4d) These results demonstrated that

heat-killed MoLac-1

4d). These results demonstrated that

heat-killed MoLac-1 induced IFN-γ production by NK cells via IL-12 secretion from macrophages and activated NK cells in vitro. Oral administration of LAB has been reported to augment NK activity in mouse and clinical studies (Ogawa et al., 2006; Takeda et al., 2006; Koizumi et al., 2008). Oral administration of heat-killed MoLac-1 increased the population of NK cells in the spleen, but did not affect the expression of early activation marker CD69 on NK cells PD0325901 (Fig. 6). Takagi et al. (2001) suggested that the enhancement of NK activity in splenocytes from LAB-fed mice was caused by the increased proportion of NK cells but not by the increased cytotoxicity of individual NK cells. Thus, oral administration of MoLac-1 might enhance

NK activity by increasing the population of NK cells, but further investigation such as functional assay of NK cells is needed for evaluating the possible effect on the activity of NK cells. NK cells and IFN-γ produced by NK cells are crucial to the early natural defenses against IFV infection (Stein-Streilein & Guffee, 1986; Monteiro et al., 1998). As in vitro studies demonstrated that heat-killed MoLac-1 cells induced the production of IFN-γ by NK cells, see more the in vitro immunomodulating effects of MoLac-1 might be associated with the alleviation of IFV infection; however, involvement of NK cells in the anti-infective effects of MoLac-1 is not clear and further investigation is needed. Substantial interest has been aroused in the application of nonviable microorganisms in food or food supplements. At first, the use of nonviable microorganisms could solve the problem concerning Progesterone the stability of active constituents in handling and preservation, and could prolong the shelf life of the products. Furthermore, nonviable microorganisms could eliminate the risks of microbial translocation, invasion, and toxin production (Taverniti & Guglielmetti, 2011). Concerns have been raised for safety aspects in the application of live bacteria in food or food supplements

(Wassenaar & Klein, 2008). In summary, we demonstrated that heat-killed MoLac-1 would have the potential to modulate innate immunity and might be useful for alleviation of symptoms of IFV infection. This strain was found to induce dose dependently IL-12p40 production by human PBMCs (data not shown). Further studies are anticipated to assess the usefulness of heat-killed MoLac-1 in clinical experiments. “
“OTHER ARTICLES PUBLISHED IN THIS MINI-REVIEW SERIES ON Th17 CELLS Function and regulation of human T helper 17 cells in health and disease. Clin Exp Immunol 2009; doi:10.1111/j.1365-2249.2009.04037.x Are T helper 17 cells really pathogenic in autoimmunity? Clin Exp Immunol 2009; doi:10.1111/j.1365-2249.2009.04039.x CD4+ T helper cells: functional plasticity and differential sensitivity to regulatory T cell-mediated regulation.

Biochemically he was under-dialysed with a urea of 28 mmol/L and

Biochemically he was under-dialysed with a urea of 28 mmol/L and creatinine 1180 μmol/L. Hypertension had been complicated by severe left ventricular hypertrophy, diastolic dysfunction and moderate pulmonary hypertension. Other comorbidities were renal osteodystrophy and renal anaemia. Previous liver biopsies STI571 molecular weight and his hepatitis C viral loads by polymerase chain reaction suggested that this disease was quiescent with no evidence of cirrhosis. The donor was a 46-year-old, brain dead man.

There was a 5/6 HLA mismatch with a cold ischemic time of 15.5 hours. Serology showed cytomegalovirus donor and recipient positivity. Transplantation was planned with ‘standard’ induction therapy including basiliximab, methylprednisone, tacrolimus and mycophenolate mofetil. Standard prophylactic agents including valganciclovir, trimethoprim/sulfamethoxazole, pantoprazole and nystatin were also commenced. Hypertension was aggressively managed prior to transplant. The transplant surgery was complicated by donor kidney core biopsy-related haematuria and subscapular bleeding with blood pressure instability. Because of the likelihood of need for dialysis after transplant surgery, the surgeon opted to leave the Tenckoff catheter Selleck RG 7204 in situ.

Dialysis was not required. However, residual peritoneal fluid became infected with methicillin-resistant Staphylococcus aureus (MRSA). The infected Tenckoff catheter was removed 9 days after transplantation, and a 2 week course of intravenous vancomycin for MRSA peritonitis was completed. Immunosuppression was also switched from Mycophenolate to azathioprine in view of severe diarrhoea, and valganciclovir and bactrim were stopped secondary to leucopenia. Despite the intra- and postoperative complications, there was immediate and good graft function, with a discharge

creatinine on day 25 of 75 μmol/L. On week 7 after transplantation, a computed tomography (CT) scan with contrast was performed to investigate new onset abdominal cramps and diarrhoea. This showed a large perigraft collection with large check details volume ascites, peritoneal enhancement, and thickened small bowel loops. Percutaneous drainage of the collection and ascites revealed frank pus that cultured positive for MRSA. Abdominal drains were left on free drainage and antibiotics recommenced for MRSA peritonitis, but as a result of ongoing abdominal cramps and diarrhoea the patient returned to theatres for a laparotomy and abdominal washout. This showed that the intra-abdominal space and small bowel were covered with pus and loculations. There were organising fibrin bands throughout the small bowel. An extensive division of adhesions was performed, and a peritoneal biopsy obtained.

[16, 17, 25] Clearly new therapeutic strategies are required for

[16, 17, 25] Clearly new therapeutic strategies are required for this deadly disease. Such potential novel therapies can be better designed with comprehensive understanding of the mechanism of infection and its related host defence. Iron uptake from the host by

microorganisms is essential for the establishment and progression of infection since this element is required for the survival of living cells.[26] In a normal host, free iron is restricted by highly efficient iron sequesters such as transferrin, ferritin and lactoferrin.[26] Pathogens either devise strategies to obtain iron from the host by stripping iron from these sequesters (e.g. by siderophore production), or the tightly controlled free iron becomes more available in certain medical conditions. The unique susceptibility of certain patient populations to mucormycosis, but not to other pathogenic JAK inhibitor fungi, point to the importance of iron uptake in the pathogenesis of mucormycosis.[3, 23] These include, hyperglycaemic, DKA and other forms of Sorafenib molecular weight acidosis patients as well as deferoxamine-treated patients. All these patient categories suffer from elevated available serum iron. For example, the excessive glycosylation of proteins such as transferrin and ferritin, due to constant hyperglycaemia result in decreased iron affinity of these sequesters

which leads to the release of free ion in the blood stream and in cells.[27] Similarly, DKA and other forms of acidosis cause proton-mediated dissociation of iron from iron-sequestering proteins.[28] The increased levels of available iron enable enhanced growth of Mucorales in serum.[9, 28, 29] It is also known that DKA mice are more susceptible to mucormycosis infection than normal mice and iron chelation therapy using deferiprone or deferasirox protects DKA mice from mucormycosis.[29, 30] Subsequent studies confirmed the efficacy of deferasirox in treating experimental mucormycosis using the Drosophila fly model.[31] Patients with iron overload toxicity were used Thiamine-diphosphate kinase to be treated with the bacterial iron-siderophore, deferoxamine.

These patients were found to be extremely susceptible to deadly form of mucormycosis.[32-34] Subsequent studies demonstrated that although deferoxamine is an iron chelator from the perspective of the human host, Rhizopus spp. utilise ferrioxamine (the iron-rich form of deferoxamine) as a xeno-siderophore to obtain previously unavailable iron.[35, 36] It was found that ferrioxamine binds to a cell surface receptor on the surface of Rhizopus and through an energy dependent reductive step releases ferrous iron prior to transporting it across the fungal cell membrane without deferoxamine internalisation.[36] Subsequent studies demonstrated that reduction in the high-affinity iron permease FTR1 copies (Mucorales are multinucleated organisms) in R.

[8] Furthermore, there is an independent, graded increased risk o

[8] Furthermore, there is an independent, graded increased risk of death and cardiovascular (CV) events associated with reduced eGFR,[6] BMS907351 and this relationship is also seen in survivors of acute MI (AMI) and NSTE-ACS.[9-11] Medical management of ACS, which include STEMI and NSTE-ACS, and chronic stable CAD has been extensively studied in the general population leading to evidence-based national clinical practice guidelines.[7-9] There are RCTs that have firmly established roles for reperfusion and primary PCI, antiplatelet and anticoagulant therapies, beta-blocker therapy, and ACEi or ARB therapy for ACS in the

general population. In the majority of these trials patients with moderate-to-severe renal impairment have been excluded, leading to unanswered concerns about efficacy and safety, and consequently significant underuse

of these therapeutic options in CKD patients.[9-11] The aim of this guideline is to examine the benefits and harms of medical management, specifically reperfusion therapy, antiplatelet and anticoagulant therapies, beta-blocker therapy, and ACEi/ARB therapy (but excluding lipid-lowering therapy), of ACS and chronic stable CAD in patients with CKD, including the dialysis and transplant populations. The benefits examined are: The risk of MI and CV death in patients presenting https://www.selleckchem.com/products/BIBW2992.html with ACS, including the risk of coronary

restenosis in patients with an ACS undergoing a PCI and receiving associated antiplatelet and/or anticoagulant therapy. The risk of MI and CV death in patients with chronic stable CAD. The harms examined relate to serious adverse Adenosine events reported in the literature in relation to the aforementioned medical therapies. There is little high quality evidence regarding the management of ACS or chronic stable CAD in patients with CKD. The RCT data examining the therapeutic options for the medical management of ACS or chronic stable CAD are all taken from post-hoc analyses of RCTs from the general population where patients with CKD were identified based on serum creatinine and/or eGFR, and outcomes analysed. These limitations also apply to assessing harms of ACS therapies. Specifically with regards to harm of anticoagulant therapies, data have been extrapolated from trials using anticoagulants for non-cardiac indications. Prospective and retrospective registry data or observational cohorts provide a significant proportion of the evidence for ACS therapies. The management of ACS in the general population has been published in the extensive guidelines available.[7-9] These guidelines support the use of PCI in favour of thrombolysis without specifically including or excluding CKD patients.

(2010)

(2010) selleck demonstrating a significant reduction in intestinal pro-inflammatory TNF-α expression in synbiotic-treated patients. Moreover, the results from this investigation provide evidence to suggest that early treatment with synbiotic combination of probiotic La and prebiotic inulin can effectively prevent pathogen-induced intestinal inflammation

by affecting NF-κB and Smad 7 signaling within the intestinal epithelium. Prebiotics are known to help colonization of beneficial probiotics. While early administration of a synbiotic combination of probiotic La and prebiotic inulin attenuated the secretion and expression of pro-inflammatory cytokines and inflammation, supporting a potential indirect role of prebiotic inulin in regulating mucosal immune response

by modulating the colonic microbial communities. Our results are supported by previous observations showing that a diet supplemented with Fructooligosaccharides (FOS) and inulin can trigger and stimulate the gut mucosal immune system (Benyacoub et al., 2008). Our observations also are in line with the results of randomized controlled trials, which provide evidence to Selleckchem BAY 73-4506 suggest that synbiotic therapy can be more effective in the treatment IBD than therapies limited to probiotics or prebiotics (Fujimori et al., 2009; Macfarlane et al., 2009; Steed et al., 2010). In the current study, we found that prebiotic (inulin) treatment of young mice resulted in a reduction in fecal C. rodentium output after the bacterial infection (Fig. 2b and c). It was reported previously that feeding rats with an inulin-oligofructose diet resulted in reduced numbers of Salmonella Typhimurium in the content of ileum and cecum (Kleessen & Blaut, 2005). However, contradicting results have also been reported. Petersen et al. (2009) reported that BALB/c mice fed diets containing prebiotics (FOS or xylo-oligosaccharide) had significantly higher

numbers of S. Typhimurium, translocated into liver, spleen, and MLN compared with mice fed with control diet. In contrast, no increased translocation of S. Typhimurium was found in mice fed inulin (Petersen find more et al., 2009), in that same study. Nevertheless, most prebiotics and/or probiotics have not been shown to cause illness, but additional research is needed to determine the safety of prebiotics and probiotics in young children or people whose immune system is compromised. The observations showing an enhanced colonic TGF-β and IL-10 responses in mice with early synbiotic or probiotic treatments provided evidence to support the idea that these treatments may modulate gut mucosal inflammatory responses by promoting immunological regulatory mechanisms, which parallel results by Roller et al.

However, lung larvae are smaller at day 1 in WT FVB/N hosts and d

However, lung larvae are smaller at day 1 in WT FVB/N hosts and do not grow to the extent seen in the more permissive CBA/Ca host strain (77). Thus, IL-5 Tg

and WT FVB/N mice, which represent two quite different host models, are highly resistant in the early stages of primary N. brasiliensis infections. This is also analogous to the resistance seen during re-challenge of WT host strains susceptible to primary infections (69,75,76) and even with secondary exposure in the IL-5−/− and ΔdblGATA deletion mutant strains (69). In addition, for those larvae that are able to migrate to https://www.selleckchem.com/products/MLN8237.html the gut in resistant hosts, it is likely that damage mediated prior to arrival in the lungs render them less capable of maturation or colonization of the gut. Leucocytes are recruited into the skin within the first hour of a primary infection with N. brasiliensis

L3 (65), and this is initially dependent on activation of complement protein C3 via the alternative pathway and generation of the chemotactic factor C5a (75,78,79). The C5a receptor inhibitor PMX53 can inhibit both neutrophil and eosinophil recruitment in this model (75). C3 deposition on larvae and eosinophil recruitment and degranulation within the first 30 min of infection are reduced in complement factor B deficient/IL-5 Tg double mutant mice (75). Whilst C3 deposition on larvae is inhibited for at least 150 min in these animals, Doxorubicin concentration at this stage of the infection complement is no longer essential for leucocyte recruitment, adherence to larvae or degranulation nor for larval

aggregation (75). Larval escape from the skin is enhanced in mice deficient in either factor B or C3, but this does not occur when C1q is absent (75). However, complement-deficient/IL-5 Tg double-mutant mice have few intestinal worms at day 6 pi. and those Rucaparib purchase that are present produce almost no eggs. In addition, single-mutant mice deficient in complement proteins C1q, factor B or C3 are also highly resistant, with few parasites at either the lung or gut stage of secondary infections (75). These experiments, together with in vitro studies (78,79), show that although complement is important for leucocyte recruitment and attachment to N. brasiliensis larvae, even in the vital first few hours of infection, when larvae are attempting to escape from the skin, other factors can compensate. We investigated the possibility that eotaxin-1, a potent and largely eosinophil-specific chemotactic factor at sites of inflammation in the skin, lungs and gut (80–83), might compensate for loss of C5a activity. Eosinophil recruitment into the skin is diminished in both primary and secondary N. brasiliensis infections in eotaxin-1−/−/IL-5 Tg double mutants, but not in eotaxin−/− single mutants and is not essential for resistance (76). Experiments with multiple and simultaneous deletion of complement, eotaxin-1, eotaxin-2 and other chemokines or receptors are required.

Data analysis was performed with the softwares spss version 10 0

Data analysis was performed with the softwares spss version 10.0 (SPSS Inc., Chicago, IL, USA) and stata version 9.0 (StataCorp LP, College Station, TX, USA). In addition selleck inhibitor to the cut-off point of 1.5 that was originally recommended by the manufacturer of the GM Platelia kit, 1.0, 0.7 and 0.5 cut-off points were also used to calculate sensitivity, specificity, negative and positive predictive values. Calculations were made separately for single positive

values and at least two consecutive positive results (within 1 week) as well as classifying the data as proven plus probable cases or proven plus probable plus possible cases. A total of 83 hospitalisation episodes were included in the study; however, 25 episodes were excluded from analysis because of the death of the patients soon after their inclusion in the study (n = 8), neutropenia <10 days (n = 7), absence of neutropenia (n = 6), problems with the venous access route (n = 1) and short period of hospitalisation (n = 3). Fifty-eight hospitalisation

episodes in 45 patients were eligible for final analysis (Table 1). The underlying haematological malignancy was acute myeloblastic leukaemia in 35 patients, acute lymphoblastic leukaemia in six patients, chronic myelocytic leukaemia-blastic click here transformation in two patients, biphenotypical leukaemia in one patient and high-grade non-Hodgkin lymphoma in one patient. According to the EORTC-MSG case definitions, one patient had proven IA (sinopulmonary aspergillosis). The diagnosis was confirmed by the demonstration of invading hyphae in the necrotic specimen taken from the lateral BCKDHA wall of the nose. Probable IA was

diagnosed in four and possible IA was diagnosed in 20 episodes. Thirty-three episodes were defined as not having IA. Dyspnoea and cough were the leading complaints in proven and probable IA cases (Table 2). Bacteraemia was present in 21.2%, 30% and 60% of the episodes without IA, with possible IA and with probable/proven IA, respectively. One case of candidaemia and one case of disseminated fusariosis were identified, both of which did not have IA according to EORTC-MSG criteria. Aspergillus flavus was cultured from either blood, sputum or bronchoalveolar lavage in three episodes of three different patients, while Aspergillus fumigatus was cultured from bronchoalveolar lavage in two episodes of probable IA. Bronchoalveolar lavage could only be performed in nine episodes overall. At least one thoracic CT was performed in 36 episodes. CT was ordered by the ward staff when the patient had prolonged fever without a focus, pulmonary signs and symptoms or pathological findings on plain radiograms. Among the 22 episodes in which no thoracic CT was performed, 12 had prolonged fever and neutropenia despite broad-spectrum antimicrobial therapy. Although indicated theoretically, CT was not ordered in these episodes at the discretion of the ward staff.

tuberculosis Furthermore, the purified proteins were used for fu

tuberculosis. Furthermore, the purified proteins were used for functional

characterization in terms of immunogenicity in rabbits for induction of antigen-specific antibodies. find more Antigen-specificity and polyclonal nature of the antibodies were determined by testing the rabbit sera with recombinant proteins and overlapping synthetic peptides covering the sequence of each protein. Bacterial strains and plasmids.  The plasmid pGEM-T Easy (Promega corporation, Madison, WI, USA) was propagated in E. coli strain DH5αF’ (Gibco-BRL, Paisley, UK), and pGES-TH-1 was propagated in E. coli strain BL-21 (Novagen, Madison, WI, USA), as described previously [24, 26]. M. tuberculosis H37Rv was obtained from PCI-32765 nmr the American Type Culture Collection (Rockville, MD, USA) and served as the source of genomic DNA for the amplification and cloning of the mycobacterial genes. All DNA manipulations, plasmid isolations, restriction endonuclease digestions and transformations were carried out according to standard procedures, as described previously [24, 26]. Synthetic peptides.  Overlapping synthetic peptides (25-mers overlapping neighbouring peptides by 10 amino acids) covering

the sequence of Rv3874, Rv3875 and Rv3619c proteins were obtained commercially (Interactiva Biotechnologies GmbH, Ulm, Germany). The stock concentrations (5 mg/ml) of the peptides were prepared in normal saline (0.9%) by vigorous pipetting, and the working concentrations were prepared by further dilution in phosphate-buffered saline (PBS, pH 7.0), as described previously [27–29].

Oligonucleotide primers.  The gene-specific forward (F) and reverse (R) oligonucleotide primers for the amplification of full-length rv3874, rv3875 and rv3619c genes by polymerase Epothilone B (EPO906, Patupilone) chain reaction (PCR) were designed on the basis of nucleotide sequences of these genes in the M. tuberculosis genome [30]. Furthermore, each F and R primer contained additional sequences at 5′ end (bold face nucleotides), including a BamH I and a Hind III restriction site (bold face and underlined nucleotides), respectively, for efficient cloning of PCR-amplified DNA in the cloning and expression vectors. The DNA sequences of F and R primers for each gene are shown below: Rv3874 F 5′-AATCGGATCCATGGCAGAGATGAAGACCGATGCC-3′ Rv3874 R 5′-ACGTAAGCTTGAAGCCCATTTGCGAGGACAG-3′ Rv3875 F 5′-AATCGGATCCATGACAGAGCAGCAGTGGAATTTC -3′ Rv3875 R 5′-ACGTAAGCTTTGCGAACATCCCAGTGACGTT-3′ Rv3619c F 5′-AATCGGATCCATGACCATCAACTATCAATTCGGGGAC-3′ Rv3619c R 5′-ACGAAGCTTGGCCCAGCTGGAGCCGACGGCGCT-3 Cloning and expression of rv3874, rv3875 and rv3619c genes in E. coli.  The DNA corresponding to rv3874, rv3875 and rv3619c genes were amplified by PCR using the respective F and R primers and genomic DNA from M. tuberculosis as the template, as described previously [20], except that for the amplification of rv3619c, 1% dimethyl sulfoxide (DMSO) was also added to the reaction mixture.

In developing this anti-CVB3 antibody

detection system, w

In developing this anti-CVB3 antibody

detection system, we generated new peptide sequences that specifically recognize the anti-CVB3 antibody produced during viral infection. We selected the peptide sequences by predicting the antigenicity and hydrophobicity of regions of the whole sequence of the enterovirus capsid protein. We confirmed the antibody SCH727965 mw production induced by the synthesized peptides with a rabbit immunization test. The synthetic peptides significantly recognized the anti-CVB3 antibodies in immunized mouse serum. This system also succeeded in detecting anti-virus antibodies in the serum of a human patient with viral myocarditis. This assay is the first to use detection of CVB3-induced IgG antibodies in patient serum for diagnostic purposes. Selection of peptides was based on amino-acid sequences of the CVB3-H3 Woodruff variant strain (locus accession number U57056). FXR agonist The two peptides with strongest antigenicity and lowest hydrophobicity were selected, namely VP2 and VP1. These peptides were synthesized and rabbits (NZW, Orient Bio, Seoul, Korea) immunized with 500 µg of each of them with IFA three times every second week for 6 weeks. One week after the final immunization, the rabbits were killed to collect their sera and IgG antibody production measured by ELISA) All these steps were performed by Ab-Frontier (Seoul,

Korea). The HeLa cervical Methane monooxygenase cancer cell line was obtained from the American Type Culture Collection (ATCC, Rockville, MD, USA) and maintained in Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, CA, USA) supplemented with 10% heat-inactivated

FBS (Invitrogen). The H3 variant of CVB3, the Woodruff strain, was obtained from a cDNA copy. The viral titer was determined with a plaque assay in HeLa cells, as described previously [10, 11]. The cells were lysed in SDS sample buffer (25 mM Tris–HCl [pH 6.8], 2% w/v SDS, 10% glycerol, 50 mM dithiothreitol, 0.1% w/v bromophenol blue). Aliquots (10 µg) of total cell extracts were resolved on a 10% SDS–PAGE gel and transferred to a Hybond-ECL nitrocellulose membrane (Amersham, Buckinghamshire, UK). The membrane was blocked with 5% nonfat dried milk solution (in Tris-buffered saline) containing 0.1% Tween 20. The protein bands were probed with anti-VP2 and anti-VP1 sera, anti-enteroviral VP1 antibody (Novocastra, Newcastle, UK), and anti-GAPDH antibody. The bands were visualized with an enhanced chemiluminoscence kit (Thermo Scientific, Barrington, IL, USA) according to the manufacturer’s instructions [11]. Balb/c mice (5 weeks old, n = 20) were intraperitoneally infected with 2 × 103 plaque forming units of CVB3 [10-13]. Antisera were collected from three mice on each of Days 3, 7, 14, and 21 post-infection. The mice were anesthetized with 2% isoflurane, after which blood was collected from the carotid artery after decapitation.

Mixtures of opsonized Candida in mouse autologous serum (10%) wer

Mixtures of opsonized Candida in mouse autologous serum (10%) were added to 0.2 mL of macrophage suspension. The mixture was incubated for 30 min at 37°C. The percentage of phagocytosis was expressed as the percentage of phagocytosing macrophages in 200 cells counted using an optical microscope (15). Alveolar and peritoneal macrophages selleck inhibitor monolayers were prepared as described above. In order to determinate the influence of lactobacilli on the capacity of macrophages to produce cytokines, alveolar and peritoneal macrophages were challenged in vitro with heat-killed C.

albicans AV4 at a concentration of 107 cells/mL. After incubation at 37°C in 5% CO2, the supernatant was recovered and kept frozen until cytokine analyses.

IL-1β and TNF-α were determined using the corresponding mouse ELISA kits ABT-263 purchase (R & D Systems). In order to evaluate the influence of lactobacilli treatments on the immune response against C. albicans in vivo, challenges with pathogenic C. albicans AV4 were performed. Yeast cells were grown in Sabouraud broth at 37°C until the log phase was reached. The pathogens were harvested by centrifugation at 3600 g for 10 min at 4°C and washed three times with sterile PBS. Intraperitoneal challenge with C. albicans AV4 was performed on the day after the end of each Lactobacillus treatment (third or sixth days). The mice were challenged with injections of 200 μL of an inoculum containing 108 cells. For yeast cell counts in blood, liver and spleen, mice were killed on day 2 post-infection. The livers and spleens were excised, weighed and homogenized in 5 mL of sterile peptone water. The homogenates were diluted appropriately, plated in duplicate on Sabouraud agar and Phloretin incubated at 37°C. C. albicans colonies were counted and the results expressed as log10 CFU/g of organ or mL of blood. Intranasal challenge

with C. albicans AV4 was performed on the day after the end of each Lactobacillus treatment (third or sixth days). The mice were challenged nasally with the pathogen by dripping 25 μL of an inoculum containing 107 cells into each nostril. To facilitate migration of the inoculum to the alveoli, the mice were held in a head-up vertical position for 2 min. For yeast cell counts in lung and blood, mice were killed on day 2 post-infection. The lungs were excised, weighed and homogenized in 5 mL of sterile peptone water. The homogenates were diluted appropriately, plated in duplicate on Sabouraud agar and incubated at 37°C. The C. albicans colonies were counted and the results expressed as log10 CFU/g of organ or ml of blood. In order to evaluate innate immune responses after challenges, the concentrations of TNF-α and IFN-γ and the number of leukocytes and neutrophils were determined in BAL and peritoneal fluid according to techniques described in a previous report (15).