@phdthesis{Chaudhari2013, author = {Chaudhari, Sweena M.}, title = {Role of Hypoxia-Inducible Factor (HIF) 1α in Dendritic Cells in Immune Regulation of Atherosclerosis}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-91853}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2013}, abstract = {Atherosclerosis is the underlying cause of cardiovascular diseases and a major threat to human health worldwide. It involves not only accumulation of lipids in the vessel wall but a chronic inflammatory response mediated by highly specific cellular and molecular responses. Macrophages and dendritic cells (DCs) play an essential role in taking up modified lipids and presenting them to T and B lymphocytes, which promote the immune response. Enhanced activation, migration and accumulation of inflammatory cells at the local site leads to formation of atherosclerotic plaques. Atherosclerotic plaques become hypoxic due to reduced oxygen diffusion and high metabolic demand of accumulated cells. The various immune cells experience hypoxic conditions locally and inflammatory stimuli systemically, thus up-regulating Hypoxia-inducible factor 1α. Though the role of HIF1α in macrophages and lymphocytes has been elucidated, its role in DCs still remains controversial, especially with respect to atherosclerosis. In this project work, the role of HIF1α in DCs was investigated by using a cell specific knockout mouse model where HIF1α was deleted in CD11c+ cells. Aortic root sections from atherosclerotic mice showed presence of hypoxia and up-regulation of HIF1α which co-localized with CD11c+ cells. Atherosclerotic splenic DCs also displayed enhanced expression of HIF1α, proving non-hypoxic stimulation of HIF1α due to systemic inflammation. Conditional knockout (CKO) mice lacking HIF1α in CD11c+ cells, under baseline conditions did not show changes in immune responses suggesting effects of HIF1α only under inflammatory conditions. When these mice were crossed to the Ldlr-/- line and placed on 8 weeks of high fat diet, they developed enhanced plaques with higher T-cell infiltration as compared to the wild-type (WT) controls. The plaques were of a complex phenotype, defined by increased percent of smooth muscle cells (SMCs) and necrotic core area and reduced percent of macrophages and DCs. The mice also displayed enhanced T-cell activation and a Th1 bias in the periphery. The CKO DCs themselves exhibited increased expression of IL 12 and a higher capacity to proliferate and polarize naive T cells to the Th1 phenotype in vitro. The DCs also showed decreased expression of STAT3, in line with the inhibitory effects of STAT3 on DC activation seen in previous studies. When STAT3 was overexpressed in DCs in vitro, IL 12 was down-regulated, but its expression increased significantly on STAT3 inhibition using a mutant vector. In addition, when STAT3 was overexpressed in DCs in vivo using a Cre regulated lentiviral system, the mice showed decreased plaque formation compared to controls. Interestingly, the effects of STAT3 modulation were similar in WT and CKO mice, intending that STAT3 lies downstream of HIF1α. Finally, using a chromatin immunoprecipitation assay (ChIP), it was confirmed that HIF1α binds to hypoxia responsive elements (HREs) in the Stat3 gene promoter thus regulating its expression. When DCs lack HIF1α, STAT3 expression is not stimulated and hence IL 12 production by DCs is uninhibited. This excessive IL 12 can activate naive T cells and polarize them to the Th1 phenotype, thereby enhancing atherosclerotic plaque progression. This project thus concludes that HIF1α restrains DC activation via STAT3 generation and prevents excessive production of IL 12 that helps to keep inflammation and atherosclerosis under check.}, subject = {Dendritische Zelle}, language = {en} }