The MRDS is currently involved in two major projects. One project is investigating the molecular mechanism(s) of 7-ketocholesterol (7KCh)-mediated toxicity and inflammation. The other project involves the development of 7KCh-mediated angiogenesis models in rat and rabbit for the purpose of testing drugs that may be useful in the treatment of age-related diseases where chronic inflammation resulting from the accumulation of oxidized lipids may be involved. 7-KCh is an oxidized derivative of cholesterol found in very high concentrations in atherosclerotic plaques (as much as 25% weight) and in the back of the retina in deposits in Bruchs membrane (BM) and the choriocapillaris (CH). 7-KCh forms by the autooxidation of cholesterol and especially cholesterol-fatty acid esters (CEs) in the presence of oxygen and a transition metal such as Cu+2 or Fe+2. This is the main reason why the CE-rich LDL deposits are so readily oxidized. A recent and yet unpublished study using monkey eyes has shown a large increase in 7KCh in the RPE and CH with aging. Monkey 16 years of age and older contain 60-70 times more 7KCh than monkeys less than 5 years of age. Drusen deposits also seem to contain large amounts of 7KCh. Preliminary results using frozen sections of drusen-containing human eyes indicate that the drusen-containing sections have 7KCh levels 100 time greater than sections without drusen. A more comprehensive collaborative study is in progress to determine the levels of 7KCh in drusen deposits. The precise mechanism of action of 7KCh is not known. Studies performed by the MRDS and by other investigators have shown that 7-KCh causes endoplasmic reticulum (ER) stress and sets off a variety of inflammatory pathways mostly mediated by NFkB. Using sterculic acid (a potent 7KCh antagonist) and a variety of other inhibitors and siRNAs we have been able to systematically eliminate all of the inflammatory pathways downstream from the ER. Presently the MRDS is focusing on determining which of the upstream pathways that lead to ER stress are activated by 7KCh and inhibited by sterculic acid. The metabolism of 7KCh outside of the liver is not known. Various investigators have reported hydroxylation by the cytochrome P450, CYP27A or sulfation by SULT2B1. These enzymes are able to use 7KCh as substrate in vitro but not in vivo. SULT2B1 expression in the retina is nil and CYP27A1 doesn't seem to come in contact with 7KCh. 7-KCh localized to lipid deposits in Bruchs membrane and in the capillary endothelial cells of the choriocapillaris and the neural retina. We have extensively searched using LCMS for hydroxylated and sulfated 7KCh metabolites in both cultured cells treated with 7KCh and in retinal and RPE extracts from monkeys and humans and have not found any. However, what we do find is an abundance of 7KCh-fatty acids esters (7KFAEs) in both in vitro and in vivo extracts. Our experiments indicate that these esters are formed by ACAT-1 (SOAT-1) in conjunction with the calcium-dependent PLA2. In vitro we have found that as much as 25% of the 7KCh that enters the cell is converted into 7KFAEs. We have also found that HDL is crucial in promoting 7KCh efflux and this occurs independently of the ABCA1 and ABCG1 transporters. Commercially purchased HDL also contains significant amounts of 7KFAEs. This suggests that HDL is a major transporter of 7KCh and its esters back to the liver. The MRDS has also been involved in developing an angiogenesis model in rats and rabbits. In this model a 0.5 mm disk- shape implant containing 7KCh (10% w/w) mixed with hydrogel and polyethylene glycol is placed in the anterior chamber of the rat eye. Within 7 days the 7KCh-containing implant becomes tumor-like doubling in size and drawing large numbers of neovessels from the limbus. Implants containing cholesterol only dissolve and dissipate without causing any inflammatory response and angiogenesis. This not only demonstrates the ability of 7KCh to cause angiogenesis but also has created a very useful angiogenesis model. The transparency of the cornea allows the monitoring and quantification of the neovessels by fluorescein angiography using a dissecting microscope equipped with a fluorescent lamp and a camera. Anti-angiogenic drugs are being tested by simply incorporating them directly into the implants or by eye-drop delivery. Aside from sterculic acid and sterculia oil we have found two naturally occurring novel compounds that inhibit angiogenesis. Oral doses of various compounds are also being tested. The rabbit with its larger eye would make a better model to test delivery mechanisms for anti-angiogenic drugs. In this model a 1 mm implant containing 20% 7KCh is surgically placed in the suprachoroidal space. The inflammation and angiogenesis are monitored by OCT, fluorescein and Indocyanine green angiography. While massive inflammation and edema have been observed, the choroidal vessels have not been able to grow through Bruchs membrane into the neural retina. We are hoping to achieve this by breaking Bruchs membrane at the site of the implant using a laser. In summary, the MRDS has made considerable progress in defining the mechanism of action of 7KCh, its metabolism and its potential role in age-related diseases. Is has also developed a very useful rat angiogenesis model for drug testing. Using this model the MRDS has demonstrated that sterculic acid and sterculia oil may be useful in the treatment of angiogenesis associated with oxidized lipids.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIAEY000307-18
Application #
8556814
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
18
Fiscal Year
2012
Total Cost
$1,241,893
Indirect Cost
Name
U.S. National Eye Institute
Department
Type
DUNS #
City
State
Country
Zip Code
Huang, Jiahn-Dar; Amaral, Juan; Lee, Jung Wha et al. (2014) 7-Ketocholesterol-induced inflammation signals mostly through the TLR4 receptor both in vitro and in vivo. PLoS One 9:e100985
Wang, Minhua; Wang, Xu; Zhao, Lian et al. (2014) Macroglia-microglia interactions via TSPO signaling regulates microglial activation in the mouse retina. J Neurosci 34:3793-806
Amaral, Juan; Lee, Jung Wha; Chou, Joshua et al. (2013) 7-Ketocholesterol induces inflammation and angiogenesis in vivo: a novel rat model. PLoS One 8:e56099
Huang, Jiahn-Dar; Amaral, Juan; Lee, Jung Wha et al. (2012) Sterculic acid antagonizes 7-ketocholesterol-mediated inflammation and inhibits choroidal neovascularization. Biochim Biophys Acta 1821:637-46
Pascual, Iranzu; Larrayoz, Ignacio M; Campos, Maria M et al. (2010) Methionine sulfoxide reductase B2 is highly expressed in the retina and protects retinal pigmented epithelium cells from oxidative damage. Exp Eye Res 90:420-8
Larrayoz, Ignacio M; Huang, Jiahn-Dar; Lee, Jung Wha et al. (2010) 7-ketocholesterol-induced inflammation: involvement of multiple kinase signaling pathways via NFýýB but independently of reactive oxygen species formation. Invest Ophthalmol Vis Sci 51:4942-55
Friedman, James S; Chang, Bo; Krauth, Daniel S et al. (2010) Loss of lysophosphatidylcholine acyltransferase 1 leads to photoreceptor degeneration in rd11 mice. Proc Natl Acad Sci U S A 107:15523-8
Pascual, Iranzu; Larrayoz, Ignacio M; Rodriguez, Ignacio R (2009) Retinoic acid regulates the human methionine sulfoxide reductase A (MSRA) gene via two distinct promoters. Genomics 93:62-71
Moreira, Ernesto F; Larrayoz, Ignacio M; Lee, Jung Wha et al. (2009) 7-Ketocholesterol is present in lipid deposits in the primate retina: potential implication in the induction of VEGF and CNV formation. Invest Ophthalmol Vis Sci 50:523-32
Rodriguez, Ignacio R; Fliesler, Steven J (2009) Photodamage generates 7-keto- and 7-hydroxycholesterol in the rat retina via a free radical-mediated mechanism. Photochem Photobiol 85:1116-25

Showing the most recent 10 out of 11 publications