02.10.2015 Views

studies

2015SupplementFULLTEXT

2015SupplementFULLTEXT

SHOW MORE
SHOW LESS
  • No tags were found...

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

HEPATOLOGY, VOLUME 62, NUMBER 1 (SUPPL) AASLD ABSTRACTS 647A<br />

rates by regulating endocrine hormone levels such as thyroid<br />

hormone levels. Fasting is an adaptive mechanism developed<br />

in mammals for nutrient depravation. Fasting is characterized<br />

with increase in free fatty acid levels in serum and liver causing<br />

hepatic steatosis along with several other physiological<br />

changes. These physiological changes in fatty acid metabolism<br />

during fasting are similar to diabetes condition. In this study<br />

we aim to identify role of Tgr5 in fatty acid metabolism using<br />

fasting model. Methods: Male C57BL/6J (WT) and Tgr5 knock<br />

out (Tgr5-/-) mice in pure C57BL/6J background were either<br />

fed ad libitum or fasted for 24 hrs. Lipid profiles of serum, liver<br />

and adipose tissues were analyzed. Quantitative real-time PCR<br />

was used to assay mRNA expression levels of the genes in bile<br />

acid, cholesterol and lipid metabolism. A Tgr5 specific agonist<br />

INT-777 (Intercept Pharma) was used to activate Tgr5 signaling<br />

in mice. Results: Fasting increased hepatic lipid accumulation<br />

in both WT and Tgr5-/- mice. Lack of Tgr5 attenuated fasting-induced<br />

hepatic triglyceride and fatty acids accumulation compared<br />

to WT mice, without changes in adipose tissue lipolysis.<br />

Gene expression analysis showed Tgr5-/- mice have increased<br />

expression of hepatic lipolysis genes such as Cpt1, Pgc-1α<br />

and autophagy gene LC3II levels. Comprehensive laboratory<br />

animal monitoring system (CLAMS) metabolic <strong>studies</strong> showed<br />

Tgr5-/- mice have increased physical activity and energy<br />

expenditure. Tgr5-/- mice also showed decrease in expression<br />

of hepatic fatty acid transporter Cd36 and decreased hepatic<br />

fatty acid uptake. Additionally, Tgr5-/- mice had increased<br />

hepatic Stat5 activation, which is known to regulate hepatic<br />

lipid metabolism. Moreover, a Tgr5 agonist INT-777 treatment<br />

increased hepatic Cd36 expression and decreased hepatic<br />

Lc3II levels in fasted mice. Conclusion: Although Tgr5 is not<br />

expressed in hepatocytes, Tgr5 may play a role in regulation<br />

of hepatic fatty acid metabolism during fasting. Lack of<br />

Tgr5 attenuates fasting-induced hepatic steatosis by decreasing<br />

hepatic fatty acid uptake and increasing hepatic lipolysis.<br />

Disclosures:<br />

The following authors have nothing to disclose: Ajay C. Donepudi, Shannon M.<br />

Boehme, John Chiang<br />

886<br />

Hepatic NOD-like receptors, pyrin domain-containing<br />

3 (NLRP3) inflammasome activation is associated with<br />

histological severity in patients with nonalcoholic fatty<br />

liver disease<br />

Hironori Mitsuyoshi, Kohichiroh Yasui, Tasuku Hara, Hiroyoshi<br />

Taketani, Hiroshi Ishiba, Akira Okajima, Yuya Seko, Atsushi<br />

Umemura, Taichiro Nishikawa, Kanji Yamaguchi, Michihisa Moriguchi,<br />

Yoshio Sumida, Masahito Minami, Yoshito Itoh; Kyoto Prefectural<br />

University of Medicine, Kyoto, Japan<br />

Background and Aims: Nonalcoholic fatty liver disease<br />

(NAFLD) is the hepatic manifestation of metabolic syndrome,<br />

which is associated with dysregulated inflammasome activation<br />

caused by obesity. Inflammasomes are intracellular multiprotein<br />

complexes, comprising nucleotide-oligomerization and binding<br />

domain (NOD)-like receptors, the apoptosis-associated specklike<br />

protein containing a CARD (ASC), and procaspase-1.<br />

Assembly of these components in response to metabolic stress<br />

results in caspase-1 activation, leading to interleukin-1 beta<br />

(IL-1β) and IL-18 activation, which exacerbates inflammation<br />

in systemic organs, including the liver. Here, we examined the<br />

role of inflammasomes in the development of NAFLD. Methods:<br />

Biopsy-proven patients with NAFLD (n = 91) were enrolled.<br />

Liver histology was assessed according to the NAFLD activity<br />

score (NAS). Hepatic (n = 91) and blood (n = 37) levels of the<br />

NOD-like receptor family, pyrin domain-containing 3 (NLRP3),<br />

ASC, procaspase-1, IL-1β, and IL-18 were quantified by realtime<br />

polymerase chain reaction (PCR). Adiponutrin polymorphisms<br />

(rs738409, C>G) were determined by TaqMan PCR.<br />

Serum IL-1β and IL-18 levels were measured by ELISA and<br />

liver tissue caspase-1 expression was evaluated by immunostaining.<br />

Results: Hepatic mRNA levels of NLRP3, ASC, procaspase-1,<br />

IL-1β, and IL-18 were significantly higher in patients<br />

with NAFLD compared with control livers. Blood procaspase-1<br />

mRNA levels were significantly higher in NAFLD patients compared<br />

with healthy controls. In contrast to the blood mRNA<br />

levels, hepatic mRNA levels of NLRP3 inflammasome components<br />

were significantly associated with adiponutrin G alleles.<br />

Hepatic procaspase-1 and IL-1β mRNA levels correlated significantly<br />

with lobular inflammation, hepatocyte ballooning,<br />

and total NAS. Serum IL-18 levels were significantly higher<br />

in NAFLD patients compared with controls, while IL-1β levels<br />

exhibited a non-significant increase. Serum IL-1β and IL-18 levels<br />

were significantly correlated with steatosis, total NAS and<br />

serum transaminase levels. Caspase-1-positive cells were scattered<br />

within the portal tracts, inflammatory foci, and ballooning<br />

hepatocytes. Immunofluorescence staining showed colocalization<br />

of caspase-1 with the macrophage marker CD68. Conclusions:<br />

Our results suggest that NLRP3 inflammasomes are<br />

primed in the liver in NAFLD patients and that this process is<br />

influenced by adiponutrin genotypes. Furthermore, our results<br />

indicate that NLRP3 inflammasomes are activated exclusively<br />

in Kupffer cells or infiltrating macrophages, leading to histological<br />

NAFLD progression through IL-1β and IL-18 production.<br />

Disclosures:<br />

Yoshito Itoh - Grant/Research Support: MSD, Chugai Phamaceutical CO., Bristol-Myers<br />

Squibb, Dainippon Sumitomo, Otsuka, Takeda, Fujifilm medical, Eisai,<br />

Mitsubishi Tanabe, AstraZeneca; Speaking and Teaching: MSD<br />

The following authors have nothing to disclose: Hironori Mitsuyoshi, Kohichiroh<br />

Yasui, Tasuku Hara, Hiroyoshi Taketani, Hiroshi Ishiba, Akira Okajima, Yuya<br />

Seko, Atsushi Umemura, Taichiro Nishikawa, Kanji Yamaguchi, Michihisa Moriguchi,<br />

Yoshio Sumida, Masahito Minami<br />

887<br />

Liver MicroRNA-21 is Overexpressed in Non Alcoholic<br />

Steatohepatitis in Patients and Contributes to this Disease<br />

In Two Mouse Models by restoring PPARα expression<br />

Xavier Loyer 1 , Valerie Paradis 3 , Carole Hénique 1 , Anne-Clémence<br />

Vion 1 , Nathalie Colnot 3 , Coralie L. Guerin 1 , Cécile Devue 1 , Sissi<br />

On 1 , Jérémy Scetbun 1 , Mélissa Romain 1 , Jean-Louis Paul 4 , Marc<br />

E. Rothenberg 5 , Patrick Marcellin 2 , Francois Durand 2 , Pierre<br />

Bedossa 3 , Carina Prip-Buus 6 , Eric Baugé 7 , Bart Staels 7 , Chantal<br />

Boulanger 1 , Alain Tedgui 1 , Pierre-Emmanuel Rautou 1,2 ; 1 INSERM,<br />

U970, Paris Cardiovascular Research Center - PARCC@HEGP,<br />

Paris, France; 2 Hepatology, Hôpital Beaujon, APHP, Clichy,<br />

France; 3 Pathology, Hôpital Beaujon, APHP, Clichy, France; 4 Service<br />

de Biochimie, Hôpital Européen Georges Pompidou, AP-HP,<br />

Paris, France; 5 Cincinnati Children’s Hospital Medical Center, University<br />

of Cincinnati College of Medicine, Division of Allergy and<br />

Immunology, Department of Pediatrics, Cincinnati, OH; 6 INSERM,<br />

U1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris<br />

Cité, Paris, France; 7 European Genomic Institute for Diabetes<br />

(EGID); INSERM UMR1011, University of Lille; and Institut Pasteur<br />

de Lille, Lille, France<br />

Objective: Previous <strong>studies</strong> suggested that microRNA-21<br />

may be up-regulated in the liver in non alcoholic steatohepatitis<br />

(NASH), but its role in the development of this disease<br />

remains unknown. This study aimed to determine the role of<br />

microRNA-21 in NASH. Design: We assessed features of<br />

NASH in two mouse models of NASH where microRNA-21<br />

was either pharmacologically inhibited or genetically deleted:

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!