. tuberculosis[26]. In a previous study we described the generation of the antisense-strain M. bovis BCG (pAS-MDP1) which carries the plasmid pAS-MDP1 causing a reduction of MDP1 expression in BCG by about 50% [27]. We analysed BCG (pAS-MDP1) with respect to general growth characteristics. The down-regulated BCG grew faster in broth culture and achieved a higher cell mass in the stationary phase. Similarly, growth was enhanced in human and murine macrophage-like cell lines. A further important finding was the reduced protein synthesis occurring under hypoxic conditions [27]. These findings support a role of MDP1 in growth regulation of M. bovis BCG.

1 and 448 1 respectively This experiment was performed twice wit

1 and 448.1 respectively. This experiment was performed twice with similar results. Figure 4  Leptospira interrogans  endogenously expresses N-acetylneuraminic acid (Neu5Ac). L. interrogans was grown in EMJH medium or in a chemically defined medium containing no exogenous sialic acid (this was confirmed by HPLC, not shown). Covalently bound

Sias were released by mild acid hydrolysis and analyzed by DMB-derivatization and HPLC as described in previous AR-13324 clinical trial figures and Materials and Methods. This experiment was performed twice with similar results. Composition and phylogenetic analysis of NulO biosynthetic gene clusters and enzymes Next we performed analysis of the composition and phylogeny of the putative NulO biosynthetic gene clusters and the enzymes they encode in L. interrogans serovars Lai (strain 56601) and Copenhageni (strain L1-130). Consistent with

the biochemical analysis of L. interrogans, genomic analysis of the NulO gene cluster reveals that the organism encodes a complete pathway for di-N-acetylated nonulosonic acid biosynthesis (see Table 1 in comparison with Figure 5). There are multiple distinct open reading frames encoding synthesis of aminotransferases, NulO synthases, and CMP-NulO synthetases (see Table 1 and Figure 5), suggesting that L. interrogans may learn more express multiple nonulosonic acid species, a conclusion supported by our biochemical investigations (Figure 2 and Figure 3). Table 1  L. interrogans  encodes a complete pathway for legionaminic acid synthesis  Campylobacter enzymes for legionaminic acid biosynthesis[14, 17–21]  C. jejuni Pathway number (Figure 5)  L. interrogans L1-130 & 56601 NCBI accession numbers Predicted L. interrogans Pathway number (Figure 5) Predicted enzymatic Function PmtE (cj1329) Florfenicol 1 YP_002106 1 Glc-1-P guanyltransferase     NP_711792     GlmU 2 YP_000413 2 (housekeeping)     NP_714003   N-acetyltransferase

LegB (cj 1319) 3 YP_002111 3 4,6-dehydratase     NP_711787     LegC (cj1320) 4 YP_002110 4 Aminotransferase in legionaminic acid synthesis (Figure 6A)     NP_711788         YP_002103 4, 13, or ? Aminotransferase     NP_711795     LegH (cj1298) 5 YP_002109 5 N-acetyltransferase     NP_711789     LegG (cj1328) 6 YP_002107 6 2-epimerase/NDP sugar hydrolase in legionamimic acid synthesis     NP_711791     LegI (cj1327) 7 YP_002108 7 Legionaminic acid synthase (Figure 6B)     NP_711790         YP_002104 10 Legionaminic or neuraminic acid synthase (Figures 6B & 7)     NP_711794     LegF (cj1331) 8 YP_002102 8 or 11 CMP-Legionaminic acid or neuraminic acid synthetases (Figure 6C)     NP_711796         YP_002112 8 or 11       NP_711786     Figure 5 Schematic of pseudaminic, legionamimic, and neuraminic acid biosynthetic pathways. Studies of nonulosonic acid biosynthesis at the enzymatic level have been carried out with greatest resolution using C. jejuni and H. pylori as model systems [14, 17–21, 35].

difficile surface layer protein (SLP) has been

difficile surface layer protein (SLP) has been Selleck MDV3100 shown to contain antigenic epitopes and play role in colonization of the bacterium to gastrointestinal tissues [8, 10]. Complete genome sequences for three of its widely studied strains; C. perfringens strain 13, C. perfringens ATCC 13124T (a gas gangrene isolate and the species type strain), and C. perfringens SM101 (enterotoxin-producing food poisoning strain) have been recently determined

and compared [12, 13]. Several striking findings have emerged from the complete genome sequencing data of this clostridial pathogen. Comparisons of the three genomes have revealed considerable genomic diversity with >300 unique “”genomic islands”" identified and using PCR based analysis it was also demonstrated that the large genomic islands were widely variable across a large collection of C. perfringens strains [12]. Proteome maps of

sequenced organism are important research tools for the authentication of hypothetical proteins, the identification of components of different cellular proteome fractions and for yielding information concerning the occurrence and abundance of proteins. Such proteome maps in the public domain have been generated for many pathogens GSK1120212 mouse and are of great value in identifying new virulence factors and the antigens of potential diagnostic and/or curative value against infections with pathogens. Despite a sudden spurt of activity towards proteomic characterization of bacterial

pathogens, for reasons unknown, clostridia have largely been ignored. Clostridium difficile is the only clostridial species for which analysis of proteome has been carried out to some extent [8, 10, 14]. To invade, multiply and colonize tissues of the host, a pathogen must be able to evade the host immune system, and obtain nutrients essential for growth. The factors involved in these complex processes are largely unknown and of crucial importance to understanding microbial pathogenesis. Growth of microorganisms FER in vitro, under conditions which mimic certain aspects of the host environment, such as temperature [15], pH [16], nutrient conditions, and interaction with host derived cells [17], can provide valuable information on microbial pathogenesis. Proteome analysis is one of the best tools for understanding the basic biology of microorganisms including pathogenesis, physiology, and mechanisms of avoiding host immune system. In this study we report identification of major surface and cell envelope proteins from Clostridium perfringens ATCC13124 and those differentially expressed in cells grown on cooked meat medium (CMM) in comparison with cells grown in reference state TPYG (tryptose-yeast extract-glucose) medium. Cooked meat medium [18] provides substrate in the form of muscle tissue, for the myonecrotic cells of C. perfringens which produces phospholipase C as one of its major virulence factor.

For bacteremia, cure rates were 71 4% (15 of 21 subjects) compare

For bacteremia, cure rates were 71.4% (15 of 21 subjects) compared with 58.8% (10 of 17 subjects) for the ceftaroline and ceftriaxone groups, respectively (difference 12.6%, 95% CI −17.6% to 41.6%) [44]. At the late

follow-up visit (21–35 days after completion of therapy), relapse rates between the two treatment arms were similar in the CE population: 1.9% for the ceftaroline group and 1.2% for the ceftriaxone group (difference 0.7%, 95% CI −1.4% to 2.9%) [44]. Pooled post hoc exploratory analysis requested by the FDA to assess clinical improvement on day 4 of study therapy in participants with a confirmed bacterial pathogen at baseline showed a weighted difference in clinical response of 11.4% (95% CI 0.6–21.9%) in favor of ceftaroline 4-Hydroxytamoxifen [48]. Table 3 Summary of clinical cure rate at the test-of-cure visit in the co-primary analysis populations, FOCUS and CANVAS trials [12–15, 44, 47] Trial MITTE CE FOCUSa Clinical cure % (no. of cured/total no.) Differenceb (95% CI) Clinical cure % (no. of cured/total no.) Differenceb (95% CI) Ceftaroline Ceftriaxone Ceftaroline Ceftriaxone selleck kinase inhibitor 1 83.8 (244/291) 77.7 (233/300) 6.2 (−0.2, 12.6) 86.6 (194/224) 78.2 (183/234) 8.4 (1.4, 15.4) 2 81.3 (235/289) 75.5 (206/273) 5.9 (−1.0, 12.7) 82.1 (193/235) 77.2 (166/215) 4.9 (−2.5, 12.5) 1 and 2 82.6 (479/580) 76.6 (439/573) 6.0c

(1.4, 10.7) 84.3 (387/459) 77.7 (349/449) 6.7c (1.6, 11.8) Trial MITT CE CANVASa Clinical cure % (no. cured/total no.) Differenceb (95% CI) Ceftaroline Vanc/Az Ceftaroline Vanc/Az 1 86.6 (304/351) 85.6 (297/347) 1.0 (−4.2, 6.2) 91.1 (288/316) 93.3 (280/300) −2.2 (−6.6, 2.1) 2 85.1 (291/342) 85.5 (289/338) −0.4 (−5.8, 5.0) 92.2 (271/294)) 92.1 (269/292) 0.1 (−4.4, 4.5) 1 and 2 85.9 (595/693) 85.5 (586/685) 0.3 (−3.4, learn more 4.0) 91.6 (559/610) 92.7 (549/592) −1.1 (−4.2, 2.0) CE clinical efficacy population, CI confidence interval, MITT modified intent-to-treat population, MITTE modified intent-to-treat efficacy population, Vanc/Az vancomycin plus aztreonam combination aNon-inferiority margin was set at −10% for both FOCUS and CANVAS trials bTreatment

difference: cure rate ceftaroline − cure rate comparator group cWeighted treatment difference The CANVAS Trials The CANVAS (CeftAroliNe Versus vAncomycin in Skin and skin structure infections) 1 and 2 studies (NCT00424190 and NCT00423657, respectively) were multinational, multicenter, phase 3, double-masked, randomized, active comparator-controlled trials designed to evaluate the safety and efficacy of monotherapy with ceftaroline fosamil 600 mg IV every 12 h compared with a combination of vancomycin 1 g every 12 h plus aztreonam 1 g every 12 h IV for 5–14 days for the treatment of ABSSSI [14, 15, 45, 47] Dose adjustments for renal impairment by unblinded pharmacists were based on creatinine clearance and institutional guidelines.

Am J Physiol 1998, 274:G1061–1067 PubMed 14 Khuri FR, Wu H, Lee

Am J Physiol 1998, 274:G1061–1067.PubMed 14. Khuri FR, Wu H, Lee JJ, Kemp BL, Lotan R, Lippman SM: Cyclooxygenase-2 overexpression is a marker of poor prognosis in stage I non-small cell lung cancer. Clinical Cancer Research 2001, 7:861–867.PubMed 15. Xu Z, Choudhary S, Voznesensky O, Mehrotra M, Eltanexor Woodard M, Hansen M: Overexpression of COX-2 in human osteosarcoma cells decreases proliferation and increases apoptosis. Cancer Res 2006, 66:6657–6664.PubMedCrossRef 16. Klein RD, Van Pelt CS, Sabichi AL, Dela Cerda J, Fischer SM, Furstenberger G: Transitional cell hyperplasia and carcinomas in urinary bladders of transgenic mice with keratin 5 promoter-driven cyclooxygenase-2

overexpression. Cancer Res 2005, 65:1808–1813.PubMedCrossRef 17. Thun MJ, Henley SJ, Patrono C: Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, Cell Cycle inhibitor pharmacologic, and clinical issues. J Natl Cancer Inst 2002, 94:252–266.PubMed 18. Fosslien E: Biochemistry of cyclooxygenase (COX)-2 inhibitors and molecular pathology of COX-2 in neoplasia. Crit Rev Clin Lab Sci 2000, 37:431–502.PubMedCrossRef 19. Wang R, Wang X, Lin F, Gao P, Dong K, Zhang HZ: shRNA-targeted cyclooxygenase (COX)-2 inhibits proliferation, reduces invasion and enhances chemosensitivity in laryngeal carcinoma cells. Mol Cell Biochem 2008, 317:179–188.PubMedCrossRef 20. Fujita H,

Koshida K, Keller ET, Takahashi Y, Yoshimito T, Namiki M: Cyclooxygenase-2

promotes prostate cancer progression. Prostate 2002, 53:232–240.PubMedCrossRef 21. Klimp AH, Hollema H, Kempinga C, van der Zee AG, de Vries EG, Daemen T: Expression Oxymatrine of cyclooxygenase-2 and inducible nitric oxide synthase in human ovarian tumors and tumor-associated macrophages. Cancer Res 2001, 61:7305–7309.PubMed 22. Hida T, Yatabe Y, Achiwa H, Muramatsu H, Kozaki K, Nakamura S: Increased expression of cyclooxygenase 2 occurs frequently in human lung cancers, specifically in adenocarcinomas. Cancer Res 1998, 58:3761–3764.PubMed 23. Hwang D, Scollard D, Byrne J, Levine E: Expression of cyclooxygenase-1 and cyclooxygenase-2 in human breast cancer. J Natl Cancer Inst 1998, 90:455–460.PubMedCrossRef 24. Attiga FA, Fernandez PM, Weeraratna AT, Manyak MJ, Patierno SR: Inhibitors of prostaglandin synthesis inhibit human prostate tumor cell invasiveness and reduce the release of matrix metalloproteinases. Cancer Res 2000, 4629–4637. 2000/09/02 ed 25. Tsujii M, DuBois RN: Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase 2. Cell 1995, 83:493–501.PubMedCrossRef 26. Fujita T, Matsui M, Takaku K, Uetake H, Ichikawa W, Taketo MM: Size- and invasion-dependent increase in cyclooxygenase 2 levels in human colorectal carcinomas. Cancer Res 1998, 58:4823–4826.PubMed 27.