43,44 (2) Enhanced peroxisomal FAO, the first enzymatic step of w

43,44 (2) Enhanced peroxisomal FAO, the first enzymatic step of which produces H2O2.5,45 (3) Higher mtFAO, which is also able to generate ROS, possibly at the level of electron transfer flavoprotein-ubiquinone oxidoreductase,46,47 or downstream within the MRC.5 Reduced ROS detoxification could also favor oxidative stress. Indeed, several studies showed lower GSH levels in NAFLD, including within the mitochondria.48-51 Decreased expression and/or activity of antioxidant

enzymes such as GPx and SODs could also occur, in particular at the mitochondrial level.48,52-55 NAFLD has been associated with higher hepatic expression of the inducible nitric oxide (NO) synthase (iNOS), mainly as a consequence of tumor necrosis factor-α

(TNF-α) overproduction by the Kupffer cells.56,57 Increased expression of the neuronal NOS (nNOS) could be also a significant source selleck of RNS.58 NO can readily react with the superoxide anion, thus generating the RNS peroxynitrite, which has deleterious effects on mitochondrial function and genome.20,58 Genetic susceptibilities RG-7388 ic50 could increase the risk of developing fatty liver, but also its progression to NASH in some individuals. Thus far, a polymorphism in the adiponutrin (PNPLA3) gene seems to be the most robust genetic determinant associated with fatty liver.59 Genetic polymorphisms affecting the mitochondrial ability to oxidize fat could also modulate the risk of NAFLD, in particular in genes encoding PPARα, leptin, adiponectin, or receptors of these adipokines.5,59,60 Polymorphisms in the TNF-α, transforming growth factor-β, and MnSOD genes have been shown to favor NASH.5,59,61 During fatty liver, several metabolic adaptations can restrain fat accretion (Fig. 2). A major mechanism is the stimulation of mitochondrial and peroxisomal FAO.5,6,10,62,63 Increased mitochondrial oxidation of FAs and other substrates could also be an adaptation to produce more ATP needed for DNL and gluconeogenesis.64 However, higher FAO can be associated

with different mitochondrial abnormalities, as discussed below (Table 1). Another important adaptation in fatty 上海皓元医药股份有限公司 liver could be an increased release of VLDL.65-67 Thanks to different noninvasive methods, increased hepatic mtFAO was found in patients with fatty liver, or in obese individuals with prodromal features of the metabolic syndrome.68-70 Importantly, higher fat oxidation seems to persist in patients with NASH,42,71,72 as discussed later on (Table 1). In rodents, enhanced mtFAO was found in fatty liver induced by overfeeding,64,73-79 or by monosodium L-glutamate.53 Hepatic ketogenesis was also augmented in mice fed a high-fat diet (HFD),80 thus suggesting that higher acetyl-CoA generation by way of mtFAO is followed by efficient KB production. However, during severe IR, excess acetyl-CoA could also enter the TCA cycle, in particular to serve as a carbon source for gluconeogenesis.

4C), we characterized these individuals using platelet counts and

4C), we characterized these individuals using platelet counts and APRI,[25] which are the readily available surrogate markers for liver fibrosis. We first determined the cutoff values of platelet counts and APRI for predicting HCC development by ROC analyses. Accordingly, platelet counts <150 × 103/μL and APRI >0.96 were identified as cutoff values, and the areas under the ROC curve for platelet counts and APRI were 0.715 (95% CI: 0.675-0.755) and 0.740 (95% CI: 0.701-0.779), respectively (Supporting Figure). Even in

individuals without advanced fibrosis (F1 and F2 patients), the proportion of patients with platelet counts <150 × 103/μL or APRI >0.96 was significantly higher in patients with HCC than in those without HCC (platelet counts, 53.0% [35/66] versus 31.3% [387/1238], P = 0.0002; APRI, 53.0% [35/66] versus 26.4% [325/1229], P < 0.0001). SB203580 supplier Moreover, GDC-0199 supplier the cumulative incidence of HCC development was significantly higher in patients with platelet counts <150 × 103/μL or APRI >0.96 in the subgroups without advanced fibrosis (Supporting Figure). Therefore, patients with low platelet counts or high APRI still have a substantial risk for HCC development even though they were diagnosed with mild fibrosis by liver biopsy. To characterize SVRs without ALT and AFP normalization after IFN therapy, we evaluated the percentage of severe hepatic

steatosis in these patients. Accordingly, the percentages of severe hepatic steatosis were significantly higher in SVRs without ALT and AFP normalization than in those with normal ALT and AFP (ALT, 37.9% [36/95] versus 13.8% [77/557], P < 0.0001; AFP, 31.6% [31/98] versus 14.8% [82/554], P < 0.0001). Therefore, it is likely that presence of hepatic steatosis is 上海皓元医药股份有限公司 associated with ALT and/or AFP elevation, and it is one of the risks for HCC development even after achieving SVR. This large-scale, long-term cohort study establishes important findings, which demonstrate a strict association between hepatocarcinogenesis and post-IFN

treatment ALT and AFP levels in patients with CHC. This association was notable in both SVR and non-SVR subgroups, and suppression of these values by IFN therapy reduced the hepatocarcinogenesis risk despite failure of HCV eradication. These data, which demonstrate the efficacy of IFN against HCC development associated with suppression of AFP, have clinically important implications for physicians. Although there have been reports on the association between baseline pretreatment AFP levels and HCC risk,[26-35] little is known regarding the effects of IFN therapy on change in post-IFN treatment AFP and its relation to HCC risk.[36] Although a previous report demonstrated that a decrease in AFP levels in patients receiving IFN therapy reduced the incidence of HCC,[37] this study was performed in a small number of patients (n = 382), and cutoff values, relation to ALT, or histological findings were not determined.

3 and Bettermann et al4 have identified transforming growth fact

3 and Bettermann et al.4 have identified transforming growth factor (TGF) β-activated kinase 1 (TAK1) as an essential inhibitor of this fatal series of events in hepatocytes. TAK1 belongs to the mitogen-activated protein kinase kinase kinase (MAP3K) family and acts as a mediator of stress, apoptosis, and inflammatory signals in the liver. Upon ligand activation, Toll-like receptor/interleukin-1 receptor (TLR/IL1R) and tumor necrosis factor receptor (TNFR) recruit and phosphorylate TAK1 through TNFR-associated

factors (TRAFs). Phosphorylated TAK1 activates IκB kinase (IKK) and MAP kinase kinase 4/7 (MKK4/7), leading to the activation of nuclear factor-κ B (NF-κB) and c-Jun N-terminal kinase (JNK), respectively. NF-κB and JNK are important for mounting an immune response and causing tissue inflammation. Moreover, the current belief is that NF-κB protects hepatocytes from death, whereas JNK promotes apoptosis. Consequently, NF-κB and JNK act as regulators of injury, this website death, proliferation, and dysplastic transformation of hepatocytes,5, 6 making any molecule that activates these pathways, such as TAK1, a potential therapeutic target. A role for TAK1 in the liver was first reported in 2001 in two studies from the Brenner group describing how TAK1 activated JNK and maintained hepatocyte quiescence7 and controlled the proliferation of stellate cells.8 One year later,

Liedtke et al.9 elucidated TAK1′s role in hepatocyte apoptosis by showing increased apoptosis after inhibition of the TAK1/JNK pathway. Because of the lack of mice with inactivation of Map3k7, the gene encoding TAK1, these studies relied on selleckchem adenoviruses to express dominant-negative sequences in mice. Despite these intriguing findings, TAK1 was one of the least studied MAP3Ks in the last 上海皓元医药股份有限公司 decade. However, the generation of mice carrying floxed Map3k7 alleles restored interest in this regulator.10 For example, Tang et al. crossed these mice with mice transgenic

for Mx1-Cre, an interferon-inducible Cre recombinase. Upon injection of the interferon inducer polyinosinic:polycytidylic acid, Cre-mediated recombination resulted in TAK1 deficiency mainly in hematopoietic cells, but also in hepatocytes.11 Surprisingly, this caused cholestasis, massive hepatocyte apoptosis, and destruction of the normal liver architecture followed by death from liver failure 8-10 days after polyinosinic:polycytidylic acid injection. These results provided a starting point for two studies published this year that examined the function of TAK1 specifically in the liver. The first study by Inokuchi et al. appeared in Proceedings of the National Academy of Sciences of the U.S.A.3 and was quickly followed by the study from Bettermann et al. published in Cancer Cell.4 Both studies focused on the role of TAK1 in HCC development. Inokuchi et al. crossed mice carrying floxed Map3k7 alleles with mice expressing Cre from an albumin enhancer/promoter construct (Table 1).

Lcn2 was also reported to be a biomarker in cholangiocarcinoma;[3

Lcn2 was also reported to be a biomarker in cholangiocarcinoma;[35] these findings suggest that the role of Lcn2 is dependent on the liver tumor with an epithelial phenotype. Our results are in good agreement with previous observations in cancer of the ovary, where Lcn2 expression is almost completely absent in tissue samples from normal ovaries, but strongly expressed in both borderline and grade

1 tumors, and weakly to moderately expressed in grade 2 and 3 tumors.[4] Furthermore, moderate to strong expression of Lcn2 was observed in the epithelial ovarian cancer cell lines SKOV3 and OVCA433, while no expression was detected in mesenchymal-like OVHS1, PEO36, or HEY cell lines. Taken together, Lcn2 expression appears to be linked find more to the epithelial phenotype of tumors and is lost as the tumor progresses and becomes undifferentiated. Lcn2 has been implicated in the induction of cellular proliferation because its expression is associated with a variety of proliferative cells.[36, 37] Lcn2 expression is required for Bcr-Abl-induced http://www.selleckchem.com/PARP.html tumorigenesis in leukemia

cells.[38] Lcn2 expression promotes breast tumor growth and progression,[39-41] and also increases colon cancer migration and invasion.[42] Furthermore, down-regulation of Lcn2 by antisense RNA suppresses human esophageal MCE carcinoma SHEEC cell invasion in vivo by reducing matrix metalloproteinase (MMP)−9 activity.[43] In the present study, Lcn2 suppressed the proliferation, migration, and invasion of EMT phenotypic HCC cells in vitro and tumor growth and metastasis in vivo. This is consistent with results

from a previous study that reported that Lcn2 suppressed Ras-transformed 4T1 mouse mammary tumor cell invasiveness in vitro and tumor growth and lung metastases in vivo.[9] Furthermore, Lcn2 blocked human colon cancer KM12SM cell invasion and liver metastasis.[10] A recent study also proposed that Lcn2 may act as a suppressor of invasion and angiogenesis in advanced pancreatic cancer cells.[11] These apparently conflicting observations could be due to distinct functions of Lcn2 in different cell types. Our focus in this study was to determine the mechanisms by which Lcn2 inhibits growth factor-mediated EMT in association with invasion and metastasis in HCC. Loss of E-cadherin expression has been associated with activation of the EGF/EGFR cascade in several cancer types, including pancreatic cancer and cervical cancer.[25, 27] EGF-induced EMT phenotypes were inhibited in the presence of AG1478, an inhibitor of EGF receptor tyrosine kinase activity. Lim et al.[4] reported that EGF down-regulated both E-cadherin and Lcn2 expression in ovarian cancer cells.

For in vitro experiments, 38 paired fresh tissues were used from

For in vitro experiments, 38 paired fresh tissues were used from HCC patients,

including 30 HBV+ cases and eight HBV− alcoholic cases in different experiments. Fresh HCC tissues and surrounding nontumor adjacent liver tissues (at least 3 cm distant from the tumor site) were used Z-VAD-FMK ic50 for the isolation of tumor- and nontumor-infiltrating leukocytes. For survival analysis, we followed 99 HBV-associated HCC patients after surgical resection from January 2007 to April 2010 (Table 1). The research was approved by the Institutional Review Board of Tongji Medical College of Huazhong University of Science and Technology. Both written and oral consent was obtained before samples were collected. Immune cells were obtained from peripheral blood and fresh liver tissues as described.19 CD14+ tumor-associated Kupffer cells (KCs) and Tim-3+CD4+ T cells were isolated with paramagnetic beads (StemCell Technology, Canada) and sorted.

Cell purity was >90% as confirmed by flow cytometry (LSR II, Becton Dickinson). Immune cells were stained extracellularly with fluorochrome-conjugate-specific antibodies against human antibodies, then fixed and permeabilized with Perm/Fix solution (eBioscience), and stained for intracellular cytokines and Ki67 (eBioscience). Tim-3+CD4+ T cells (5 × 105/ml) were cocultured with CD14+ KCs (105/mL) from the same HCC tissue from six patients for 5 days in the presence of antihuman CD3 (2.5 μg/mL, BD Biosciences) and antihuman CD28 (1.25 μg/mL) or with autologous HCC (105/mL). Neutralizing monoclonal antibody (mAb) medchemexpress against 17-AAG cell line human Tim-3 (10 μg/mL, Biolegend) or isotype controls were added to the culture. The resultant cells were collected for flow cytometry analysis or for ELISPOT assay with ImmuneSpot analyzer (Cellular Technology).

Carboxyfluorescein succinimidyl ester (CFSE)-labeled Tim-3+CD4+ T cells were incubated with CD14+ KCs from the same HCC tissue from six patients for 5 days. Cell division was determined based on CFSE dilution by flow cytometry analysis. Frozen tissue sections were stained with primary antibodies, rat monoclonal antihuman Tim-3 (clone: 344823, 1/200, IgG2a, R&D Systems), mouse antihuman CD4 (Clone: RPA-T4, 1/500, IgG1, eBioscience), mouse antihuman galectin (clone: 9M1-3, 1/500, IgG1, Biolegend), and CD68 (clone: Y1/82A, 1/500, IgG2b, eBioscience), and subsequently stained with secondary antibodies, Alexa Fluor 568-conjugated goat antirat IgG2a, Alexa Fluor 488-conjugated goat antimouse IgG1, and Alexa Fluor 568-conjugated goat antimouse IgG2b (all 2 μg/mL, Invitrogen). Hoechst 33342 (Invitrogen) was used for nuclear staining. Images were acquired by fluorescence microscope and positive cells were quantified by ImagePro Plus software (Media Cybernetics, Bethesda, MD) and expressed as the mean of the percentage of positive cells ± standard error of the mean (SEM) in five high-powered fields.

Such induction was higher in STAT3Mye−/− mice but lower in STAT3H

Such induction was higher in STAT3Mye−/− mice but lower in STAT3Hep−/− and STAT3Mye−/−Hep−/− mice (Fig. 4 and Supporting Fig. 5), which is consistent with the grade of liver regeneration in these mice, as illustrated in Fig. 2. In

addition, SOCS3 but not selleck chemicals SOCS1 was induced after PHx in wild-type mice, consistent with earlier findings.11 Similar induction of SOCS3 was also observed in STAT3Mye−/− mice. Interestingly, SOCS1 but not SOCS3 was significantly induced after PHx in both STAT3Hep−/− and STAT3Mye−/−Hep−/− mice. pSTAT3 and pSTAT1 activation were also examined in liver leukocytes after sham operation or PHx. pSTAT3 was detected post-PHx in the liver leukocytes from wild-type and STAT3Hep−/− mice but not from STAT3Mye−/− and STAT3Mye−/−Hep−/− mice (Fig. 4B). Constitutive activation of pSTAT1 was detected in the liver leukocytes of STAT3Mye−/− mice before or after sham or PHx, in agreement with previous reports.17 pSTAT1 was detected in the liver leukocytes 3 hours post-PHx in all groups with the highest levels in STAT3Mye−/−Hep−/− mice. The above data (Fig. 4) indicate increased activation of pSTAT1 in the inflammatory cells of STAT3Mye−/− mice Transmembrane Transporters modulator and in the liver of STAT3Hep−/− mice, respectively, and increased activation in both the inflammatory cells and the liver in STAT3Mye−/−Hep−/− mice. Because STAT1 plays a key role in the induction of inflammation, cell apoptosis and

cell cycle arrest,21 it is possible that elevation of STAT1 in hepatocytes contributes to reduced liver regeneration in STAT3Hep−/− mice, elevation of STAT1 in inflammatory cells contributes to enhanced inflammation in STAT3Mye−/− mice, while the simultaneous elevation of pSTAT1 in both inflammatory cells and the liver contributes to liver failure and impaired liver regeneration in STAT3Mye−/−Hep−/− mice. To test these possibilities, we generated STAT3Hep−/−STAT1−/−, MCE STAT3Mye−/− STAT1−/−, and STAT3Mye−/−Hep−/−STAT1−/− mice. Expression of STAT1 protein in the liver was induced in STAT3Hep−/− mice but not in wild-type mice (Fig. 5A), which is consistent

with previous findings.12 Western blot analyses confirmed the absence of STAT1 and STAT3 protein expression in the liver of STAT3Hep−/−STAT1−/− mice (Fig. 5B). All STAT3Hep−/−STAT1−/− mice survived after PHx (Fig. 5C) and had a greater number of Brdu+ hepatoctyes than STAT3Hep−/− mice after PHx (Fig. 5D), suggesting that deletion of STAT1 in STAT3Hep−/− mice restores the ability of the liver to regenerate. Treatment with a low dose of IFN-γ induced stronger pSTAT1 activation in STAT3Hep−/− than in wild-type hepatocytes (Fig. 5E). As expected, no STAT1 or STAT3 proteins were detected in STAT3Hep−/−STAT1−/− hepatocytes. Furthermore, STAT3Hep−/− hepatocytes were more susceptible to IFN-γ inhibition of cell proliferation, an effect that was abolished in STAT3Hep−/−STAT1−/− hepatocytes. Western blot analyses (Fig.

Furthermore,

as per the Asian standard of BMI categories,

Furthermore,

as per the Asian standard of BMI categories, the distribution pattern of LSM would not have been U-shaped. Although underweight subjects had significantly higher LSM values than healthy and preobese subjects, the mean difference in absolute value was only 0.5 KPa, suggesting that the body-size effect is not perceptible using the current FibroScan machines. The ULN of LSM has been reported to vary from 5.3 to 7.0 KPa in various studies,2, 3 including one study based on histopathology.2 Thus, 8.5 kPa as the ULN of LSM seems too high for a healthy liver in any given population across the world. Such a high cutoff may erroneously cause the exclusion of healthy subjects or patients who would require further evaluation. Though this value corresponded to the 95th percentile, the dispersion selleckchem of data (mean, 95%

confidence interval) revealed that 414 of 418 healthy subjects actually had LSM values within 6.5 KPa, as shown in Fig. 2 of the Das et al. study.1 Notably, sufficient investigations were not done to exclude potential underlying liver disease in healthy subjects with high LSM. Also, approximately 16% of LSM results are known to be unreliable. In our experience, in a cohort of 445 healthy adult subjects, the mean LSM value was 5.1 ± 1.1 KPa, and the 95th percentile value was 7.07 KPa. LSM values increased with increasing BMI categories (4.1 ± 0.7, 5.08 ± 0.6, and 6.08 ± 1.2 KPa in healthy BMI, overweight, and obese subjects, respectively).3 None of our patients belonged to the underweight category; hence, the distribution of LSM cannot be compared with

the present selleck kinase inhibitor study. To conclude, the present study results seem to have limited external validity. However, the background population and properly validated regional data are important while interpreting LSM 上海皓元 using FibroScan. Ramesh Kumar M.D., D.M.*, Manoj Kumar Sharma M.D., D.M.*, Shiv Kumar Sarin M.D., D.M.*, * Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India. “
“A 46 year-old man with hypertension, diabetes and end stage renal disease on peritoneal dialysis (PD) presented with generalized abdominal pain, distention and constipation. His abdominal exam revealed diminished bowel sounds, dullness to percussion and diffuse tenderness to deep palpation without guarding or rebound tenderness. He had a PD catheter in the right lower quadrant. Laboratory analysis revealed a WBC of 9600 cells/mm3, eosinophil count of 730 cells/mm3, hematocrit 27%, creatinine 6.2 mg/dL, blood urea nitrogen 27 mg/dL and albumin 1.5 g/dL. Serum aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, total bilirubin, amylase and lipase levels were within normal limits. Peritoneal fluid aspiration through the PD catheter revealed bloody ascites. Halfway through aspiration, the syringe plugged. Careful, brief tension was applied and a white, smooth, cylindrical specimen measuring 15.0 × 0.3 cm was extracted into the syringe (Figure 1).

In this strain, all four 16S rRNA genes were identical, which is

In this strain, all four 16S rRNA genes were identical, which is in agreement with a study of Engene and Gerwick (2011). Two operons contained nearly identical 16S-23S ITS regions with both tRNA genes, and two operons contained nearly identical 16S-23S ITS regions lacking tRNA genes. This result Ibrutinib nmr was consistent with what we found in all other Cylindrospermum strains, and we assume that all strains had this pattern even when one of the ITS operons was not recovered. The 16S-23S ITS regions were highly similar within the Cylindrospermum taxa in clades X and Y, permitting alignment of both operon 1 and operon 2. Phylogenies of these ITS regions had high bootstrap support,

and are superior to the 16S rRNA phylogenies for elucidating relationships among species (Fig. 1, b and c). These trees were in fairly close agreement with each other and with the 16S phylogeny. The ITS phylogenies were not in disagreement with our species assignments, i.e. no species was clearly not monophyletic. Secondary structure of the conserved domains of the ITS revealed a number of species-specific patterns. The D1-D1′ helices were identical for C. muscicola SAG 44.79, C. licheniforme CCALA 995, C. badium CCALA 1000, and the five strains of

C. catenatum, although sequence differences existed (note ambiguous bases W and R, group I; Fig. 2a). Cylindrospermum pellucidum CCALA 989/CCALA 992, C. stagnale PCC 7417, and C. moravicum CCALA www.selleckchem.com/products/icg-001.html 993 possessed helices structurally similar to the D1-D1′ helices of the C. catenatum group, but differed in minor

ways (Fig. 2, b and c). Cronbergia also had a D1-D1′ helix similar to all of the above (Fig. 2d). Cylindrospermum stagnale PCC 7417 had a single nucleotide in the terminal loop that differed from the above group (Fig. 2, a–d), such that the terminal loop was divided into a small terminal loop and a subterminal bilateral bulge (Fig. 2h). The C. catenatum group could close the large terminal loop in this position as well, but this structure would be less thermodynamically stable (GC….GU binds, but more weakly than the AC….GU in C. stagnale PCC 7417). A major break in structure occurred between the group including C. catenatum and the group of species MCE公司 in the clade containing C. marchicum CCALA 1001, C. alatosporum CCALA 988/CCALA 994, and C. maius CCALA 998 (Fig. 2, e–g). The Hawaiian Cylindrospermum CCALA 1002 had different helices in the two different operons, both of which showed significant divergence from the helices of other species (Fig. 2, i and j). Aulosira bohemensis was also very distinct (Fig. 2k). The V2 helix is located between the tRNAIle and tRNAAla genes, and is thus only present in those operons, which contain the tRNA genes. This helix varied considerably among strains. C. catenatum (CCALA 990, 991, 996) and C.

, 2005) For example, a closely related species, the red-faced ci

, 2005). For example, a closely related species, the red-faced cisticola Cisticola erythrops, shows a pattern of song variability, which results from changes in syllable use and delivery order (Benedict & Bowie, 2009). In the red-faced cisticola, song form has apparently been shaped by multiple evolutionary forces, including diversifying cultural drift and stabilizing selection on syllable delivery rate that may help delimit species boundaries (Benedict & Bowie, 2009). With the data presented here, we quantify song variation across the geographic range of the rattling cisticola, we look for song features that are useful for species identification, and we discuss evolutionary processes that may have

generated the observed patterns. Rattling cisticolas are expected Selleck Protease Inhibitor Library to experience stabilizing selection on songs as indicators of species identity, so we predict that some elements of song will be stable

across the geographic range. At a local scale, rattling cisticolas live in social groups where males sing to defend territories of varying quality p38 inhibitors clinical trials (Carlson, 1986). We therefore predict that sexual selection will select for diversity of some song elements as signals of individual identity and quality (Catchpole, 1987; Andersson, 1994). Finally, we predict that patterns of syllable use and other song features are likely to vary across the large geographic range of this species. We analyzed 61 recordings (957 songs) of rattling cisticolas obtained from the British Library, the Macaulay Library of Natural Sounds and the Ditsong National Museum of Natural History (Transvaal Museum) (Appendix S1). The use of archived songs allowed access to many sites widely distributed across sub-Saharan Africa. We assessed all sound files to confirm that the songs were from a rattling cisticola and found

that every song matched the general species description of having several introductory notes followed by a trill-like end phrase. Closely related species of cisticolas that are found sympatrically with rattling cisticolas have very different song structures, making us confident in our identifications (L. Benedict, unpubl. data). Tracks varied in length 上海皓元 from 5 to 404 s (mean = 71.1 ± 73.4 s) and included from 1 to 81 songs (mean = 15.7 ± 16.3). Recordings came from 38 different sites. Based on statements and notes from the recordists, we eliminated sound files from our analysis that were duplicates of a single bird. We did include tracks that were recorded at similar locations and/or dates but were distinct in time or specific location as they are likely to represent different birds. Analyzed recordings represented 12 of 17 subspecies and covered most of the species’ geographic range (Fig. 1). We assigned subspecies identity following Erard et al. (1997). We used Google Earth (http://www.google.com/earth/index.html) to obtain elevation, latitude and longitude (Appendix S1). We examined song diversity in two ways.

Wiegand, Birgit Bremer, Andreas Geipel, Corinna M Bremer, Anika

Wiegand, Birgit Bremer, Andreas Geipel, Corinna M. Bremer, Anika Wranke, Dieter Glebe Introduction: Hepatitis delta is the most severe

form of viral hepatitis with a fast progression of fibrosis to cirrhosis. Treatment options are still very limited as PEG-interferon alfa is effective only in a minority of patients. Therefore, appropriate determination of stage of liver disease is desired. Non-invasive fibrosis scores used for other liver diseases including APRI-score, AST/ALT ratios or FIB-4 index do not perform well in hepatitis delta. We here aimed to develop novel non-invasive fibrosis scores optimized for patients with hepatitis delta. Methods: In the ongoing HIDIT-2 treatment trial Decitabine chemical structure 120 patients with chronic hepatitis delta were recruited. Liver biopsies were evaluated centrally by two independent pathologists. Additionally, Akt inhibitor 50 cytokines, chemokines, growth factors and angiogenic factors were measured in sera of 100 of the 120 patients using multiplex technology (Bio-Plex System). Anti-HDV-IgM-testing was performed in all patients by the ETI-DELTA-IGMK-2

assay (Diasorin). T-test was used to identify factors associated with cirrhosis or fibrosis. MCE With ROC curve analysis and calculation of the Youden index cut offs were determined differentiating cirrhosis and non-cirrhosis as well as fibrosis and non-fibrosis for each factor. In a last step logistic regression was used to identify the most important factors differentiating fibrosis and cirrhosis

in order to create the new score. Results: Four factors were identified differentiating between cirrhosis and Ishak scores <5 (MIF, AST/ALT ratio, age, HGF). Defined cut-offs were determined for each factor (MIF >3400 ng/ml, AST/ALT >0.8, age >35 and HGF >370 ng/ml) which were then included in the following equitation (1 point × the indicated factor for each variable if above the cut-off): 5*MIF+2*(AST/ALT)+AGE+HGF. The AUC of the new score was 0.84; >2 points predicted cirrhosis with a sensitivity of 85%, a specificity of 69%, a PPV of 72% and a NPV of 83%. In order to differentiate between fibrosis (Ishak-score >2) and non-fibrosis, another score was similarly determined based on 6 variables: 0.