There are many generic olanzapine orodispersible formulations, bu

There are many generic olanzapine orodispersible formulations, but their relative disintegration and dispersion times have never been studied to our knowledge. Variation in dispersion times might

be expected, depending on the different fast dissolve/disintegration technologies used to manufacture the tablets and/or the disintegration test used to evaluate them. Olanzapine Zydis® (also known as Velotab®) is manufactured by Catalent Pharma Solutions (Somerset, NJ, USA), and is made by a freeze drying process that provides a low-density, highly porous structure that readily Buparlisib concentration disintegrates in the oral cavity. Although bioequivalence is accepted for generic ODTs, the time it takes for an ODT to disintegrate and dissolve in the oral cavity may potentially impact clinical parameters such as patient acceptance and adherence to treatment. For olanzapine Zydis® ODT, the elapsed time for

initial and complete disintegration was measured in two small in vivo studies [14, 15]. However, these studies used different methods: one took the first measurement of initial disintegration at 15 s, while the other took the first measurement CB-5083 manufacturer at 5 s. It is desirable to compare disintegration times among different products using the same methodology. Given the obvious challenges of standardizing in vivo assessments, the objective of our current in vitro comparison was to investigate in vitro disintegration time and dissolution rate differences of various generic formulations of olanzapine ODT relative to olanzapine Zydis® in simulated saliva. We also compared the chemical and physical properties of each ODT and measured in vitro disintegration time for risperidone ODT [16] as a comparator. 2 BAY 1895344 manufacturer Materials and Methods All types of olanzapine ODT that could be obtained were evaluated (Table 1). Eleven different examples were filmed to determine disintegration times, and all were evaluated for manufacturing method, dissolution characteristics and formulation differences,

including the freeze dried Zydis® formulation of olanzapine ODT and Paclitaxel risperidone ODT. A Canon XHL1 HD camera (Canon, Tokyo, Japan) was used to capture a 3-min disintegration event for each ODT product added to 30 mL of non-agitated 37 °C (initial temperature) simulated saliva solution in a 10-cm Petri dish. Disintegration was defined as the time it took a tablet to reach full dispersion after addition to the artificial saliva (see Table 2 for the formulation, based on formulations described in Giannola et al. [17] and Gal et al. [18]). Drug product excipient data were obtained from published product literature. Dose form and manufacturing method (compressed tablet, lyophilized wafer) were determined by microscopic/visual observation.

fumigatiaffinis and A lentulus [7, 8], whereas A fumigatus is u

fumigatiaffinis and A. lentulus [7, 8], whereas A. fumigatus is usually susceptible to the GSK2245840 clinical trial antifungals that are available for clinical treatment [19, 20]. Few clinical cases of invasive aspergillosis have been reported in which the antifungal treatment was repeatedly modified until the correct identification of the fungal agent and the administration of the appropriate antifungal treatment [17, 18]. Considering that A. fumigatus may represent a considerable part of all clinical cases of aspergillosis, molecular characterization is essential for the correct identification of species within the section Fumigati. In this study, we developed a multiplex PCR strategy that was able

to differentiate A. fumigatus from all the other related species within the section Fumigati. Rabusertib mouse We could not test all of the species of section Fumigati, as some of them are extremely rare. However, we believe that the present multiplex PCR can be widely used, as A. lentulus is more closely related to A. fumigatus than most species in section Fumigati (e.g. A. viridinutans) [4, 5], and a distinct electrophoretic profile was observed with two strains

of this species. It is expected that other species of section Fumigati that are genetically distant from A. fumigatus can be distinguished by employing Y-27632 supplier this multiplex PCR (see additional file 2 in supplemental data). A simple electrophoresis profile after PCR amplification clearly separates two species, A. fumigatus and N. udagawae, from a second group of fungal isolates of section Fumigati. This method is furthermore amenable to automation. Compared to previously described methodologies for A. fumigatus identification within its section [10–13], the proposed method facilitates the molecular recognition of this species by employing a single multiplex PCR and avoiding the need for restriction enzymes and specialized Ceramide glucosyltransferase equipment. This approach is cheap and simple and would be very useful

in clinical labs that routinely screen and perform the molecular identification of several mould isolates. The proposed new assay proved to be specific and highly reproducible for targeting A. fumigatus within the section Fumigati and outside this section. A list of fungal species related to A. fumigatus could be identified by sequencing partial regions of βtub and rodA. A group of 14 unique species and two groups of species of section Fumigati were distinguished by point mutations in βtub and rodA. This work presents the first record of polymorphic sites available for the rapid identification of species within the section Fumigati following the analysis of more than 450 βtub and rodA sequences. This list represents a practical guide for the molecular recognition of rare fungal species, and it can certainly be expanded in the near future when more sequences of βtub and rodA are available.

0379; LB: fRF, p = 0 0385; LB: uRF, p = 0 0381, dRF: fRF, p = 0 0

0379; LB: fRF, p = 0.0385; LB: uRF, p = 0.0381, dRF: fRF, p = 0.0121 and fRF: uRF, p = 0.0655; dRF: uRF, p = 0.1077. Proteomics analysis (i) Bottom-up LC-MS/MS analysis of the O157 cell pellet and lysate fractions

generated in Experiment I provided insights into the proteins being expressed by O157 in different media, under different growth conditions and at extended incubation time points. A total of 585 protein (2284 spectra) hits p38 MAPK signaling were identified by setting minimum characteristics for the identification confidence. However, of these only 218 O157 proteins matched a GS-1101 cell line higher threshold cut off, with 90% protein-80% peptide probability in the Scaffold Viewer, and RG7112 cost hence, were selected for analysis. The 218 O157 proteins were differentially expressed: 90 only under aerobic conditions, 37 only under anaerobic conditions and 91 under both conditions (data not shown), accounting for fewer proteins under anaerobic conditions. Interestingly, none of the O157 proteins expressed aerobically or anaerobically in either media were associated with direct virulence (e.g., the Locus of Enterocyte Effacement [LEE]-encoded proteins or Shiga toxins)

but were primarily associated with sequences homologous to other E. coli genomes (Backbone) (Additional file 1: Table S1). Considering that the rumen is an anaerobic microbiome, the 128/218 O157 proteins expressed anaerobically were examined in greater detail. www.selleck.co.jp/products/cetuximab.html These proteins were either unique to growth in LB (93/128), dRF (2/128), fRF (10/128) or, expressed in more than one media (14/128 in LB/dRF/fRF, 9/128 in dRF/fRF) (Figure 2). Specifically, the 35 proteins expressed anaerobically in fRF and dRF (unique and shared combined), were functionally associated with the osmotic adaptation pathway (OsmE), anaerobic respiration and oxidative stress pathway (YggE, MoaB, DmsB, FdoH), heat stress response (HchA), carbon starvation response (Slp),

energy metabolism and biosynthetic pathways (glycolytic/gluconeogenesis pathway, amino acid biosynthesis: AldoC, Crr, AnsB, PykF, Eno, GpmA, GadpH, CysK, Ttc, AhpC, YhcB), chaperones (DnaK, GroEL, HchA), transport (LamB, ManX, FadL, RbsB), outer membrane proteins/porins/channel (OmpC, TolC, YdeN, Slp, OmpA), tellurite resistance (TerD), lysozyme inhibitor (Ivy), chemotaxis (GgbP), and motility (FliC) (Table 3; Additional file 1: Table S1). Figure 2 Distribution of 128 anaerobically expressed O157 proteins, identified using bottom-up proteomics, amongst the media tested. LB, Luria-Bertani broth; dRF, depleted and filtered rumen fluid; fRF, filtered rumen fluid.

PubMedCrossRef 35 Lin NT, Chiou PY, Chang KC, Chen LK, Lai MJ: I

PubMedCrossRef 35. Lin NT, Chiou PY, Chang KC, Chen LK, Lai MJ: Isolation and characterization of phi AB2 : a novel bacteriophage of Acinetobacter baumannii . Res Microbiol 2010, 161:308–314.PubMedCrossRef 36. Hagens S, Loessner MJ: Application of bacteriophages for detection and control of foodborne pathogens. Appl Microbiol Biotechnol 2007, 76:513–519.PubMedCrossRef 37. Iriarte FB, Balogh B, Momol MT, Smith LM, Wilson M, Jones JB: Factors affecting survival of bacteriophage on tomato leaf surfaces. Appl Environ Microbiol selleck products 2007, 73:1704–1711.PubMedCrossRef 38. Chang KC, Lin NT, Hu A, Lin YS, Chen LK, Lai MJ: Genomic analysis of bacteriophage ϕAB1 , a ϕKMV -like virus infecting multidrug-resistant Acinetobacter baumannii

. Genomics 2011, 97:249–255.PubMedCrossRef 39. McConnell MJ, Perez-Ordonez A, Perez-Romero P, Valencia R, Lepe JA, Vazquez-Barba I, Pachon J: Quantitative real-time PCR for detection of Acinetobacter baumannii colonization in the hospital environment. J Clin Microbiol 2012, 50:1412–1414.PubMedCrossRef 40. Yang H, Liang L, Lin S, Jia S: Isolation and characterization of a virulent bacteriophage AB1 of Acinetobacter baumannii . BMC Microbiol 2010, 10:131.PubMedCrossRef 41. Boyce JM, Kelliher S, Vallande N: Skin irritation and dryness associated with two hand-hygiene regimens: soap-and-water hand washing versus hand antisepsis with an alcoholic hand gel. Infect Control Hosp Epidemiol 2000, Temsirolimus price 21:442–448.PubMedCrossRef

42. Goroncy-Bermes P, Schouten MA, Voss A: In vitro activity of a nonmedicated handwash product, chlorhexidine, and an alcohol-based hand disinfectant against multiply resistant gram-positive microorganisms. Infect Control Hosp Epidemiol 2001, 22:194–196.PubMedCrossRef 43. Trick WE, Vernon PAK6 MO, Hayes RA, Nathan C, Rice TW, Peterson BJ, Segreti J, Welbel SF, Solomon SL, Weinstein RA: Impact of ring wearing on hand contamination and comparison of hand hygiene agents in a hospital.

Clin Infect Dis 2003, 36:1383–1390.PubMedCrossRef 44. Mendez J, Jofre J, Lucena F, Contreras N, Mooijman K, Araujo R: Conservation of phage reference materials and water samples containing bacteriophages of enteric bacteria. J Virol Methods 2002, 106:215–224.PubMedCrossRef 45. Adams M: Bacteriophages. Edited by: Hershey AD. New York: Interscience; 1959:137–159. Competing interests The authors declare that they have no competing interests. Authors’ Talazoparib in vivo contributions LKC and YLL performed the experiments and analyses. AH and KCC provided test materials and participated in the analysis of bacteria. NTL and MJL participated in the bacteriophage experiments. CCT conceived of the study and drafted the manuscript. All authors read and approved the final manuscript.”
“Background Sika deer (Cervus nippon) represent the most ancient and primitive members of the genus Cervus because of the simple structure of their antlers, which is very distinct from those of reindeer.

Using several-fold higher concentrations of the test β-lactam ant

Using several-fold higher concentrations of the test β-lactam antibiotic, compared to the probe, enhances the likelihood that the antibiotic will be the preferred substrate of the lactamase in the competition reaction in the assay. The reduced selleck chemical fluorescence indirectly reflects the ability of the β-lactamase to bind and cleave the tested antibiotic (large difference = antibiotic can be readily

bound and hence cleaved and inactivated). Notably, unlike growth based conventional AST methods, the end-point 7-Cl-O-Nec1 in vivo of the β-LEAF assay is not bacterial viability or differences in growth pattern. The read-out of the assay is fluorescence, which reflects probe cleavage due to the enzymatic activity of the β-lactamase. Importantly, the β-LEAF assay is rapid compared to the conventional growth based AST methods (1 h versus 20–24 h for disk diffusion/MIC conventionally or ~8 h with automated instruments). The observation in Figure 2 of low to negligible fluorescence in β-LEAF + cefazolin reactions with all β-lactamase ‘positives’ (#1, #6, #18, #19, #20) suggests that cefazolin can be readily targeted and inactivated by the respective lactamases, and would be anticipated to be a less effective treatment option for these bacteria. An expectation of this assay is that the reduction in probe fluorescence in the presence of an antibiotic will be inversely proportional to its predicted activity

against the pathogen. If fluorescence is completely reduced in the presence of an antibiotic, then the respective antibiotic can be readily cleaved and inactivated by β-lactamase. However, if despite the ‘saturating’ amount of antibiotic, some fluorescence DZNeP price increase reflecting probe cleavage is still observed (e.g. cefepime reactions in Figure 3), the lactamase may not be capable of effectively destroying the antibiotic, and the antibiotic predicted as likely to be active. In experiments with multiple antibiotics (Figure 3) a ratio of the cleavage rate of β-LEAF in presence of an antibiotic to the cleavage rate of β-LEAF alone, for each antibiotic tested, is shown in Table 4. For β-lactamase based resistance, the ratio of cleavage

rates closer to 1 (Table 4) would indicate greater β-lactam antibiotic efficacy. With more rigorous testing Niclosamide from multiple data sets on a large number of isolates, cut-offs could be set up to develop the ratios as a ‘β-lactamase-based antibiotic activity/susceptibility index’ within specific limits. We recognize that there are a wide variety of lactamases, and note that with appropriate kinetic analysis (such as building on our previous study [50]), the approach presented here has the potential of characterizing the different lactamases. The motivation for the choice of antibiotics used in this initial study was to test three different generations of cephalosporin antibiotics. Cephalosporins are a standard treatment for skin and soft-tissue infections [58, 59].

A mutant of Rhizobium etli, that

did not accumulate PHB,

A mutant of Rhizobium etli, that

did not accumulate PHB, was shown to significantly fix more nitrogen than the isogenic wild type [10, 11], whereas non-fixing nifH mutants of R. etli[12] and Bradyrhizobium japonicum[13] accumulated more PHB than their isogenic nitrogen-fixing parental strains. There is a conflict between rhizobia Selleck Duvelisib and legumes over the rate of PHB accumulation, due to the metabolic tradeoff between nitrogen fixation and PHB accumulation. Therefore, PHB biosynthesis and accumulation in species of rhizobia may be controlled to balance the tradeoff, but the buy CH5183284 mechanism underlying this control has not yet been fully explained. One of the best studied microorganisms with respect to PHB biosynthesis and accumulation is the Gram-negative bacterium Ralstonia eutropha[14]. It synthesizes PHB using the three PHB synthetic genes: phbA, which encodes 3-ketoacyl-CoA thiolase; phbB, which encodes acetoacetyl CoA reductase; and phbC, which encodes the enzyme PHB synthase. PHB degradation, however, is performed by PHB depolymerase, which is encoded by phaZ. Phasins, encoded by phaP, are a class of low-molecular-mass amphipathic proteins that form a layer at the surface

Selleckchem Proteasome inhibitor of the PHB granule and stabilize it [15]. The R. eutropha possesses at least four phaP paralogs identified so far [16]. Expression of the major phasin, encoded by phaP1, is regulated by the transcriptional crotamiton repressor PhaR [17, 18]. Under conditions less favorable for PHB biosynthesis, PhaR binds to the phaP1 promoter region to repress transcription of this gene. After the onset of PHB biosynthesis, when the nascent PHB granules gradually form, PhaR leaves the promoter and binds to the granules so that phaP1 is transcribed

and translated. During the later stages of PHB accumulation, PhaR is estimated to bind no longer to the granules as it is displaced by PhaP1 phasin. The displaced PhaR returns to bind to the phaP1 promoter and represses transcription again [16]. Most members of the Rhizobiaceae are known to possess single copies of the PHB biosynthesis genes. For instance, strains of Sinorhizobium meliloti, the symbionts of alfalfa, regarded as one of the model organisms to study symbiotic nitrogen fixation, are characterized to have a single set of the genes for PHB metabolism, namely phbA, phbB, phbC, and phaZ[19, 20], whereas two paralogous genes, phaP1 and phaP2, encode functional phasins [21]. On the other hand, strains of B. japonicum, the symbionts of soybean, are known to accumulate a large amount of PHB [22], and the B. japonicum USDA110 genome was found to contain five paralogs of phbC, as well as two paralogs of phbAB[23]. This genetic redundancy may suggest a functional importance that has not yet been fully elucidated. In this study, we examined the expression profiles of the paralogs relevant to PHB metabolism in free-living B.

Science 1994, 266:1380–1383 PubMedCrossRef 45 Fiala KI, Sokal RR

Science 1994, 266:1380–1383.PubMedCrossRef 45. Fiala KI, Sokal RR: Factors determining the accuracy of cladogram estimation-evaluation using computer-simulation. Evolution 1985, 39:609–622.CrossRef 46. Kingman JFC: The Coalescent. Stochastic Processes and their applications 1982, 13:235–248.CrossRef Authors’ contributions JC conceived and designed the study, performed and interpreted

the phylogenetic and statistical analyses, participated in the collection of the sequence data and animal assays, and drafted the manuscript. QC performed the PCR amplification and participated in the collection of the sequence data. LJ participated in evaluation of the results and in revision of the manuscript. CC and FB participated in the PCR amplification, biochemical tests and animal assays. JW and FM participated in the analysis of sequence data. GSK872 manufacturer WF supervised the project, participated in the design of the study and data interpretation,

and helped draft the manuscript. All authors read and approved the final manuscript.”
“Background Globally, Salmonella enterica subsp. enterica is one of the leading food-borne pathogens. For example in 2006 in the United States, Salmonella enterica subsp. enterica caused 45.808 registered GSK126 chemical structure cases of salmonellosis, corresponding to an incidence of 15 cases/100,000 inhabitants [1]. Furthermore the actual number of infections is estimated to be 38 times higher [2]. In Denmark, there were 1658 registered cases of CB-839 concentration salmonellosis (incidence of 30 cases/100,000 inhabitants) in 2006 [3]. Salmonella serotype Typhimurium, denoted S. Typhimurium, accounted for 17% of the salmonellosis cases in the USA and 25% of the Danish cases [1, 3]. The outcome of human infection ranges from mild self-limiting diarrhoea to severe diarrhoea that requires hospitalization. In rare cases, often among immunocompromised patients, salmonellosis can be fatal. Several factors in both the host and the bacteria influence the outcome of an infection. Clearly an important aspect of human infection is the immune state of the patient. It has been shown that immunocompromised Tolmetin patients are more prone to develop a severe infection

[4]. Another important aspect of human infection is the intestinal microbiota of the host. Ingestion of antibiotics is known to affect the intestinal microbiota leaving the host more prone to infection and disease caused by S. Typhimurium [5]. Significant bacterial factors for the outcome of infection are encoded by a wide range of genetic elements, including plasmids, prophages and Salmonella Pathogenicity Islands (SPIs). A total of 14 SPIs have been described so far [6]. SPI-1 encodes type 3 secretion system 1 (T3SS-1) that causes secretion and translocation of a range of bacterial proteins to the host cell. SPI-2 encodes T3SS-2 that allows intracellular survival and replication [7]. Different S. Typhimurium strains share more than 99% genomic content [8]. The detected variation within S.

6) PFGI-1 does not encode a Rep protein, and it is not clear whe

6). PFGI-1 does not encode a Rep protein, and it is not clear whether it replicates by a theta-type or strand displacement mechanism, although the latter has been suggested for pKLC102 [30]. Like some conjugative plasmids, PFGI-1 carries homologues of the stress-inducible genes umuC (PFL_4692) and umuD (PFL_4691), which encode a putative lesion bypass DNA polymerase and a related accessory protein,

respectively. Such genes may be involved in plasmid DNA repair and umuDC-mediated mutagenesis, which could allow plasmids to adapt more quickly to new bacterial hosts [41]. PFGI-1 also contains a cluster of 10 genes, pilLNOPQRSTUVM (PFL_4675 through PFL_4683) (Fig. 6), that spans over 10 kb and Selleck RSL 3 closely resembles part of the pil region from the self-transmissible E. coli plasmid R64 [42]. In E. coli, these genes are involved in production of thin flexible sex pili required for mating and transfer of R64 in liquid media. The similarity between the pil clusters of R64 and PFGI-1 suggests that the latter encode mating pili rather than type IV pili involved in bacterial twitching motility, adherence to host cells, biofilm formation and phage sensitivity [43]. P. Barasertib price fluorescens Pf-5 has the capaCity to produce type IV pili,

and the corresponding biosynthetic genes are located in at least three clusters found outside of PFGI-1. The PFGI-1 ITF2357 pil cluster contains genes for pilin protein PilS (PFL_4680), prepilin peptidase PilU (PFL_4681), outer membrane protein PilN (PFL_4676), nucleotide binding protein PilQ (PFL_4678), integral membrane protein PilR (PFL_4679), and pilus adhesin PilV (PFL_4682). Unlike R64, PFGI-1 does not include a shufflon PIK3C2G region that determines recipient specifiCity in liquid matings via generation of different adhesin types [42, 44]. Finally, PFGI-1 carries genes

encoding proteins that may be involved in conjugal DNA transfer. PFL_4696 and PFL_4706 encode for TraG-like coupling proteins that may function as membrane-associated NTPases, which during conjugation would mediate transport of DNA covalently linked to a putative relaxase protein (the product of PFL_4751). Recent studies have demonstrated that ICEs are a major component of a flexible gene pool of different lineages of Gram-negative Proteobacteria [45–47]. Metabolically versatile members of the Pseudomonadaceae are no exception, with ICEs having been identified among strains of P. aeruginosa [29–32], P. syringae [36, 48], and P. fluorescens [49]. Comparison of PFGI-1 with islands from other Pseudomonas spp. reveals at least six highly conserved gene clusters (Fig. 7).

holarctica subclades identified by Vogler et al and Svensson et

holarctica subclades identified by Vogler et al. and Svensson et al. [15, 16] (See additional file 1 for an update of these SNP positions based on the latest SCHU S4 genome NC_006570). Subclades within the B.Br.013 group are depicted in red. The Georgian isolate was placed in the basal node B.Br.013/020/023 (black arrow). (B) Maximum parsimony SNP phylogeny of four F. tularensis whole genome sequences from the B.Br.013 group. The Georgian strain

is highlighted in gray and is basal to the other three genomes. Newly identified branches (B.Br.027 and B.Br.026) are colored red and showed two major divisions within the B.Br.013 group. This phylogeny was rooted using OSU18 (not depicted). Bootstrap values are based on 1000 Ku-0059436 molecular weight replicates in PAUP using a heuristic search. Additional analyses of the B.Br.013 group are crucial for fully understanding the phylogeography of F. tularensis subsp. holarctica in Europe and Asia. This group contains significant genetic diversity based upon multi-locus variable-number

tandem repeat (VNTR) analysis (MLVA) [15], indicating that considerable phylogenetic structure may exist that could be revealed with additional analyses. In addition, this group is widely distributed, extending from Eastern Europe into the border regions of the European/Asian continents. Importantly, the eastern geographic extent of the B.Br.013 group is very poorly understood. This is because, to date, it has not been possible to place F. tularensis isolates from countries at the Phospholipase D1 boundary of the European/Asian continents and Western Asia, including Georgia, into a larger phylogeographic context. Based on growth characteristics, biochemical analyses, MAPK Inhibitor Library datasheet basic PCR methods, and DNA sequencing, we know that F. tularensis subsp. holarctica is the predominant subspecies in Georgia and in regions further east [11, 19–21], but more HDAC activity assay specific genetic information is limited.

Some isolates from the European/Asian juncture regions and East Asia have been genotyped with a subset of VNTRs but have not been part of any global analyses [10, 22, 23]. Although valuable for regional studies, homoplasy associated with these rapidly-evolving markers restricts their value for global phylogenetic analyses [24]. In this study, we determined the phylogenetic structure of F. tularensis subsp. holarctica isolates from the European/Asian juncture country of Georgia by sequencing the genome of a Georgian isolate, comparing that genome to other available whole genome sequences to discover SNPs, and screening a subset of the resulting SNPs across 25 isolates from Georgia. We examined diversity within the subclades defined by these SNPs using a multiple-locus variable number tandem repeat analysis (MLVA) system [25]. To place the Georgian isolates into an existing global phylogeographic framework [15], we also screened a canonical subset of the newly discovered SNPs across a large panel of European isolates belonging to the B.Br.013 group.

Discussion Cooked meat medium was developed by Robertson [18] in

Discussion Cooked meat medium was developed by Robertson [18] in 1916 for use in the cultivation of certain anaerobes isolated from Apoptosis inhibitor wounds. The present formulation for CMM is a modification of

Robertson’s original formula. Cooked Meat Medium is still widely used for the cultivation and maintenance of clostridia and the medium is recommended for use in the enumeration and identification of Clostridium perfringens from food [21]. Cooked Meat Medium provides a favorable environment for the growth of C. perfringens, since the muscle protein in the heart tissue granules is a source of amino acids and other nutrients. The muscle tissue also provides reducing substances, particularly glutathione, which permits the growth of strict anaerobes [22]. The Staurosporine combination of 2-DE and MS has clearly identified major proteins over-expressed in cells of C. perfringens ATCC13124 when grown on CMM. We have identified eleven prominent proteins showing over expression JAK inhibitor CMM grown whole cell proteome of C. perfringens ATC13124 cells (see Additional file 1, Figure 1). For a bacterial protein

to be considered as a candidate vaccine antigen, it should preferably be conserved (i.e. present in all strains), secreted or surface localized, and immunogenic (i.e. capable of stimulating the immune system). Ornithine carbamoyltransferase (cOTC) was an abundant protein up-regulated in CMM-grown cells. It was also identified as an immunogenic surface protein of this bacterium (spot SP15) (see Additional file 1 and 5, Figure 3). In another study, ornithine carbamoyltransferase has been isolated as putative adhesin from surface

molecule preparation of Staphylococcus epidermidis [23]. cOTC is a bonafied cell wall protein of Streptococcus agalactiae [24], S. pyogenes [25], S. sanguis [26], and S. suis [27]. Taken together, this makes cOTC a putative vaccine candidate against C. perfringens infection. Similarly, cystathionine beta-lyase (spot CMM4) that was over-expressed in CMM-grown cells of C. perfringens, has been previously shown as a dominant cell surface protein of the next bacterium, indicating a possible role of this protein in pathogenesis and a potential as putative vaccine candidate. Electron transfer flavoprotein, over-expressed in CMM grown cells has been recognized in earlier studies as cross reactive protein of C. tetani when probed with mouse anti C. perfringens (heat killed organism) polyclonal serum [28] and also as an extracellular protein in Bacillus anthracis [29] and Mycobacterium tuberculosis [30]. Antibodies from animals surviving gas gangrene infection recognized proteins from both TPYG and CMM grown cells of C.