They showed a dramatic reduction (∼35%-45%) in stellate cell chem

They showed a dramatic reduction (∼35%-45%) in stellate cell chemotaxis, proliferation, and collagen production with Ccl5−/− splenocytes. This reduction in fibrogenic activity was even greater when stellate cells were pretreated with Met-CCL5 before the treatment with WT splenocyte–conditioned media (∼75%-80%). In the in vivo studies, Met-CCL5 (administered concomitantly with either

CCl4 or the MCD diet) significantly inhibited hepatic fibrosis progression (∼20%-40%) and the expression of hepatic genes associated with fibrogenesis. In both animal models of hepatic fibrosis, CD8+ T cells and CD68+ macrophages were significantly reduced by the in vivo Met-CCL5 treatment, whereas the numbers of natural killer and natural killer T cells, B220+ B cells, and CD11c+ dendritic cells were unchanged. When daily Met-CCL5 treatments were administered after the establishment of fibrosis by an 8-week selleck screening library CCl4 injection regimen (3 days after the final CCl4 injection), they augmented the regression of hepatic fibrosis (∼50%) after 7 days. These histological changes in fibrosis were preceded by the reduced expression of both procollagen α1(I) and tissue inhibitor of metalloproteinase 1 mRNA levels in the liver. These data are particularly interesting because they suggest the potential for the treatment of established fibrosis via the accelerated regression of fibrotic tissue, although further investigations

are warranted to selleck products evaluate 4��8C the mechanisms involved in this process. In a previous study, Ruddell et al.8 identified CD45+ immune cells as a source of RANTES in another murine model of hepatic fibrosis. They used the choline-deficient, ethionine-supplemented dietary model of hepatic injury, liver progenitor cell expansion, and portal fibrosis to demonstrate a role for the tumor necrosis factor family member lymphotoxin β (LTβ) in the process of wound healing and hepatic fibrosis.8 They proposed a novel mechanism for RANTES expression by hepatic stellate cells via

direct cell contact between liver progenitor and hepatic stellate cells that is induced by the interaction of cell surface–bound LTβ on liver progenitor cells with the LTβ receptor expressed on hepatic stellate cells. In the same study, significant numbers of CD45+ T cells were also demonstrated to express RANTES in choline-deficient, ethionine-supplemented mouse livers and were observed in close spatial association with liver progenitor cells. Neither Ruddell et al. nor Berres et al.3 examined the relative contributions of either T cells or hepatic stellate cells to RANTES expression in these models of hepatic fibrosis. Although it appears that immune cells are the major source of RANTES at least in the CCl4 and MCD models, the contributions of other resident and nonresident hepatic cells require further investigation.

Peripheral venous

blood was obtained from

Peripheral venous

blood was obtained from https://www.selleckchem.com/Wnt.html healthy donors and patients with cholestatic disorders, uremia, Hodgkin’s disease, and atopic dermatitis after informed consent, according to the Declaration of Helsinki. The study was approved by the local medical ethical committees. Treatment interventions, such as colesevelam,12 RMP, MARS therapy, and nasobiliary drainage,7 were conducted, recorded, and reported on in compliance with the International Conference on Harmonization Good Clinical Practice and national regulations. Blood samples were allowed to clot for 1 hour before they were centrifuged at 4°C, and serum was cryopreserved in aliquots at −80°C. Itch intensity was quantified in all patients at the time point of blood drawing using a visual analog scale (VAS) ranging from 0 (no pruritus) to 100 (unbearable pruritus). In the colesevelam study,12 35 patients were evaluable, of whom 17 patients received colesevelam (1,875 mg twice-daily) and 18 patients were treated with an identical placebo selleck products for 3 weeks. The study population

consisted of 22 female and 13 male patients being mainly diagnosed for PBC (N = 14) or PSC (N = 14). MARS treatment was performed in 10 patients (8 female and 2 male) with intractable pruritus resulting from PBC (n = 6), PSC (n = 2), or other liver disorders (n = 2; Supporting Table 4). Choline oxidase (ChO), horseradish peroxidase (HRP), homovanillinic acid (HVA), dimethyl sulfoxide (DMSO), bovine serum albumine

(BSA), and RMP were purchased from Sigma-Aldrich (Steinheim, Germany); stearoyl LPA (18:1) and myristoyl LPC (14:0) were from Avanti Lipids (Alabaster, AL). Human HepG2 hepatoma cells were grown in Dulbecco’s modified Eagle’s medium (DMEM; Lonza BioWhittaker, Cologne, Germany) supplemented with 10% fetal Branched chain aminotransferase calf serum, 4 mM of L-glutamine, and a mixture of antibiotics (5 mg/mL of penicillin and 5 mg/mL of streptomycin). Cells were incubated at 37°C in a humidified atmosphere containing 5% CO2. For studying the effect of RMP, cells were seeded in six-well plates at a density of 8 × 105 cells/well until reaching 80% confluence. Subconfluent cells were cultured overnight in serum-free medium containing 0.2% BSA. After brief washing, cells were incubated for 24 hours in DMEM/0.2% BSA containing 10 μM of RMP. As a solvent control, 0.1% DMSO was added to control cells. HepG2 cells overexpressing PXR and PXR knock-down HepG2 cells (see below) were identically analyzed.

Key Word(s): 1 gastric

cancer; 2 serum proteomics; 3 i

Key Word(s): 1. gastric

cancer; 2. serum proteomics; 3. iTRAQ; 4. D-LC-MS/MS; Presenting Author: MALU JUN Additional Authors: LINYAO GUANG Corresponding Author: LINYAO GUANG Affiliations: guangxi medical university Objective: To study the expression of S100A11 and Beclin1 in gastric carcinoma, precancerous lesion and chronic nonatrophic pangastritis, and the relationship between S100A11 and Beclin1 expression in gastric cancerous tissues and the biological behaviour of gastric carcinoma, investigate the mechanism and clinical significance of S100A11 and Beclin1 Doxorubicin in the development of gastric carcinoma. Methods: The expression of S100A11 and Beclin1 proteins were determined by streptavidin-perosidase immunohistochemical method in 50 cases of gastric carcinoma from exairesis tissues, 30 cases of precancerous lesion and 20 cases of chronic nonatrophic pangastritis from endoscopic biopsy. Pathological image analysis system be used to analysis the grey level of S100A11 and Beclin1, then analyze the mechanism and clinical significance of S100A11 and Beclin1 in the development of gastric carcinoma.

Results: The positive expression grey level of S100A11 in gastric carcinoma was 132.9209 ± 5.649, and in precancerous lesion tissues was 133.6706 ± 5.8348, both of them were significantly lower than that of in chronic nonatrophic Montelukast Sodium pangastritis tissues (138.048 ± 3.5902), ICG-001 in vivo There were significant difference between the gastric carcinoma and chronic nonatrophic pangastritis tissues, precancerous lesion tissues and chronic nonatrophic pangastritis tissues (P < 0.05), But there was no difference between the gastric carcinoma and precancerous lesion tissues (P > 0.05). There was obvious correlation between the expression of S100A11 and the clinicopathological

factors, such as grading, infiltrating depth, lymph nodes metastasis, TNM degree (P < 0.05), but there was no correlation between the expression of S100A11 and position, knubbly diameter (P > 0.05). The positive expression grey level of Beclin1 in gastric carcinoma was 140.9705 ± 6.2019, which was significantly higher than those in precancerous lesion tissues (136.711 ± 5.5759) and in chronic nonatrophic pangastritis tissues (130.8024 ± 2.5363), there were significantly differences between two of the three tissues (P < 0.05). There was correlation between the expression of Beclin1 and grading, lymph nodes metastasis (P < 0.05), but there was no correlation between the expression of Beclin1 and position, diameter, infiltrating depth, TNM degree (P > 0.05), There existed a negative correlation between S100A11 and Beclin1 in gastric carcinoma (r = −0.156, P < 0.05).

highlights the fact that current criteria as defined by the Inter

highlights the fact that current criteria as defined by the International Ascites Club need revision. Current proposals for a working definition of HRS have largely adopted current AKI criteria,5 but we

need to recognize the clinical reality that not all patients who might be classified as having type 1 HRS should necessarily be included in one grouping, and this is where current AKI criteria or the new proposals let us down. This is not just about putting patients into brackets or classifying them, it is about understanding the mechanisms of disease. Further, the current RIFLE criteria and indeed the proposals put forward by Wong et al.5 put patients with refractory ascites into a group with chronic kidney disease, and yet much of the renal dysfunction is entirely reversible. Belcher et al. recognized this dilemma when they state “We have avoided using the https://www.selleckchem.com/products/DAPT-GSI-IX.html term ‘chronic kidney disease’ as this classically implies structural damage. Many patients with cirrhosis have a chronically depressed GFR due instead to persistent hypoperfusion and their renal function may thus be partially reconstituted with restitution of perfusion. The article by Belcher et al.7 highlights the need for all new definitions of HRS to recognize that patients with cirrhosis may develop acute

kidney injury for a variety of reasons, many of which involve bacterial infection, or rapid decompensation of liver function (e.g., alcoholic hepatitis without infection), shock, administration of a nephrotoxic drug, as well as those having “true” chronic Autophagy Compound Library price kidney disease, rather than renal hypoperfusion. We need to be able to identify patients early, both as new patients and importantly those patients who develop AKI following admission to the hospital, since this latter group have a higher mortality, and this should be preventable.6 What is the purpose of a definition of HRS? Why not just group everyone together as recently proposed?5 The purpose is to recognize the different clinical entities that arise so that we do not group all patients as being one and equal, but classify them so that we can

increase our understanding of the underlying pathophysiology and Decitabine nmr develop targeted therapies. While it is clear from the two largest trials of terlipressin in HRS2, 3 that not all patients respond to therapy, we need to identify the different clinical entities that may respond to different therapies, in the same way that pharmacogenomics is beginning to identify subsets of patients who respond to certain drugs. The article by Parikh et al. highlights the problems and dilemmas we face.6 There are major problems with the current definition of HRS, but there are also problems if we simply adopt AKI criteria. We need robust criteria to classify patients so that our future therapies are individualized to the patient, so that they can be more effective.

NAFLD was present in 59 6%, borderline NASH in 14 2% and definite

NAFLD was present in 59.6%, borderline NASH in 14.2% and definite NASH in 6.4%. Stage 1 fibrosis was present

in 15.6% and stage 2 in 2.8%; 1 subject had stage 3 and none had cirrhosis. Compared MS-275 manufacturer to subjects with Not-NAFLD and with NAFLDNot NASH, more subjects with borderline/definite NASH were male and had diabetes and hypertension. Serum ALT, AST, HOMa-IR, and triglyceride levels were also higher. Pre-operative weight loss >5% occurred in 11% of subjects but did not vary by disease grade. Conclusions: The presence of NASH in a multicenter cohort of severely obese adolescents undergoing WLS was associated with male gender and higher cardiometabolic risk. While NAFLD was common, prevalence of advanced fibrotic NASH was low. This may reflect younger age, demographic or referral patterns, or biological factors specific to severe obesity and merits further study. Characteristics and significant determinants of liver histology Not-NAFLD (n=57) NAFLD-Not NASH (n=55) Borderline/Definite NASH (n=29) NAS=NAFLD activity score Disclosures: Marc Michalsky – Grant/Research

Support: Allergan Health, Irvine CA Thomas H. Inge – Grant/Research Support: Ethicon The following people find more have nothing to disclose: Stavra A. Xanthakos, Tawny P. Wilson, David E. Kleiner, Todd M. Jenkins, Reena Mourya, Mary L. Brandt, Carroll M. Harmon, Michael A. Helmrath, Anita P. Courcoulas, Meg H. Zeller Background & Aims: Nonalcoholic fatty liver disease (NAFLD) is closely related to metabolic syndrome and obesity which are associated with an increased risk of various malignancies. In this study, we investigated the association between NAFLD and prostate cancer biochemical recurrence (BCR) after radical prostatectomy (RP). Methods: Consecutive prostate cancer patients who underwent RP between 2005 and 2008 at a single tertiary hospital in Korea were included in this study. The Celecoxib presence of NAFLD, body mass index (BMI), pre-diagnostic prostate-specific antigen

(PSA), and histological findings including Gleason score were analyzed with regard to their associations with BCR. NAFLD was diagnosed based on clinical information and ultrasonography or unenhanced CT images. BCR-free survival rates were calculated using the Kaplan-Meier method. Results: A total of 222 patients were analyzed. During a median follow-up period of 54 (inter-quartile range, 44-65) months, 45 (20.3%) patients developed BCR. The presence of NAFLD was significantly associated to longer time-to-BCR (P=0.001 by log rank test), while BMI failed to show statistical significance (P=0.861). In multivariate analysis, the presence of NAFLD (hazard ratio [HR], 0.26; 95% confidence interval [CI], 0.11-0.61; P=0.002), pathological Gleason score (compared to <7, 7: HR, 2.92; 95% CI, 1.12-7.64; P=0.029, >7: HR, 6.64; 95% CI, 2.26-19.52; P=0.001), and positive surgical margin (HR, 2.17; 95% CI, 1.18-3.99; P=0.013) were independent predictive factors of BCR.

We found a link between the expression of CTLA-4 and the proapopt

We found a link between the expression of CTLA-4 and the proapoptotic mediator Bim in HBV-specific CD8. Longitudinal study of a cohort of CHB patients commencing antiviral therapy showed that viral load reduction did not reduce CTLA-4 or Bim levels in antiviral T cells. We therefore explored the potential to manipulate this coinhibitory pathway in vitro to restore expansion of HBV-specific CD8

T cells. ALT, alanine transaminase; CHB, chronic hepatitis B virus infection; CTLA-4, cytotoxic T lymphocyte antigen-4; HBV, hepatitis B virus; OLP, overlapping peptides. The study was approved by the local Ethical Committees selleck chemicals llc and written informed consent was obtained from all patients. A total of 86 patients with CHB, three patients with resolved HBV infection, and 23 healthy volunteers participated in the study; there were no significant differences in their demographics (Table 1). All participants were HCV and HIV seronegative and cytomegalovirus (CMV) seropositive. Patients with CHB were stratified by HBV DNA levels above or below 2,000 IU/mL (determined by real-time polymerase chain reaction [PCR]), according to European Association for the Study of the Liver (EASL) guidelines.13 All CHB patients were treatment-naïve at recruitment; a subgroup of seven patients was followed

longitudinally after commencing lamivudine and adefovir (Table 2). Hepatitis B s-antigen (HBsAg) was quantitated with the Architect assay. Paired peripheral blood and liver biopsy selleck inhibitor specimens (surplus to diagnostic requirements) were obtained from eight patients with CHB (Table 1). PBMCs were isolated by Ficoll-Hypaque density gradient centrifugation and cultured on anti-CD3 monoclonal antibody (mAb)-coated plates (1 μg/mL) or in medium alone for 16 hours before analysis of CTLA-4 in total CD8 T cells. For detection of intracellular CTLA-4 on virus-specific CD8 T cells ex vivo, cells were stained with human leukocyte antigen A2 (HLA-A2)/c18-27, HLA-A2/e183-191, HLA-A2/e335-343,

and HLA-A2/e348-357 HBV dextramers (Immudex) before stimulation with HBV-specific peptides for 4 hours in the presence of Brefeldin A. CMV-specific CD8 T cells were detected by HLA-A2/NLVPMVAYV pentamers (Proimmune). For functional detection of virus-specific CD8 T cells, cells were stimulated with HBV or control viral peptide and cultured for 10 3-mercaptopyruvate sulfurtransferase days, supplemented with 20 U/mL IL-2 at 0 and 4 days, restimulated with 1 μM peptide for 16 hours in the presence of 1 μg/mL Brefeldin A (Sigma-Aldrich), and identified by intracellular staining for IFN-γ. To examine the effect of blocking inhibitory pathways, purified, NA/LE monoclonal antibodies against CTLA-4 (BD Biosciences), PD-L1, PD-L2 (eBioscience), or control IgG (BD-Biosciences) were added at 5 μg/mL with peptide at onset of culture. Responses were analyzed as described above. Liver sections from biopsies were homogenized and filtered.

Results: Sustained reduction in ALT level was similar to placebo

Results: Sustained reduction in ALT level was similar to placebo (10/58; 17%; 95% CI, 9% to 29%) in both the vitamin E (15/58; 26%; 95% CI, 15% to 39%; P = .26) and metformin treatment groups (9/57; 16%; 95% CI, 7% to 28%; P = .83). The mean change in ALT level from baseline to 96 weeks was -35.2 U/L (95% CI, -56.9 to -13.5) with placebo vs -48.3 U/L (95% CI, -66.8 to -29.8) with vitamin E (P = .07) and -41.7 U/L (95% CI, -62.9 to -20.5) with

metformin (P = .40). The mean change at 96 weeks in hepatocellular ballooning scores was 0.1 with placebo (95% CI, -0.2 to 0.3) vs -0.5 with vitamin find more E (95% CI, -0.8 to -0.3; P = .006) and -0.3 with metformin (95% CI, -0.6 to -0.0; P = .04); and in NAFLD activity score, -0.7 with placebo (95% CI, -1.3 to -0.2) vs -1.8 with vitamin E (95% CI, -2.4 to -1.2; P = .02) and -1.1 with metformin (95% CI, -1.7 to -0.5; P = .25). Among children with NASH, the proportion who resolved at 96 weeks was 28% with placebo (95% CI, 15% to 45%; 11/39) vs 58% with vitamin E (95% CI, 42% to 73%; 25/43; P = .006) and 41% with metformin (95% CI,

26% to 58%; 16/39; P = .23). Compared with placebo, neither therapy demonstrated significant improvements in other histological features. Conclusion: Neither vitamin E nor metformin was superior to placebo in attaining the primary outcome of sustained reduction in ALT level in patients with pediatric NAFLD. TRIAL REGISTRATION: clinicaltrials.gov selleck chemical Identifier: NCT00063635. Nonalcoholic fatty liver disease (NAFLD) has rapidly become the most common form of chronic liver disease in children1 and is a major reason for referral to pediatric gastroenterologists and hepatologists. This dramatic increase in the prevalence of NAFLD is a direct consequence of the childhood obesity epidemic, with an estimated 10% of U.S. children having fatty liver.2 The spectrum

of NAFLD ranges from the benign form of hepatic steatosis, with the accumulation of lipids in the liver, to the progressive form of nonalcoholic steatohepatitis (NASH), which Janus kinase (JAK) is characterized by steatosis along with inflammation, hepatocyte injury, and variable degrees of fibrosis.3 It is important to recognize that NASH may present with a distinct histological pattern in children, mainly characterized by increased portal inflammation and fibrosis, as opposed to the predominantly lobular inflammation and perisinusoidal fibrosis observed in adults.4, 5 The progression of NASH to cirrhosis that requires liver transplantation during childhood is well documented,6 making the identification of effective therapy for this condition a pressing issue. Weight loss may improve the liver disease or even lead to the resolution of NAFLD in some children,7 but other than lifestyle advice on diet and exercise, there are no approved therapies for pediatric NAFLD. Insulin resistance (IR) and oxidative stress (OS) play a significant role in disease development and progression (Fig.

F Hoffmann-La Roche Ltd-funded   HBeAg-pos (N=182) HBeAg-neg (N=

F. Hoffmann-La Roche Ltd-funded   HBeAg-pos (N=182) HBeAg-neg (N=430) Male sex, n (%) 125(69) 306(71) Caucasian/White race, n/N* (%) 80/132(61) 249/289 (86) Mean age ± SD 31.3 ±10.5 36.3 ±11.4 Mean ALT ratio ± SD 2.6 ± 2.3 1.8 ±1.9 Cirrhosis/bridging fibrosis, n/N (%) 21/151 (14) 39/384(10) Mean HBV DNA, log10 IU/mL ± SD 6.75 ± 2.06 4.14± 1.77 Mean HBsAg, log10 IU/mL ± SD 3.96 ± 0.84 3.49 ±0.89 Previous nucleos(t)ide analog, n (%) 39(21) 66(15) LBH589 manufacturer * Patients from France do not ha e race recorded due to loci 1 regulations Disclosures: Patrick Marcellin – Consulting: Roche, Gilead, BMS, Vertex, Novartis, Janssen-Tibotec, MSD, Boehringer, Pfizer, Abbott, Alios BioPharma;

Grant/Research Support: Roche, Gilead, BMS, Novartis, Janssen-Tibotec, MSD, Alios BioPharma; Speaking and Teaching: Roche, Gilead, BMS, Vertex, Novartis, Janssen-Tibotec, MSD, Abbott Wlodzimierz W. Mazur – Advisory Committees or Review Panels: Bristol-Myers-Squibb company; Speaking and Teaching: Gilead, MSD, Roche, Abvee Christophe Hezode – Speaking and Teaching: Roche, BMS, MSD, Janssen, abb-vie, Gilead Dominique Guyader – Advisory Committees or Review

Panels: Roche, Gilead, IRIS; selleck chemicals llc Board Membership: Merck; Grant/Research Support: Janssen; Speaking and Teaching: BMS Christoph Jochum – Advisory Committees or Review Panels: Gilead, Roche, Norgine, Janssen-Cilag; Speaking and Teaching: BMS, Roche, Janssen-Cilag, Gilead Graham R. Foster – Advisory Committees or Review Panels: GlaxoSmithKline, Novartis, Boehringer Ingelheim, Tibotec, Chughai, Gilead, Janssen, Idenix, GlaxoSmithKline, Novartis, Roche, Tibotec, Chughai, Gilead, Merck, Janssen, Idenix, BMS; Board Membership: Boehringer Ingelheim; Grant/Research Support: Chughai, Roche, Chughai; Speaking and learn more Teaching:

Roche, Gilead, Tibotec, Merck, BMS, Boehringer Ingelheim, Gilead, Janssen Markus Cornberg – Advisory Committees or Review Panels: Merck (MSD Ger-mamny), Roche, Gilead, Novartis; Grant/Research Support: Merck (MSD Ger-mamny), Roche; Speaking and Teaching: Merck (MSD Germamny), Roche, Gilead, BMS, Novartis, Falk Manfred Bogdan – Management Position: Roche Pharma, Germany Diethelm Messinger – Consulting: Roche, Roche Veronique Cartier – Employment: ROCHE Joerg Petersen – Advisory Committees or Review Panels: Bristol-Myers Squibb, Gilead, Novartis, Merck, Bristol-Myers Squibb, Gilead, Novartis, Merck; Grant/Research Support: Roche, GlaxoSmithKline, Roche, GlaxoSmithKline; Speaking and Teaching: Abbott, Tibotec, Merck, Abbott, Tibotec, Merck The following people have nothing to disclose: Manuela G. Curescu, Anna Piekarska, Denis Ouzan Introduction. Treatment failure to nucleos(t)ide analogues (NUC) in chronic hepatitis B (CHB) patients could occur due to limited antiviral potency, viral resistance or patient non-adherence. However, real-life prospective data on treatment adherence in CHB patients are scarce.

1 Liver stem cells, or even stem cells derived from other tissues

1 Liver stem cells, or even stem cells derived from other tissues, could potentially provide a source of

human hepatocytes for regeneration of the injured liver.3, 4 In particular, mesenchymal stem cells (MSCs), shown to be capable of in vitro differentiation into hepatocytes,5 were investigated as a possible source of hepatocytes for liver regeneration. In addition, it has been shown that secretion of trophic molecules by MSCs may favor regeneration following acute liver injury.6 In a previous study, we isolated a population of human adult liver stem cells (HLSCs) expressing MSC markers and certain embryonic and hepatic cell markers, and having multipotent differentiation capabilities and regenerative properties.7 However, the therapeutic potential of HLSCs and HLSC-conditioned medium (CM) in FLF

has not yet been evaluated. In this study we investigated SB203580 in vitro PS-341 ic50 the effect of HLSCs and HLSC-derived CM in a lethal model of liver injury induced by D-galactosamine (GalN) and lipopolysaccharide (LPS) in SCID mice. HLSCs were isolated from human cryopreserved normal hepatocytes and MSCs were obtained from Lonza (Basel, Switzerland) and were cultured as described in the online Supporting Information.7, 8 Detailed protocols for the preparation of CM from HLSCs or MSCs9 are provided in the online Supporting Information. CM, conditional medium; FLF, fulminant liver failure; GalN, D-galactosamine; HLSCs, human liver stem cells; LPS, lipopolysaccharide; MSCs, mesenchymal stem cells. The CM was analyzed

for specific proteins, using multiplex biometric immunoassay, Bioclarma (Bio-Plex Human Cytokine Assay; Bio-Rad Laboratories, Hercules, CA) and data were confirmed by enzyme-linked Succinyl-CoA immunosorbent assay (ELISA). Studies were approved by the University of Torino Ethics Committee and conducted in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals. Intramuscular injection of Zolazepam (0.2 mL/kg) and Xilazin (16 mg/kg) were used as anesthesia (40 µL/mouse). FLF was induced in male SCID mice (7-8 weeks old) (Charles River Laboratories, Milan, Italy), by intraperitoneal injection of GalN (600 mg/kg body weight) and LPS (125 ng per animal).10 Injection of GalN and LPS induced liver injury causing apoptosis and necrosis of hepatocytes with 100% lethality at 8 hours. Thirty minutes after GalN/LPS administration, mice received different treatments. The following groups were studied: group 1, FLF mice intravenously injected with vehicle alone (n = 18); group 2, healthy mice intraperitoneally injected with vehicle instead of GalN/LPS (n = 6); group 3, FLF mice intravenously injected with 2 × 106 HLSCs (n = 9, 3.3 × 105 cells given six times for a total number of 2 × 106); group 4, FLF mice intravenously injected with 2 × 106 MSCs (n = 6, 3.


“Netherlands Cancer Institute, Division of Gene Regulation


“Netherlands Cancer Institute, Division of Gene Regulation, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands Chronic hepatitis B virus (HBV) infection is a major Alectinib mw risk factor for liver cancer development. HBV encodes the hepatitis B virus X (HBx) protein that promotes transcription of the viral episomal DNA genome by the host cell RNA polymerase II. Here we provide evidence that HBx accomplishes this task by a conserved and unusual mechanism. Thus, HBx strongly stimulates expression of transiently transfected reporter constructs, regardless of the enhancer and promoter sequences. This activity invariably requires HBx

binding to the cellular UV-damaged DDB1 E3 ubiquitin ligase, suggesting a common mechanism. Unexpectedly, none of the reporters tested is stimulated by HBx when integrated into the chromosome, despite remaining responsive to their cognate activators. Likewise, HBx promotes gene expression from the natural HBV episomal template but not from PS-341 chemical structure a chromosomally integrated HBV construct. The same was observed with the HBx protein of woodchuck HBV. HBx

does not affect nuclear plasmid copy number and functions independently of CpG dinucleotide methylation. Conclusion: We propose that HBx supports HBV gene expression by a conserved mechanism that acts specifically on episomal DNA templates independently of the nature of the cis-regulatory sequences. Because of its uncommon property and key role in viral transcription, HBx represents an attractive target for new antiviral therapies. (HEPATOLOGY 2012;56:2116–2124) Chronic infection by hepatitis B virus (HBV) affects close to 400 million people worldwide and is a leading cause of hepatocellular carcinoma, one of the most common human cancers.1 HBV belongs to the hepadnavirus family of DNA viruses that also includes woodchuck hepatitis virus and ground

squirrel hepatitis virus.2 HBV replicates its genome in a manner very analogous to retroviruses, by reverse transcription (RT) of an RNA intermediate into double-stranded DNA that serves as template for transcription by the host cell RNA Polymerase II (Pol II) (reviewed3). A distinctive feature of HBV, Selleck Sunitinib however, is that the viral DNA genome does not integrate into the chromosome of the newly infected cell but instead is maintained as a covalently closed circular DNA (cccDNA) molecule. The cccDNA is transcribed into four major RNA species encoding the viral proteins, including a more than full-length transcript termed the pregenomic RNA. The pregenomic RNA is then reverse-transcribed in the cytoplasm within newly formed viral particles. As the cccDNA does not replicate, a pool of 10-100 copies of the cccDNA is maintained by recycling of a small proportion of the newly synthesized viral DNA genomes into the nucleus. HBV encodes the nonstructural hepatitis B virus X (HBx) protein that is conserved among mammalian hepadnaviruses, suggesting an important function.