ASMase+/+ PMH pre-treated

with U18666A increased lysosoma

ASMase+/+ PMH pre-treated

with U18666A increased lysosomal cholesterol levels, impaired mitophagy (LAMP-GFP/ mtKeima colocalization) and sensitized to APAP-induced cell death. Conversely, 25-HC reversed the lysosomal cholesterol accumulation induced by U18666A, improved mitophagy and protected against APAP-induced cell death. Moreover, 25-HC abolished the susceptibility of ASMase−/− PMH to APAP exposure. Treatment with Ca-074Me to inhibit cathepsin B did not affect APAP susceptibility of ASMase-/- PMH. Conclusions: Our findings GPCR Compound Library suggest that the underlying status of the pathway leading to mitophagy may be an important risk factor for APAP hepatotoxicity. The findings may have implications for patients with lysosomal storage diseases who may exhibit susceptibility to APAP-induced liver injury. Disclosures: Neil Kaplowitz – Consulting:

GlaxoSmithKline, JNJ, Merck, Novartis, Hepregen, Takeda, Otsuka, Pfizer, Geron, Daiichi-Sanyo; Independent Contractor: Acetaminophen Litigation The following people have nothing to disclose: Anna Baulies, Susana Nuñez, Vicent Ribas, Sandra Torres, Laura Martinez, Carmen Garcia-Ruiz, Jose Fernan-dez-Checa Background/Aims: Concanavalin A (ConA)-induced AT9283 liver injury is an established model of T cell-mediated hepatitis. CD4 T cells, NKT cells and Kupffer cells all were reported to contribute to ConA-induced hepatitis. We and others have shown that hepatic stellate cells (HSCs) play a major role in hepatic inflammation and immune reactions. We recently developed a novel HSC-depleted

mouse, which is resistant to ischemia/ reperfusion- and endotoxin-induced liver injury. Here, we investigated mechanisms of ConA-induced hepatitis in the HSC-depleted mouse. Methods: HSC-depleted and HSC-sufficient mice (n=6 each) were injected 20 mg/kg ConA or vehicle (PBS) (i.v.) and sacrificed 6h later. H/E-stained liver sections were examined for histopathology and serum ALT measured. mRNA expression of IFNp, TNFα, IL10, CXCL1 and CXCL10 was determined via qRT-PCR. In vitro HSCs were incubated in a medium containing 10 μg/ml ConA or vehicle for 4 and 8h and the MCE公司 medium was transferred to hepatocytes. Viability of hepatocytes was examined by phase-contrast microscopy and TUNEL staining. mRNA expression of INFp, IRF1 and CXCL1 was measured in ConA-stimulated HSCs. Generation of reactive oxygen species (ROS) in HSCs and oxidative stress in hepatocytes was determined via DCFDA fluorescence. Results: ConA treatment caused profound liver injury (primarily in zone 2) accompanied by inflammatory infiltration, sinusoidal congestion, and increased expression of IFN, TNFα, CXCL1 and CXCL10, and JNK1-MAPK activation in HSC-sufficient mice but not in HSC-depleted mice. In contrast, IL10 expression increased in ConA-treated HSC-depleted mice but not in HSC-sufficient mice.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>