Alcohol-induced liver disease is a significant clinical problem. Kupffer cells (liver resident macrophages) play crucial roles in the inflammatory responses of alcoholic liver disease. Macrophages have distinct functional states with pro-inflammatory M1 type and anti-inflammatory M2 type. The mechanisms that govern this classical polarization remain to be elucidated. The goals of this study are to: 1) Identify a novel molecular switch that determines M1 vs. M2 polarization in the context of ethanol- induced hepatic steatosis and injury and 2) Evaluate the potential of Kupffer cells as a therapeutic target. An endoplasmic reticulum (ER) resident protein, Nogo-B, also known as reticulon 4B, has been implicated in maintaining ER structure. In the liver, Nogo-B is restricted to non-parenchymal cells including Kupffer cells, liver sinusoidal endothelial cells and hepatic stellate cells, but not in hepatocytes. Our preliminary data demonstrate that Nogo-B levels correlate with the severity of alcoholic liver disease in patients. Nogo-B levels in Kupffer cells were positively associated with M1 polarization and negatively with M2 polarization in human liver specimens. In mice, the absence of Nogo-B resulted in significantly lower levels of hepatic steatosis and injury than wildtype (WT) mice in response to an ethanol diet. Kupffer cells from Nogo-B knockout (KO) mice showed significantly decreased expression of M1 markers, including inducible nitric oxide synthase (iNOS), interleukin 1? (IL1?) and tumor necrosis factor ? (TNF?), but exhibited significantly increased M2 markers, such as CD163 and arginase-1, compared to their WT counterparts. Importantly, iNOS, IL1? and TNF? have been reported to enhance hepatic steatosis in alcoholic or non-alcoholic settings and are induced by nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB). Nogo-B KO Kupffer cells exhibited significantly increased ER stress, a factor that induces M2 polarization. Based on these observations from human specimens and animal studies, we hypothesize that Nogo-B regulates Kupffer cell polarization and facilitates hepatic steatosis/injury in response to chronic ethanol consumption and that selective deletion of Nogo-B in Kupffer cells will reduce ethanol-induced hepatic injury. To test these hypotheses, we propose the following three aims: 1) Determine the mechanism by which Nogo-B facilitates M1 polarization of Kupffer cells in response to chronic ethanol consumption, 2) Determine the mechanism by which lack of Nogo-B facilitates M2 polarization of Kupffer cells in response to chronic ethanol consumption, and 3) Determine whether deletion of Nogo-B in Kupffer cells reduces hepatic steatosis and injury in ethanol-fed mice.

Public Health Relevance

Alcohol abuse causes liver disease, whose spectrum includes alcoholic fatty liver, alcoholic hepatitis, fibrosis, cirrhosis and hepatocellular carcinoma in a progressive manner. Early intervention may prevent progression to cirrhosis and hepatocellular carcinoma, but effective treatments are limited due to our incomplete understanding of the molecular and cellular mechanisms of alcohol-induced liver injury. The goal of this proposed research is to understand the mechanism of alcohol-induced liver injury, in which Kupffer cells (liver resident macrophages) have a pivotal role.

Agency
National Institute of Health (NIH)
Institute
National Institute on Alcohol Abuse and Alcoholism (NIAAA)
Type
Research Project (R01)
Project #
5R01AA025342-03
Application #
9761401
Study Section
Hepatobiliary Pathophysiology Study Section (HBPP)
Program Officer
Gao, Peter
Project Start
2017-09-15
Project End
2022-08-31
Budget Start
2019-09-01
Budget End
2020-08-31
Support Year
3
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Yale University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
06520
Saruwatari, Junji; Dong, Chao; Utsumi, Teruo et al. (2018) Integrated analysis of microRNA and mRNA expression profiles in splenomegaly induced by non-cirrhotic portal hypertension in rats. Sci Rep 8:17983
Lai, Sanchuan; Iwakiri, Yasuko (2018) Is miR-21 a potent target for liver fibrosis? Hepatology 67:2082-2084