, 2003; Brooks et al , 2006) Brooks et al demonstrated that BB0

, 2003; Brooks et al., 2006). Brooks et al. demonstrated that BB0405 was both amphiphilic and surface exposed, as determined by TX-114 phase partitioning and proteinase K accessibility, respectively. Additionally, bb0405 encodes a putative signal peptide with a signal peptidase I cleavage site, further suggesting BB0405 is a surface-localized transmembrane-spanning OMP. Consistent with the combined data indicating

that BB0405 is a surface-exposed protein, specific anti-BB0405 antibodies were observed to be bactericidal in vitro (Brooks et al., 2006). The surface localization of BB0405 suggests that it could be an excellent candidate for future Lyme disease vaccine studies. Given that glycosaminoglycans Smad inhibitor (GAGs) are present on most eukaryotic cells and that B. burgdorferi can bind GAGs, B. burgdorferi likely exploits this activity to interact with several different cell types and tissues during the infectious process. The B. burgdorferi surface protein Bgp (Borrelia glycosaminoglycan-binding protein) is encoded by ORF bb0588 and has

been shown to bind the GAGs heparin and dermatan sulfate on the surface of mammalian cells (Parveen & Leong, 2000). Bgp is not only found as an outer surface membrane protein, but it also has been shown to be secreted from the borrelial cell (Parveen & Leong, 2000; Cluss et al., 2004). Recombinant Bgp can agglutinate erythrocytes Vismodegib concentration and inhibit the interaction of B. burgdorferi and mammalian cells (Parveen & Leong, 2000), which further suggests that Bgp plays an important role in cell adhesion. Interestingly, a Bgp null strain was not required for infection of SCID mice (Parveen et al., 2006); however, it was speculated that the lack of an Glutamate dehydrogenase observed phenotype in the animal studies was likely the result of B. burgdorferi expressing other GAG-binding proteins that compensated for the Bgp deficiency in these studies. The last two decades have led to the identification of several important proteins that are located on the outer surface of B. burgdorferi. Some have been shown to be bona fide virulence factors that are needed

for mammalian infection (e.g. OspC), while others have been utilized as human vaccine targets (e.g. OspA). As outlined in Fig. 1, some surface proteins that have been identified are specifically expressed in the tick (e.g. OspA, OspB, CspA), while others are upregulated during tick feeding and transmission to the mammalian host (e.g. OspC, OspE, OspF, P66). Studies have also shown that surface-exposed lipoproteins, such as OspA, OspB, OspC, OspD, OspE, and OspF, are not only localized to the cell surface but can also be detected in the periplasmic space (Fig. 1), which is likely true of other surface-exposed lipoproteins. The differential expression of surface proteins is important in the parasitic strategy of B.

interdigitale grew in culture and also identified a dermatophyte

interdigitale grew in culture and also identified a dermatophyte species in an additional 32 specimens that were negative in selleck products microscopy and culture. None of the investigated non-dermatophytic strains was positive. Sensitivity of MX PCR was higher as compared to mycological examination (97% vs. 81.1%). MX PCR for direct detection of dermatophytes from nail samples yielded mixed flora in 32.8% of samples. MX PCR proved sensitive and adequate for the diagnosis of dermatophytic onychomycosis. It is much adapted to cases where culture is negative or contaminated by overgrowing moulds, which makes

the identification of the causal agent problematic. Onychomycosis is the most common nail disease. It Wee1 inhibitor is mainly caused by dermatophytes of the genus Trichophyton. However, various non-dermatophyte filamentous fungi are often isolated from nails and usually considered as transient contaminants and are not the actual aetiological agent. Treatment of onychomycosis is closely linked to the identity of the causative agent, particularly in terms of whether or not it is a dermatophyte.[1] Onychomycosis is routinely diagnosed by mycological examination, which includes direct examination and culture.[2] Direct microscopic examination of infected nail material may give false positive results as it is usually enable to differentiate between dermatophyte and non-dermatophyte filamentous

fungi. Conventionally the definitive diagnosis is based on culture isolation, but culture of toenails is often associated with contaminants. Furthermore, morphological and physiological Oxalosuccinic acid characteristics may vary; the phenotypic features can be influenced by factors such as temperature variation and medium.[3] Routine identification by microscopy and culture requires considerable training of personnel and considerable supervisory expertise. Molecular approaches have been developed to provide more rapid and accurate alternatives for dermatophyte

identification. These methods include restriction fragment length polymorphism analysis RFLP,[1, 4, 5] PCR-ELISA,[6] double-round PCR,[7] nested PCR,[8, 9] real-time PCR,[10, 11] PCR using (GACA)4 primer,[12] sequencing.[13, 14] The main targets have been the following genes or DNA fragments: the ribosomal DNA region,[1, 3, 10, 13, 15, 16] DNA topoisomerase II genes,[6] actin gene[7] and the chitin synthase gene.[8, 14, 17, 18] In recent years, the multiplex (MX) PCR assay has been used to identify a great variety of fungi including dermatophytes.[15-21] However, Trichophyton mentagrophytes complex was not included in these studies despite its high frequency in nail infection.[14, 22, 23] The primary aim of this study was to design and develop a MX PCR assay to identify dermatophytes on one hand and T. rubrum and T. mentagrophytes complex on the other hand directly from nails samples.

A practical consequence of these observations for a long-term ant

A practical consequence of these observations for a long-term antimalarial strategy is that drug targets should be encoded by genes located in cold spots rather than hot spots. Genome-wide proteomic analyses have generated a high number

of potential new vaccine candidates. Several new parasite surface antigens have recently been discovered throughout the malaria parasite life cycle (33–35,38,39). The availability of the P. falciparum genome has also allowed the development of new genome-wide learn more protein microarrays to probe human plasma from individuals before and after malaria season. These novel genome-wide methods have already delivered important insights into parasite proteins associated with immunoreactivity in an unbiased manner (99–101). It is highly probable that these studies will

soon improve our understanding of the molecular basis of protective immunity and facilitates the discovery of new efficient vaccine strategies. All together, the increasing number and performances of genome-wide technologies is transforming the scientific field. Genomics and systems biological studies have already contributed significantly to a better understanding of the malaria parasite’s biology. Most importantly, they have generated an exceptional pipeline of new drugs targets and vaccine candidates. The challenge today will be to bring these achievements to efficient and affordable antimalarial products. Constantly diminishing costs of high-throughput selleck chemicals llc genomics and DNA sequencing technologies have dramatically changed the way science is being done over the past few years. These changes should soon transform the way we assess genetic risk factors and the way we think about medicine, treatments and possible disease eradication in developing countries. Genomics has already greatly contributed to Thalidomide our understanding of the malaria parasite and the human genetic factors that influence the susceptibility and the response to both malaria

and antimalarial drugs/vaccines. The full integration of the newly acquired knowledge to the disease strategy will undoubtedly provide bases to prevent the resurgence of malaria [e.g. Peru (95)] and the arising and spread of resistances by analysing parasites’ population dynamics and evolution (e.g. resistances to artemisinin in south-east Asia). The catalogue of putative drugs and drug targets has already increased together with the panel of candidates for vaccination strategies. Beyond drug discovery, genomics was recently proven to be particularly efficient in the discovery of a drug mechanism of action within a 2-year time span by coupling drug screening and genomics (97). Ultimately, diagnostic and curative treatment could be improved by genotyping both the host and the infecting parasite. Such optimized treatment would contribute to a better use of drugs and a better management of the spread of resistances.

56–60 In contrast to HLA-B, some HLA-A, -C, and -DRB1 alleles are

56–60 In contrast to HLA-B, some HLA-A, -C, and -DRB1 alleles are common over very large areas of the world, whereas others enjoy high frequencies only in specific regions. For example, the HLA-A*23:01 allele is one of the FMF alleles in African [Sub-Saharan Africa (SSA) and North Africa (NAF)] populations, but not selleck compound in other populations, while A*02:01/*02:01:01G is one of the FMF alleles in all regions but Oceania (OCE), where it is ranked fifth (data not shown). Similarly, HLA-C*07:01G is one of the FMF alleles in Africa, Europe (EUR), and Southwest Asia (SWA), while *07:02G is one of

the FMF alleles in EUR, Southeast Asia (SEA), OCE, Northeast Asia (NEA), and the Americas [North America (NAM) and South America (SAM)]. At the DRB1 locus, DRB1*11:01 is one of the FMF alleles in SSA, SWA and OCE, and *15:01 is one of the FMF alleles in NAF, EUR, SWA, OCE and NEA. Based on their CAFs, the FMF alleles at these loci represent 40–70% of the allelic diversity in each region. Patterns of allelic diversity at the class I and DRB1 loci differ considerably from those at DQA1, DQB1, DPA1 and DPB1. At the latter loci, a small number of alleles are observed

at high frequencies all over the world (resulting in most cases, at least for DPB1, in ‘L-shaped’ rather than even frequency distributions). The DQA1*03:01/*03:01:01G and *05:01/*05:01:01G alleles are two of the FMF alleles in all regions; the DQB1*0301/*03:01:01G allele is one of the Venetoclax manufacturer FMF alleles in all regions; DPA1*01:03, *02:01, and *02:02 are three of the FMF alleles in all surveyed regions (and are the only DPA1 alleles observed in SAM); and

the DPB1*04:01 and Histidine ammonia-lyase *04:02G alleles are one or two of the FMF alleles in all regions. Moreover, based on their CAFs, the FMF alleles at these loci represent 60–90% of the allelic diversity in each region. The trends observed for the DQ and DP loci contrast markedly with those for the DRB1 locus, and the differences may reflect divergent strategies of class II allelic diversification. Although there is low diversity in the genes that encode the α and β subunits of the DQ and DP proteins, a population may display greater diversity of heterodimeric DQ and DP proteins than DR proteins because the DQ and DP heterodimers may be encoded both in the cis and the trans positions of their genes (although for DQA1 and DQB1, particular combinations form unstable dimers61,62). As there is much less variation of the DRA gene, this may be driving DRB1 to diversify in a manner more similar to the class I loci. Despite evidence of natural selection acting on the evolution of the HLA polymorphism, as discussed above, this immunogenetic system is highly informative for anthropological studies, as the patterns of HLA genetic variation reveal spatial and demographic human populations expansions that occurred in the past.

As BAFF is able to induce CSR, the intestinal immunoglobulins may

As BAFF is able to induce CSR, the intestinal immunoglobulins may well be of another isotype than IgE. The results indicate that BAFF might be particularly involved in non-IgE-mediated reactions. Determination of BAFF levels in different body fluids, as in gut lavage fluid in our study, thus

supports the notion that BAFF is produced locally in different compartments of the body, not only in joints and airways but also in the gut, in response to inflammation and allergic reactions. In addition, our study raises the possibility that Selleckchem AZD4547 delayed-type hypersensitivity reactions to food may result from a unique immunoglobulin class switching in the intestine. Enhanced BAFF expression has been noted in several viral infections such as in human immunodeficiency virus (HIV), Epstein–Barr virus (EBV) and hepatitis C virus (HCV) infections [45–47]. Studies in patients with HIV suggest that these patients have increased levels of BAFF and IL-10 in their serum, and BAFF concentration increased with disease progression [45, 46]. EBV-infected

B cells have been shown to express BAFF [4, 47]. In patients with HCV, increased BAFF levels in serum were associated with the presence of arthritis/arthralgia and/or vasculitis, and high values at onset of acute HCV infection can predict its evolution to chronic infection [48]. Another Nutlin-3 chemical structure significant association was found between increased serum BAFF levels and liver fibrosis in HCV-infected patients, showing that patients with cirrhosis have more BAFF expression than non-cirrhotic patients [49, 50]. B-cell expansion and lymphoproliferation are common features in patients chronically infected with HCV [51]. Induction of BAFF expression during HIV, EBV Suplatast tosilate and HCV infections may explain the connection

between viral infections and the occasional development of autoimmunity. Persistent viral infection may enhance cell apoptosis and the release of various nuclear antigens including heat shock proteins and the binding of toll-like receptors (TLRs) [52, 53]. Following such activation, dendritic cells become overactivated and increase their production of proinflammatory cytokines, one of which is BAFF, which may terminate B-cell tolerance and stimulate autoreactive B cells to produce autoantibodies. Neoplastic B cells express one or more of the receptors for BAFF on their surface, and impaired TACI upregulation contributes to hyperactive B cells and cancer development [3, 4]. In addition to autoimmune and allergic diseases, high BAFF levels were demonstrated in the serum of patients with B-cell chronic lymphocytic leukaemia (CLL), multiple myeloma and non-Hodgkin’s lymphoma [54–57]. One study showed that many patients had increased levels of BAFF on circulating CLL compared with non-transformed B cells [54]. In different types of non-Hodgkin’s lymphoma, BAFF concentrations were at least threefold higher in serum of patients with follicular lymphoma [56, 58].

We found that the numbers of CXCR3-expressing FOXP3+ Tregs increa

We found that the numbers of CXCR3-expressing FOXP3+ Tregs increased over the first 6–12 months post transplantation in two of these patients, (Fig. 8C). In the other two, expression remained at low levels. Although these results are observational, the two patients with higher numbers

of CXCR3-expressing Tregs had excellent renal function at 24 and 36 months Selleck HKI-272 post transplantation. In contrast, both patients with low levels of CXCR3 on circulating Tregs had acute rejection episodes within the first post transplant year, and one patient developed graft failure by 24 months post transplantation. Thus, kidney transplant recipients treated with mTOR-inhibitor therapy have circulating CXCR3-expressing Tregs. It will be selleckchem intriguing to determine whether the patterns of expression seen in this small cohort of patients are associated with differences in long-term graft outcome. In this report, we demonstrate that CXCR3 is expressed on human FOXP3+CD4+ T-cell subsets, and that CXCR3hiCD4+ Treg subsets function as potent immunoregulatory

cells to suppress allogeneic and mitogen-induced effector T-cell activation in vitro. We also find that CXCR3+ Tregs migrate toward their chemokine ligand IP-10, and their directional persistence and chemotaxis response is significantly greater than that of CXCR3neg Tregs. We interpret these observations to suggest that the expression of CXCR3 on Tregs may facilitate their accumulation at sites of inflammation including allografts undergoing

rejection. Understanding the compartmentalization and migration of Tregs is an area of intense study, and is likely of great importance for tolerance induction following solid organ transplantation 16–18. Tregs are well established to express both Aspartate adhesion and chemokine receptors 20, 22, 23, and they have potential to suppress anti-donor immune responses following transplantation 16–18. The trafficking of Tregs into secondary lymphoid organs as well as into the periphery has been proposed to be important for alloimmune tolerance induction 16, 18, and for the prevention of chronic rejection 17. Indeed, recently, it was observed that effective immunoregulation in vivo was not achieved in the absence of defined patterns of migration 18. In these studies, we found that greater than 80% of human Tregs express the lymph node homing receptor CD62L. Also, consistent with others 22, 24, 25, we find that CXCR3+FOXP3+ Tregs co-express the peripheral homing receptors CCR4 and CCR5. However, we also find notable differences in the expression of additional homing receptors on Tregs versus T effector cells including α-integrins, β-integrins and PSGL-1 (p<0.01, p<0.05 and p<0.01 respectively, data not shown), further indicating the potential that human Tregs have potential to traffic to lymph nodes as well as to peripheral sites of inflammation, as observed in mouse models 16–18.

3a) Next, we examined several cell surface markers of MLN B cell

3a). Next, we examined several cell surface markers of MLN B cells isolated from 15-week-old SAMP1/Yit mice by flow cytometry. As shown in Fig. 3(b), there were no differences between cell surface markers from SAMP1/Yit Linsitinib in vitro and AKR/J mice. In addition, the expression patterns of MLN B cells in these mice were similar to those in BALB/c mice. To know whether innate immune

responses by MLN B cells are associated with the pathogenesis of ileitis that develops in SAMP1/Yit mice, we examined the production of IL-10 and TGF-β1 by TLR-mediated MLN B cells isolated from SAMP1/Yit and AKR/J mice. To achieve this, at first the surface phenotypes of the sorted B cells were checked by their presence of the commonly encountered markers CD19, CD20, B220 and PDCA-1 (Fig. 4a). The CpG-DNA induced production of IL-10 by MLN B cells from all age groups of SAMP1/Yit mice, which were significantly lower than those from AKR/J mice (Fig. 4b). Selleck IWR1 Interleukin-10 production in response to CpG-DNA was markedly higher than that in response to LPS. Although lower production of TGF-β1 after stimulation with TLR ligands was observed in all samples tested, CpG-DNA significantly induced TGF-β1 production by MLN B cells isolated from 15- and 30-week-old AKR/J mice (Fig. 4b). Interleukin-10 is expressed not only by regulatory

B cells, but also by the monocytes and type 2 helper T cells (Th2), mast cells, regulatory T cells, and in a Sclareol certain subset of activated T cells. Similarly, TGF-β1 has also been produced by a wide variety of cells to generate diverse immune-regulatory phenotypes. We therefore aimed to carry out experiments to estimate IL-10 and TGF-β1 contents

in purified T cells after stimulation with LPS and CpG-DNA. To achieve this, MLN T cells from SAMP1/Yit and AKR/J mice were isolated using CD90.1 microbeads. According to our findings, in contrast to regulatory B cells (Fig. 4b), sorted T cells from both SAMP1/Yit and AKR/J mice produced very small quantities of IL-10 and TGF-β1 in both LPS-treated and CpG-DNA-treated conditions (Fig. 4c), which we think was a result of their weak innate immune responses when stimulated with those TLR ligands. In light of these findings, we conclude that the regulatory B cells produced copious amount of IL-10 and TGF-β1 which may generate immune modulating role during intestinal inflammation. In terms of logistics, one important point is that stimulation with antigens or TLR ligands may sometimes induce apoptosis or immune tolerance in B cells. To address this, we duly checked B-cell apoptosis status in our system after stimulation with TLR ligands LPS and CpG-DNA and observed that an insignificant portion of B-cell population can undergo apoptosis upon LPS and CpG-DNA stimulation (data not shown). Beside these, we also assessed B-cell activation upon TLR stimulation by screening the B-cell activation marker CD25 in isolated B220+ cells from both AKR/J and SAMP1/Yit mice.

Psoriasis is mediated by T cells that trigger keratinocytes to hy

Psoriasis is mediated by T cells that trigger keratinocytes to hyperproliferate and perpetuate the disease [9]. T helper (h)17 and Th1 cells and the cytokines produced by these cells are found in increased levels within psoriasis plaques [10] as well as in the circulation [11] and are thought to have an important role in psoriatic inflammation. The relationship between Th1 and Th17 cells is still unclear. The tissue-specific

localization of T cells is thought to be guided by the skin-homing molecules such as cutaneous lymphocyte-associated antigen (CLA), various chemoattractants and their receptors, including chemokine receptors 4 (CCR4) and 10 (CCR10) [12]. In addition, adhesion molecules are thought to mediate T cell migration and retention in cutaneous tissue, such as the αE (CD103) β7 integrin that is overexpressed Nutlin-3a solubility dmso in psoriasis skin [13]. The main objective of this study was to evaluate the immunological therapeutic effect

of two treatment protocols on psoriasis, Selleck GSK2126458 focusing on the main inflammatory cytokines and effector T cell phenotypes known to be important for skin homing and tissue retention, thus potentially providing new insight into the immunopathogenesis of psoriasis. Our results confirm the role of Th1 and Th17 effector T cells in psoriasis. It also provides insight into the role of CD8+ T cell secreting IFN-γ (Tc1) and IL-17 (Tc17) and CLA+/CD103+ effector T cells in its immunopathology. The Icelandic National Bioethics Committee (Nr. 08-010-S1) and the Icelandic Data Protection Authority approved the study. After providing informed consent, twelve patients with plaque psoriasis entered the study. They were assessed at baseline (W0), one (W1), three (W3) and Rapamycin research buy eight (W8) weeks after starting treatment. Disease severity was assessed by the same physician (J.H.E.) at each time point

with Psoriasis Area and Severity Index (PASI) [14] score and photographic documentation, and punch biopsies and blood samples were obtained. Eligible patients were recruited to the study from January to May 2008. They were referred by dermatologists, and they were randomly assigned to two treatment groups. Patients were excluded if they had other forms of psoriasis, had other skin diseases or had received systemic psoriasis therapy, phototherapy or topical treatment within the previous 4 weeks. Of the 12 patients enrolled, six received inpatient treatment at the BL clinic for two weeks and 6 were treated with NB-UVB therapy three times weekly for 8 weeks. Psoriasis treatment at the BL clinic included bathing in geothermal seawater twice daily for at least 1 h combined with NB-UVB therapy 5 days per week for 2 weeks. After treatment at the BL clinic, patients used moisturizing creams for 6 weeks.

Because the Th1-dominated IFN-γ-producing CD4+ T-cell response in

Because the Th1-dominated IFN-γ-producing CD4+ T-cell response in control B6 mice is replaced by a Th17-dominated IL-17-producing CD4+ T-cell response in mice with combined defects in IL-12 and type I IFN

receptor,30 the relative production of IL-17 and IFN-γ by L. monocytogenes-specific CD4+ T cells in mice with combined defects in IL-21, IL-12 and type I IFN receptor (TKO) was compared with that in DKO mice, IL-21-deficient mice and control B6 mice (Fig. 4). Surprisingly, the additive effect of IL-21 deficiency in mice with combined defects in IL-12 and type I IFN receptor not only did not ablate, but accentuated IL-17 production after stimulation with the L. monocytogenes-specific I-Ab class II peptide LLO189–201 (Fig. 4a,b). Importantly, increased IL-17 production by L. monocytogenes-specific CD4+ CH5424802 manufacturer T cells, which occurs with IL-21 deficiency,

was not restricted only to mice with combined defects in IL-12 and type I IFN receptor because despite sharp reductions in the magnitude of IL-17-producing CD4+ T cells, a similar twofold increase in percentage and total number of IL-17-producing L. monocytogenes-specific CD4+ T cells was found for IL-21-deficient mice compared with B6 control mice (Fig. 4a,b). Interestingly, despite the increased production of IL-17 that occurs in the absence of IL-21, the percentage and absolute numbers of IFN-γ-producing Lenvatinib solubility dmso CD4+ T cells were not reciprocally reduced in IL-21-deficient compared with control B6 mice (Fig. 4c). Taken together, these results indicate that IL-21, IL-12 and type I IFNs synergize and play additive inhibitory roles in the differentiation of L. monocytogenes-specific IL-17-producing CD4+ T cells. Interleukin-21 therefore plays dramatically opposing roles in Th17 CD4+ T-cell differentiation under infective and non-infective conditions.

To identify the individual and collective roles of IL-21, IL-12 and type I IFNs in priming protective immunity to secondary L. monocytogenes infection, the susceptibility to re-challenge with virulent L. monocytogenes was enumerated for each tuclazepam group of mice. Thirty days after primary L. monocytogenesΔactA inoculation, groups of B6, IL-21-deficient, DKO and TKO mice were each challenged with 105 CFUs of virulent Lm-OVA.30,32 Compared with naive mice, L. monocytogenesΔactA-primed mice in each group were uniformly highly protected, and by day 3 after re-challenge contained four to five log10 reductions in recoverable L. monocytogenes CFUs (Fig. 5a). Moreover, by day 5 after re-challenge, virulent L. monocytogenes was cleared from both the spleen and liver in L. monocytogenesΔactA-primed mice in each group. The marked reductions in bacterial burden after re-challenge in L. monocytogenesΔactA-primed compared with naive mice in each group was associated with robust secondary expansion of L.

015) Furthermore, a similar expression was detected on neutrophi

015). Furthermore, a similar expression was detected on neutrophils incubated with chamber fluid and 100 ng/ml IL-8, and both had a significantly higher expression compared with cells incubated with cell culturing medium alone (P < 0.01). Figure 4 views the correlation between the concentration of IL-8 in the chamber fluid and the percentage of neutrophils that expressed the CD11b activation epitope following incubation with the same chamber fluid, at P < 0.05 and R = 0.72. Statistically significant correlations to other mediators in the

chamber fluid were not present. Peripheral leucocytes from three healthy study subjects were incubated with recombinant IL-8 in concentrations corresponding to serum and chamber fluid. The expression of CD11b activation epitope on IL-8-activated Doxorubicin neutrophils Veliparib datasheet is presented in Fig. 5, which display a dose-dependent expression of the CD11b activation epitope at P < 0.05 and R = 0.79, assessed by Spearman’s rank order analysis. In the present article, we demonstrate the induction of a variety of inflammatory mediators in a skin chamber and the

physiological effect of the microenvironment on neutrophil function. Moreover, we report a correlation between IL-8 and the expression of CD11b activation epitope, which may account for correlations between IL-8 and neutrophil transmigration. During the onset of inflammation, inflammatory mediators are produced by resident cells, and after a few hours, extravasated leucocytes make significant contributions to the inflammatory milieu. The diverse contribution by different cell types is reflected by the mixture of mediators that are released during the incubation. Pro- and anti-inflammatory cytokines such as IL-1, IL-4, IL-6, IL-7, IL-10, IL-12, TNF and interferon (IFN) were significantly induced along with growth factors such as granulocyte

colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF), as well as chemokines such as IL-8, MCP and MIP. The current results are comparable with the results by Kuhns Bacterial neuraminidase et al. [2] that demonstrated a dynamic production of inflammatory mediators in a skin chamber. In the former publication by Kuhns et al., following 8 h of incubation, IL-1β, IL-6, IL-8, TNF-α and GM-CSF were produced at comparable or slightly lower concentrations, which might reflect the use of 70% serum instead of 100% as in the current article, as well as the shorter time span between blister induction and application of the skin chamber. Interestingly, many of the assessed mediators in the present study are associated with lymphocyte differentiation and activation, despite that very few lymphocytes were detected in the skin chamber after 10 h of incubation.