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 847A<br />

1292<br />

Implants of matrix-embedded endothelial cells rescue<br />

ischemic tissue and liver engraftment in hepatectomized<br />

mice<br />

Pedro Melgar-Lesmes 1 , Mercedes Balcells 2,1 , Elazer R. Edelman 1,3 ;<br />

1 Edelman Lab, Institute for Medical Engineering and Science,<br />

Massachusetts Institute of Technology, Cambridge, MA, USA,<br />

Cambridge,, MA; 2 Bioengineering department, Institut Químic de<br />

Sarrià, Ramon Llull Univ, Barcelona, Spain; 3 Cardiovascular Division,<br />

Brigham and Women’s Hospital, Harvard Medical School,<br />

Boston, MA<br />

Background and aims: Ischemia during liver transplantation<br />

promotes a cascade of cellular injury responses that lead to<br />

inflammation, cell death, and organ dysfunction. Matrix-embedded<br />

endothelial cells (MEECs) have immunomodulatory<br />

effects in vitro and in vivo. Therefore we investigated the benefits<br />

of implants of MEECs as modulators of inflammation and<br />

regeneration after liver engraftment in mice. Methods: Partial<br />

hepatectomy (70%) was performed by excising the left lobe<br />

and half the median lobe of the mouse liver. Four groups of<br />

10 animals were distributed as follows: 1) acellular matrices<br />

in the interface between the remaining median lobe and an<br />

autograft from the left lobe; 2) MEECs between the remaining<br />

median lobe and an autograft from the left lobe; 3) acellular<br />

matrices between the remaining median lobe and an allograft<br />

from the left lobe of another mouse; 4) MEECs between the<br />

remaining median lobe and an allograft from the left lobe of<br />

another mouse. Vascular architecture was analysed using angiography<br />

with FITC dextran 2000 kDa by fluorescent multiphoton<br />

microscopy 7 days post-op. Hepatic DNA fragmentation<br />

and apoptosis were quantified in the median lobe and grafts<br />

by TUNEL assay and Western Blot of activated caspase 3,<br />

respectively. Serum markers of liver damage Alanine Aminotransferase<br />

(ALT) and Aspartate Aminotransferase (AST) were<br />

quantified in all animal groups. The phenotype of lymphocyte<br />

subsets in the liver after implantation of allografts was quantified<br />

by gene expression of Th1 (interferon gamma: INFγ; interleukin<br />

2: IL-2) and Th2 (interleukin 4: IL-4; interleukin 10: IL-10)<br />

genes by Real-Time PCR. Results: Implants with MEECs created<br />

a functional vascular splice between the ischemic median lobe<br />

and autografts or allografts improving the rate of liver donor<br />

regeneration by 15% compared to acellular controls. MEECs<br />

prevented apoptosis by 85% in donor liver lobes, by 85%<br />

in autologous grafts and by 79% in allogeneic engraftments.<br />

As a result, serum levels of ALT and AST were significantly<br />

reduced after autologous or allogeneic engraftments. The beneficial<br />

immunomodulatory effects of MEECs promoted allograft<br />

immunotolerance expressed as a significant reduction of Th1<br />

(INFγ and IL-2) and increase of Th2 (IL-4 and IL-10) cytokine<br />

expression. Conclusions: Implants of MEECs are endothelial<br />

cell-based scaffolds with potential to be used to improve current<br />

surgical procedures in liver transplantation and to reduce<br />

ischemic and inflammatory disorders. Their application may<br />

contribute obtaining functional liver grafts of adequate size for<br />

the recipient without subjecting the donor to undue risk.<br />

Disclosures:<br />

The following authors have nothing to disclose: Pedro Melgar-Lesmes, Mercedes<br />

Balcells, Elazer R. Edelman<br />

1293<br />

Identification of liver monocytic myeloid-derived suppressor<br />

cells and elucidation of their roles in non-alcoholic<br />

fatty liver disease<br />

Liying Yao, Masanori Abe, Teruki Miyake, Yoshiko Nakamura,<br />

Yusuke Imai, Yohei Koizumi, Takao Watanabe, Osamu Yoshida,<br />

Masashi Hirooka, Yoshio Tokumoto, Bunzo Matsuura, Yoichi<br />

Hiasa; Department of Gastroenterology and Metabology, Ehime<br />

Universiy Graduate School of Medicine, Ehime, Japan<br />

Background/Aim: Myeloid-derived suppressor cells (MDSCs)<br />

comprise a heterogeneous population of myeloid cells and are<br />

recognized as suppressors of T cell functions. In mice, these<br />

cells identified by the co-expression of CD11b and Gr-1 surface<br />

markers. Previous <strong>studies</strong> revealed that liver MDSC exhibit<br />

phenotypic and functional diversity; however, information<br />

regarding their characteristics and mechanisms of suppression,<br />

reported in these <strong>studies</strong>, was conflicting in nature. Immune<br />

responses may contribute to the pathogenesis of non-alcoholic<br />

fatty liver disease (NAFLD); however, little is known regarding<br />

the role of myeloid regulatory cells. In this study, we characterized<br />

the phenotype of liver MDSCs in a murine model of<br />

NAFLD and explored the mechanism underlying their immunosuppression.<br />

Methods: C57BL/6 mice were fed a normal<br />

diet (ND) or a high-fat diet (HFD) for 12 months. Different subtypes<br />

of CD11b + Gr-1 + cells were sorted from liver non-parenchymal<br />

cells by FACS. CD11b + Gr-1 + cells were co-cultured<br />

with T cells in the presence of anti-CD3 beads or allogeneic<br />

dendritic cells, and suppressive functions were investigated<br />

using a carboxyfluorescein succinimidyl ester assay. Nitric<br />

oxide (NO) concentrations in culture supernatants were measured<br />

using Griess reagent. In some experiments, L-NIL, an<br />

inducible NO synthase (iNOS) inhibitor, was added at the<br />

start of the culture. Results: In the livers of HFD-fed mice, the<br />

frequency of CD11b + Gr1 dim cells (monocytic myeloid cells),<br />

but not CD11b + Gr1 hi cells, was higher than that in the livers<br />

of ND-fed mice over time. CD11b + Gr1 dim cells were divided<br />

into SSC high and SSC low populations. The frequency of SSC low-<br />

CD11b + Gr1 dim cells increased in the livers of HFD-fed mice<br />

to a greater extent than that observed in the livers of ND-fed<br />

mice. In addition, SSC low CD11b + Gr1 dim cells suppressed T cell<br />

proliferation in a dose-dependent manner; however, this suppression<br />

was not observed in SSC low CD11b + Gr1 high cells. We<br />

also found that SSC low CD11b + Gr1 dim cells expressed iNOS<br />

after co-culture with T cells, and the supernatants obtained from<br />

the co-culture of SSC low CD11b + Gr1 dim cells with T cells had<br />

higher concentrations of NO. By adding iNOS inhibitor L-NIL<br />

to the SSC low CD11b + Gr1 dim cells and T cells co-culture, T cell<br />

proliferation was restored. Conclusion: SSC low CD11b + Gr1 dim<br />

cells represent authentic monocytic MDSCs in the liver, and<br />

their numbers were increased in the livers of NAFLD mice. The<br />

suppressive function of monocytic MDSCs was dependent on<br />

NO production by iNOS. These cells might have a role in the<br />

pathogenesis of NAFLD.<br />

Disclosures:<br />

The following authors have nothing to disclose: Liying Yao, Masanori Abe, Teruki<br />

Miyake, Yoshiko Nakamura, Yusuke Imai, Yohei Koizumi, Takao Watanabe,<br />

Osamu Yoshida, Masashi Hirooka, Yoshio Tokumoto, Bunzo Matsuura, Yoichi<br />

Hiasa

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

Saved successfully!

Ooh no, something went wrong!