Virologic response was compared between the two treatment groups

Virologic response was compared between the two treatment groups. Results: At baseline, all patients had genotypic resistances: YMDD-motif mutations, 80; YMDD mutations with adefovir- or entecavir-resistant

mutations, 25 and 32, respectively; YMDD mutations with adefovir- and entecavir-resistant mutations, 14. Median serum HBV DNA level was higher, and virologic breakthrough to last antiviral agents before enrollment (last drugs) was more frequent in teno-fovir Nutlin-3a price group than in maintenance group (all, P=0.001). Overall cumulative virologic response rates were higher in tenofovir group than in maintenance group (64.9% vs. 15.3%, 76.5% vs. 19.9%, 85.9% vs. 38.9% at 6, 12, 18 months, respectively; P<0.001). In subgroup analysis according to virologic breakthrough or suboptimal response to last drugs, cumulative virologic response rate was higher in tenofovir group than in maintenance group (all, P<0.001). In mono-resistance (YMDD mutations) or multi-drug resistance (YMDD mutations ± adefo-vir-resistant mutations ± entecavir-resistant CP-868596 mutations) subgroup analysis, cumulative virologic response rate was also higher in tenofovir group than in maintenance group (P<0.001, P=0.001; respectively). Regardless of final drugs prior to enrollment, cumulative virologic response rate was higher in tenofovir group than in maintenance group (P<0.001 in lami-vudine+adefovir

and telbivudine+adefovir, P=0.024 in entecavir). Conclusion: Tenofovir monotherapy is an effective rescue therapy for patients with this website antiviral drug resistance. Disclosures: The following people have nothing

to disclose: Tae Jung Yun, Soon Ho Um, Chang Ho Jung, Tae Hyung Kim, Seok Bae Yoon, Sun Young Yim, Bora Keum, Yeon Seok Seo, Hyung Joon Yim, Yoon Tae Jeen, Hong Sik Lee, Hoon Jai Chun, Chang Duck Kim, Ho Sang Ryu Background: Factors relevant to relapse in a long-term follow-up after cessation of nucleus(t)ide analogues (NUCs) treatment have yet to be identified. We aimed to determine off-therapy durability in response to telbivudine (LdT) and lamivudine (LAM) by analyzing the factors associated with the relapse. Methods: 60 NUCs-naïve CHB patients treated with LdT (n = 26) or LAM (n = 34) who achieved indication for off-therapy, had consolidation therapy, and followed by cessation of treatment were followed for up to 10-years. HBV-DNA, viral serology and biochemistries were periodically (every 1-3 months) determined at baseline, on-treatment, and after off-therapy. COX model was used to predict the risk of relapse. Results: Relapse occurred in 50.0% of the 60 patients during follow-up for a median of 115-months (range 3-120). 90.0% of the relapses occurred in < 4-years. Cumulative relapse rates in HBeAg-positive (n = 46) and -negative (n = 14) patients were 30.8% and 72.7%, respectively (P < 0.01).

[6] Similar programs could be implemented globally especially whe

[6] Similar programs could be implemented globally especially where subspecialty referral is impossible. Broadening the scope to new HCV providers will be dependent on a simpler algorithm of care, which seems highly likely in the near future. Efforts should be made to create policy not only to educate nonspecialist providers in HCV care, but also to incentivize these physicians to treat patients infected with HCV as more efficient therapies evolve. In conclusion,

there has been considerable enthusiasm regarding the use of DAAs to treat HCV. Efforts are being made through increased awareness and recommendations for age-based screening to identify more patients with HCV. However, the current complexity

of treatment is a significant therapeutic barrier. Directing resources to BMS-907351 in vivo support drug development plans to simplify treatment algorithms, even at the expense of optimized SVR rates, in addition to taking creative approaches to expand HCV care into a primary care setting are essential steps in ultimate viral eradication. Complex, individualized care is not the solution for control of the HCV epidemic. True evolution of therapy will likely require Navitoclax price broadly available, simple, and accessible treatment. Andrew Aronsohn, M.D.Donald Jensen, M.D. “
“A woman, aged 48 years, had an upper abdominal ultrasound study that showed a 3 cm hypoechoic mass in segment III of the liver. Four years previously, she had been treated for breast cancer. A contrast-enhanced computed tomography (CT) scan confirmed the presence of a solid mass with enhancement in all three phases. The differential diagnosis included hepatocellular

carcinoma, hepatic adenoma, a hypervascular metastasis and an atypical hemangioma. However, a positron emission tomography scan with CT (PET/CT) using 18F-fluorodeoxyglucose as well as a 99mtechnecium-labeled red blood cell scan were negative. Because of this, the preferred diagnosis became focal nodular hyperplasia. Additional investigations included a 99mtechnecium-sulphur colloid scan with CT (SPECT/CT) and a 99mtechnecium-mebrofenin scan. Both scans showed that the find more lesion was photopenic for the tracers consistent with the absence of both Kupffer cells and functioning hepatocytes. This appeared to exclude both focal nodular hyperplasia and hepatic adenoma. The final investigation was a regional 11C-acetate PET/CT (BiographTM 40 LSO HI-REZ) performed 30 minutes after the administration of 11C-acetate (555MBq). The lesion in segment III showed markedly increased 11C-acetate metabolism with a lesion to liver standard uptake value of 2.8 (Figure 1). This result was not typical for hepatocellular carcinoma and raised the possibility of an angiomyolipoma in the liver.

Nonetheless, lithium carbonate has been demonstrated to provide s

Nonetheless, lithium carbonate has been demonstrated to provide significant benefit in the treatment of CCH. Its efficacy for treating CCH has been demonstrated in the investigation discussed in “First-Line Therapy” and in a study Venetoclax research buy of 8 additional CCH patients.34,38 In the latter study, all

8 patients had at least a 75% improvement within the first 2 weeks of therapy. However, only 1 of 3 who were followed long-term had continued improvement after 18 months of therapy. The evidence for the utility of lithium carbonate for the treatment of ECH is less clear, with generally small studies providing contradictory results.34,38,39 Lithium carbonate doses of 600 mg to 900 mg per day are typically needed to obtain target therapeutic serum lithium levels of 0.4 to 0.8 mEq/L. Lithium serum levels, renal function, and thyroid function should

be monitored during lithium therapy. Common AEs to lithium include diarrhea, tremor and polyuria. Symptoms and signs of toxicity include nausea, vomiting, diarrhea, confusion, nystagmus, extrapyramidal signs, ataxia, and seizures. Topiramate, in doses ranging from 50 mg to 200 mg per day, is considered second-line therapy for CH prophylaxis. Although we have designated topiramate as second-line therapy, consistent with the Grade B recommendation in the European Federation of Neurological Societies guidelines, topiramate use for CH prophylaxis has been investigated in open-label studies only.40-42 Common AEs to topiramate include cognitive dysfunction, paresthesias, alteration in taste, weight loss, fatigue, click here and dizziness. Patients with a history of nephrolithiasis Regorafenib cost should not receive topiramate because of an increased risk of recurrent stones while taking this medication. Third-Line Therapy.— Other therapies that may be effective for maintenance cluster prophylaxis include methysergide, valproic acid, melatonin, gabapentin, baclofen, clonidine, and botulinum toxin. Although methysergide is likely effective for preventing CH, it is not available in the USA and long-term use is associated with fibrotic complications. Thus, we cannot recommend its use. Valproic

acid has been shown to provide benefit in open-label and retrospective studies only.43,44 A double-blind placebo-controlled study of sodium valproate did not support its efficacy; however, this may have been due to an exceedingly high response rate of 62% in the placebo group.45 Effective doses range from 500 mg to 2000 mg daily in divided doses. Common AEs include weight gain, fatigue, tremor, hair loss, and nausea. Monitoring with complete blood counts and liver function tests are necessary during valproic acid therapy. Limited evidence supports the use of melatonin for cluster prophylaxis. In a double-blind, placebo-controlled trial of 10 mg melatonin, 5 of 10 subjects randomized to melatonin had cluster remission within 5 days while none of the 10 subjects taking placebo went into remission.

Prednisolone administration attenuated ConA- and α-GalCer-induced

Prednisolone administration attenuated ConA- and α-GalCer-induced hepatitis and systemic inflammatory responses. Treating mice with prednisolone also check details suppressed inflammatory responses in a model of hepatotoxin (CCl4)-induced hepatitis, but surprisingly exacerbated

liver injury and delayed liver repair. In addition, administration of prednisolone also enhanced acetaminophen-, ethanol-, or ethanol plus CCl4-induced liver injury. Immunohistochemical and flow cytometric analyses demonstrated that prednisolone treatment inhibited hepatic macrophage and neutrophil infiltration in CCl4-induced hepatitis and suppressed their phagocytic activities in vivo and in vitro. Macrophage and/or neutrophil depletion aggravated CCl4-induced liver injury and impeded liver regeneration. Finally, conditional disruption of glucocorticoid receptor in macrophages and neutrophils abolished prednisolone-mediated exacerbation of hepatotoxin-induced liver injury. Conclusion: Prednisolone treatment prevents T/NKT cell hepatitis but exacerbates hepatotoxin-induced liver injury by inhibiting macrophage- and neutrophil-mediated phagocytic and hepatic regenerative

functions. These findings may not only increase our understanding of NVP-BKM120 order the steroid treatment mechanism but also help us to better manage steroid therapy in liver diseases. (Hepatology 2014;59:1094–1106) “
“The efficacy of treatment with multispecies probiotics on irritable bowel syndrome (IBS) symptoms and the alterations of gut microbiota in patients who have taken probiotics were investigated. This randomized, double-blind,

placebo-controlled trial involved 49 IBS patients (probiotics: 25, placebo: 24) diagnosed according to the Rome III criteria. Patients were randomly assigned to two groups: either to receive multispecies probiotics (a mixture of Bifidobacterium longum, B. bifidum, B. lactis, Lactobacillus acidophilus, L. rhamnosus, and Streptococcus thermophilus) twice a day for 4 weeks or to receive a placebo twice a day for 4 weeks. The primary efficacy end-point was the proportion of participants whose IBS symptoms were substantially relieved at week 4. Secondary end-points were the intensity of abdominal selleck products pain/discomfort, bloating, stool frequency/consistency, alterations in fecal microflora over the 4 weeks. Fecal microflora were analyzed in 34 patients (probiotics: 17, placebo: 17) by quantitative real-time polymerase chain reaction assays. The proportion of patients whose IBS symptoms were substantially relieved at week 4 was significantly higher in the probiotics group than in the placebo group: 68.0% (17/25) versus 37.5% (9/24) (P < 0.05). Secondary end-points such as improvement in abdominal pain/discomfort and bloating occurred in the probiotics group but not in the placebo group. Fecal analysis revealed that B. lactis, L.

pylori, may access the

central nervous system (CNS) throu

pylori, may access the

central nervous system (CNS) through blood, the nasal olfactory pathways, and the gastrointestinal system, especially in regard to the fact that gastrointestinal immune system (GIS) represents a primary immune organ with specialized immunoregulatory and anti-inflammatory functions. H. pylori would be capable of inducing humoral and cellular immune responses that, owing to the sharing of homologous epitopes (molecular mimicry), cross-react with CNS components thereby contributing and possibly perpetuating neural tissue damage. Thus, H. pylori would be implicated in the development and regulation of several autoimmune and degenerative diseases of the CNS. Shiota et al. [35] found no association between Gemcitabine supplier H. pylori infection and Alzheimer’s

BKM120 mw disease in a Japanese cohort of patients. In their commentary, Kountouras et al. [36] stressed out that this study was underpowered, owing to small number of patients enrolled and relatively high H. pylori infection prevalence in general Japanese population; thus, the study would not be comparable to European studies indicating the association between H. pylori infection and Alzheimer’s disease. Based on the studies published previously, several authors hypothesized that H. pylori infection could indirectly affect neural and brain tissue by disrupting the brain–neural barrier and blood–brain barrier, by release of numerous proinflammatory cytokines (IL-1β, IL-6, TNF-α), acting at the distance and being involved in pathogenesis of inflammatory demyelinating neuropathies [37], and epilepsy [38]. The underlying mechanism of a probable selleck inhibitor association between H. pylori infection

and epilepsy would be the action of TNF-α, leading to upregulation of matrix metalloproteinases that cause the disruption of the blood brain barrier. A high prevalence of H. pylori infection was reported by several authors in patients with diabetes mellitus (DM), but the clinical consequences in terms of metabolic control seem to be low [2]. In a review article [39], Albaker stressed out that the association between DM and H. pylori infection remains controversial, although some studies showed a high prevalence of this infection in both Type 1 DM and Type 2 DM. Although some studies spoke in favor of an association of CagA+ virulent strains with microangiopathy, neuropathy, and microalbuminuria in Type 2 diabetic patients, the results of The Freemantle Diabetes Study did not confirm the CagA seropositivity as a risk factor for chronic vascular complications of Type 2 DM [40]. Metabolic syndrome is one of the most prevalent global health problems that predisposes to Type 2 DM and it is linked to insulin resistance. A very interesting study on 462 elderly Koreans supported the hypothesis that H. pylori infection plays a role in promoting atherosclerosis by modifying lipid metabolism [41]. In a systematic review, Polyzos et al.

Disclosures: R Todd Stravitz – Grant/Research Support: Exalenz B

Disclosures: R. Todd Stravitz – Grant/Research Support: Exalenz Biosciences, LTD William M. Lee – Consulting: Eli Lilly, Novartis; Grant/Research Support: Gilead, Roche, Vertex, BI, Anadys, BMS, merck; Speaking and Teaching: Merck The following people have nothing to disclose: Caitlyn Ellerbe, Valerie Durkalski, Adrian Reuben Objective: Whether the use of mTOR inhibitor corticosteroids following hepatoportoenterostomy

(HPE) is effective and/or safe in improving clinical endpoints in infants with biliary atresia (BA) is unknown. We conducted the Steroids in Biliary Atresia Randomized Trial (START) to determine whether the addition of high dose corticosteroids is superior to surgical therapy alone. Methods: Subjects were enrolled from 14 US centers participating in the NIDDK-sponsored ChiLDREN Network and randomized to receive I. V. methylprednisolone/oral prednisolone (4 mg/kg/day x 2 wk, 2 mg/kg x 2 wk, followed by a tapering protocol over the next 9 wk) or placebo within 72 hours of HPE. All infants received post-operative care including antibiotic prophylaxis,

ursodeoxycholic acid, fat-soluble vitamins and standardized nutrition according to guidelines developed for the trial, and were followed until 2 years of age. this website The primary endpoint was the percent of subjects with serum total bilirubin <1.5 mg/dL with their native liver at 6 months after HPE (improved bile drainage). An intent-to-treat analysis was performed, using multiple logistic regression. Treatment differences in transplantfree survival over the entire period were assessed using a Cox model. Results: 140 BA subjects were randomized (70 per group); 91% achieved the pre-defined study endpoints. Demographics and baseline characteristics were comparable between the two groups: mean age at randomization was 2.3 months, mean total bilirubin prior to HPE was 7.7mg/dL, and 5 subjects

had BASM. Bile drainage was not significantly improved by corticosteroids at 6 months post-HPE (primary endpoint; steroid 58.6% vs placebo 48.6%, adjusted relative risk [RR] [95% CI]: 1.14 [0.83, 1.57], P=0.43), or at 24 months of age (steroid: 49.4% vs placebo: 39.8%, adjusted hazard ratio [HR] [95% CI]: 0.8 [0.5, 1.2], P=0.29). Transplant-free selleck screening library survival at 24 months was similar between groups (steroids: 58.7% vs placebo: 59.4%, adjusted HR [95% CI]: 1.0, [0.6, 1.8], P=0.99). There were no significant treatment differences in important safety outcomes: % of subjects with SAEs (steroids 81.4% vs placebo 80%, P=1.0), weight and height Z-scores over the study period (P=0.16 and 0.28, respectively), number of infectious SAEs per patient (RR=1.12, 95% CI [0.86, 1.44], P=0.40), time to first episode of cholangitis (P=0.63), or number of episodes of cholangitis per patient (P=0.64).

However, no information exists on the long-term efficacy and safe

However, no information exists on the long-term efficacy and safety of terlipressin therapy in type 2 HRS.13 The influence of terlipressin on cerebral blood flow, especially in patients with cirrhosis with hepatic encephalopathy needs further study. The patient under discussion had acute variceal bleeding and probably type 1 HRS. Such patients should be admitted to the intensive JNK inhibitor libraries care unit for continuous

monitoring of heart rate, mean arterial pressure, and central venous pressure. Because the hemoglobin was 8.6 g/dL, this patient did not require any red cell transfusion; however, measurement of the hemoglobin after volume resuscitation will give a more accurate assessment of the need for red cell transfusions. Prophylactic parenteral ceftriaxone 1 g intravenously daily for 5 days should be given in view of advanced liver

disease. Combination of endoscopic band ligation and vasoactive drug is the treatment of choice for treatment of AVB. Terlipressin should be started after excluding any obvious contraindication ICG-001 price and at least 30 minutes before endoscopy at a dose of 2 mg every 4 hours. Endoscopic band ligation should be carried out when the patient is hemodynamically stable, and within 12 hours of admission. As this patient also had Type 1 HRS, terlipressin should be supplemented with albumin at a dose of 1 g/kg body weight to maintain the central venous pressure at 8-12 mmHg. The patient should be monitored regularly for any side effects of terlipressin. The hematocrit, serum creatinine, and serum sodium should be monitored daily to determine control

of bleed and hyponatremia. This patient’s baseline serum creatinine, bilirubin, and absence of alcoholic hepatitis favor response to terlipressin. Based on day 3 serum creatinine levels, the dose of terlipressin could be decreased to 1 mg every 4 hours if the level is <1.5 mg/dL or 30% lower than baseline. If the decrease in serum creatinine is not greater than 30% compared to baseline, terlipressin at a dose of 2 mg every 4 hours is continued until the serum creatinine is <1.5 mg/dL, or for a maximum of 15 days of therapy. The patient requires 5 days of therapy with terlipressin in view MCE公司 of the variceal bleed. If the serum creatinine increases on treatment, terlipressin should be continued after 5 days. Finally, this patient should be listed for liver transplantation as definitive therapy. “
“Balloon-occluded retrograde transvenous obliteration (B-RTO) is recognized as the standard therapy for patients with gastric fundal varices in Japan; however, the procedure is difficult when drainage veins other than the gastrorenal shunt developed. The efficacy and safety of B-RTO using a microballoon catheter for such patients were evaluated. The subjects were 99 patients with gastric fundal varices who fulfilled the criteria for receiving endoscopic and/or interventional therapies.

cDNA was amplified using TaqMan Fast Universal PCR Master Mix (Ap

cDNA was amplified using TaqMan Fast Universal PCR Master Mix (Applied Biosystems) Birinapant supplier with validated gene-specific assays (Applied Biosystems) for CCL11 (eotaxin-1), CCL24 (eotaxin-2), and β-actin on an Applied Biosystems 7500 Fast Real-Time PCR System. RNA expression was reported relative to messenger RNA (mRNA) expression of β-actin for each sample. Serum protein levels of CCL11 and CCL24 were quantified using CCL11 and CCL24 DuoSet ELISA kits (R&D Systems, Minneapolis, MN) following the manufacturer’s protocols. Eosinophils were depleted by pretreating female Balb/cJ mice with 25 μg of sodium azide-free and low endotoxin-tested Siglec-F mAb (E50-240, BD Pharmingen) or isotype

control (Rat IgG2a,κ, R35-95, BD Pharmingen) intraperitoneally in 100 μL of sterile PBS, 24 hours prior to halothane treatment. Since the depleting antibody (anti-Siglec-F) was the same clone as the antibody used to detect eosinophils (PE-anti-Siglec-F), it was anticipated that the mean fluorescent intensity (MFI) of PE on Siglec-F+ cells from the livers of anti-Siglec-F-pretreated mice would decrease in part without the cells being depleted due to competitive binding. To ensure the magnitude of anti-Siglec-F depletion

was check details not overestimated by flow cytometry, all CD11c− CD11b+ Gr-1low Siglec-F+ and CD11c− CD11b+ Gr-1high Siglec-Flow/neg cells were back-gated to forward- and sidescatter area plots to demonstrate similar granularity and size as the eosinophils and neutrophils isolated from isotype-treated

mice. Similarly, neutrophils were depleted by pretreating female Balb/cJ mice with 10, 20, 25, or 50 μg of sodium azide-free and low endotoxin-tested Gr-1 antibody (RB6-8C5, Bio X Cell, West Lebanon, NH) or rat IgG2b MCE isotype control (LTF-2, Bio X Cell) intraperitoneally in 100 μL of sterile PBS, 24 hours prior to halothane treatment. Hepatic eosinophils and neutrophils were quantified by flow cytometry as outlined above. Liver homogenates were prepared and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) immunoblot analysis was performed as described,21 except that TFA31 and β-tubulin (Clone AA2, EMD Millipore, Billerica, MA) antibodies were used at 1/2,000 and 1/5,000 dilutions, respectively. Detection of mouse MBP in fixed tissue sections was performed using the established method with rat antimouse MBP (MT-14.7, provided by Drs. Nancy and James Lee at Mayo Clinic Arizona, Scottsdale, AZ) or Rat IgG1,κ isotype control (ab18407, Abcam, Cambridge, MA),32 with some modifications (see Supporting Material for details). All data presented are reported as mean ± standard error of the mean (SEM). Statistical significance between two groups was determined by two-tailed Student’s t test, while statistical differences between multiple groups were determined by one-way analysis of variance with Newman-Keuls post-test analysis. Differences were considered significant when P < 0.05.

Results: The database comprises 3895 PBC patients of which 2924 U

Results: The database comprises 3895 PBC patients of which 2924 U D C A-treated patients with available lab measurements; mean age of 52.3 (±12.2) yrs, female: 91%, AMA+: 88%. Median follow up time Selleckchem JNK inhibitor was 7 (IQR 3-11) yrs. LTX-free survival was significantly better for patients responding to treatment as assessed by all of the models. Rotterdam and Paris I criteria were the most powerful predictors, hazard ratio (HR) respectively: 3.92 (3.17-4.85) and 4.25 (3.53-5.11) for non-responders versus responders. According to Rotterdam and Paris I criteria 10-yrs survival was 84.1 % and 88.1% for responders and 42.7% and 50.1% for nonresponders.

Cox regression analysis showed Barcelona, Paris I, Rotterdam and Toronto criteria were independently associated with LTX-free survival (c-statistics: 0.78 (0.74-0.81)). 38% of patients responded according to all criteria (10-yrs survival: 96.7%, sensitivity: 88.6%), while 10.4% did not respond according to any criteria (10-yrs survival: 58.0%, HR=7.7 (5.510.7)). Conclusions: This analysis of a large pooled UDCAtreated PBC cohort

confirms the prognostic value of previously proposed response criteria. Paris I and Rotterdam were the most powerful predictors. Four of the five criteria Gemcitabine solubility dmso contribute independently in a combined analysis of prognostic significance, suggesting that the optimal response criteria await to be defined. Barcelona (normal ALP or >40% decrease) Paris I (ALP≦ 3xULN, AST≦ 2xULN, normal bili) Rotterdam (normaliation of abnormal bili and/or albumin) Toronto (ALP<1.67xULN) Paris II (ALP≦ 1.5xULN, AST≦ 1.5xULN, normal bili) HR no response vs response (95% CI) 1.95 (1.62-2.34) 上海皓元医药股份有限公司 4.25(3.53-5.11) 3.92 (3.17-4.85) 2.60(2.13-3.17) 2.99 (2.40-3.71) at 10 year sensitivity specificity PPV NPV 63% 59% 68% 53% 71% 72% 75% 69% 83% 59% 72% 73% 66% 60% 54% 71% 45% 84% 77% 57% c-statistics (95% CI) 0.69 (0.66-0.72) 0.76 (0.74-0.78) 0.74 (0.71-0.78) 0.71 (0.69-0.74) 0.71 (0.68-0.73) Disclosures: Gideon M. Hirschfield – Advisory Committees or Review Panels: Centocor/J&J, Medigene, Intercept, Falk Pharma; Consulting: Lumena,

Intercept Harry L. Janssen – Consulting: Abbott, Bristol Myers Squibb, Debio, Gilead Sciences, Merck, Medtronic, Novartis, Roche, Santaris; Grant/Research Support: Anadys, Bristol Myers Squibb, Gilead Sciences, Innogenetics, Kirin, Merck, Medtronic, Novartis, Roche, Santaris Cyriel Y. Ponsioen – Consulting: AbbVIE; Grant/Research Support: AbbVIE, Schering Plough, Dr. Falk Pharma, Tramedico Netherlands Marlyn J. Mayo – Consulting: Mitsubishi, Regeneron; Grant/Research Support: Intercept, Lumena Jayant A. Talwalkar – Consulting: Lumena; Grant/Research Support: Intercept, Salix, Gilead Frederik Nevens – Grant/Research Support: Ipsen, Roche, MSD, Astellas, CAF Andrew L. Mason – Grant/Research Support: Abbott, Gilead Kris V.

Ig) The recombinant plasmid was transfected into HEK293 cells us

Ig). The recombinant plasmid was transfected into HEK293 cells using Lipofectamine 2000 (Invitrogen), and then VSIG4.Ig was purified from the culture supernatant using HiTrap Protein G HP Columns according to

the manufacturer’s recommendations (GE Healthcare). Mice were injected intravenously with either a lethal dose (25-30 mg/kg) or a sublethal dose (15 mg/kg) of ConA (Sigma-Aldrich). Serum alanine aminotransferase (ALT) levels were measured using a transaminase kit (Asan Pharmaceutical) according to the manufacturer’s XL184 concentration instructions. For adoptive transfer of KCs, KCs (3 × 106) isolated from VSIG4 WT or KO mice were injected intravenously into VSIG4 KO mice by way of the tail vein. Mouse livers were fixed in 4% paraformaldehyde, dehydrated, and embedded in paraffin. 5-μm sections were stained with hematoxylin and eosin using a standard procedure and analyzed by light microscopy. Liver MNCs were isolated by the collagenase digestion method with some modification.12–13 Briefly, mouse liver was perfused in situ with Hank’s buffered salt solution (HBSS) containing 0.025% collagenase, this website removed, and passed through 70-μm stainless

steel mesh. Initial cell suspension that was resuspended in 40% Percoll was overlaid onto 70% Percoll and centrifuged at 750g for 20 minutes. MNCs were collected from the interface. For purification of KCs, liver MNC suspension was overlaid onto Percoll gradient (25%/50%), and centrifuged at 1,800g for 30 minutes. MCE KC-enriched MNCs located in the interface were harvested and stained with FITC-conjugated

anti-F4/80 (clone BM8, eBioscience). F4/80 positive KCs were purified using anti-FITC Microbeads (Miltenyi Biotech) according to the manufacturer’s protocols. KC isolates were 95% pure and KCs were the only cell fraction expressing VSIG4 among liver APCs (Supporting Fig. 1). For purification of splenic DCs, splenocytes were incubated with anti-CD11c Microbeads (Miltenyi Biotech) and enriched by the MACS system according to the manufacturer’s protocols. For purification of liver T- and NKT-cells, liver MNCs were stained with FITC-conjugated-NK1.1 mAb and PE Cy5-conjugated anti-TCR-β mAb, and then TCR-β+NK1.1+ NKT and TCR-β+NK1.1− T-cells were sorted using a BD FACSAria. T-cells (105) were plated in 96-well flat-bottom plates that were precoated with indicated concentrations of mouse anti-CD3e antibody (145-2C11) together with VSIG4.Ig or control Ig (10 μg/mL). [3H]-Thymidine (1 μCi/well) was added 16 hours prior to harvesting of the cultures. [3H]-Thymidine incorporation was measured with a Wallac MicroBeta TriLux Liquid Scintillation counter (PerkinElmer). In some experiments, purified DO11.10 T-cells (105) were incubated with KCs (1-10 × 103) in the presence of OVA323-339 (10 μg/mL) for 3 days before [3H]-thymidine incorporation. For liver NKT-cell tolerance induction, mice (Balb/c background) were injected intraperitoneally with α-GalCer (0.