MicroRNA-155 Deficiency Results in Decreased Macrophage Inflammation and Attenuated Atherogenesis in Apolipoprotein E–Deficient Mice

Arteriosclerosis, Thrombosis, and Vascular Biology - Tập 34 Số 4 - Trang 759-767 - 2014
Fu‐Sheng Du1, Fang Yu1, Yuzhen Wang1, Yvonne Hui1, Kevin Carnevale1, Mingui Fu1, Hong Lü1, Daping Fan1
1From the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Wuhan University, Wuhan, PR China (F.D.); the Department of Cell Biology and Anatomy (F.D., F.Y., Y.W., Y.H, H.L., D.F.) and the Department of Pathology, Microbiology, and Immunology (K.C.), University of South Carolina School of Medicine, Columbia; Department of Nutrition and Food Hygiene, the Fourth Military Medical University, Xi’an, Shaanxi, PR China (F.Y.); and Department of Basic Medical Science, School of...

Tóm tắt

Objective— microRNA-155 (miR155) plays a critical role in immunity and macrophage inflammation. We aim to investigate the role of miR155 in atherogenesis. Approach and Results— Quantitative real-time polymerase chain reaction showed that miR155 was expressed in mouse and human atherosclerotic lesions. miR155 expression in macrophages was correlated positively with proinflammatory cytokine expression. Lentivirus-mediated overexpression of miR155 in macrophages enhanced their inflammatory response to lipopolysaccharide through targeting suppressor of cytokine signaling-1 and impaired cholesterol efflux from acetylated low-density lipoprotein–loaded macrophages, whereas deficiency of miR155 blunted macrophage inflammatory responses and enhanced cholesterol efflux possibly via enhancing lipid loading–induced macrophage autophagy. We next examined the atherogenesis in apolipoprotein E–deficient (apoE −/− ) and miR155 −/− /apoE −/− (double knockout) mice fed a Western diet. Compared with apoE −/− mice, the double knockout mice developed less atherosclerosis lesion in aortic root, with reduced neutral lipid content and macrophages. Flow cytometric analysis showed that there were increased number of regulatory T cells and reduced numbers of Th17 cells and CD11b+/Ly6C high cells in the spleen of double knockout mice. Peritoneal macrophages from the double knockout mice had significantly reduced proinflammatory cytokine expression and secretion both in the absence and presence of lipopolysaccharide stimulation. To determine whether miR155 in leukocytes contributes to atherosclerosis, we performed a bone marrow transplantation study. Deficiency of miR155 in bone marrow–derived cells suppressed atherogenesis in apoE −/− mice, demonstrating that hematopoietic cell–derived miR155 plays a critical role. Conclusions— miR155 deficiency attenuates atherogenesis in apoE −/− mice by reducing inflammatory responses of macrophages, enhancing macrophage cholesterol efflux and resulting in an antiatherogenic leukocyte profile. Targeting miR155 may be a promising strategy to halt atherogenesis.

Từ khóa


Tài liệu tham khảo

10.1056/NEJMra043430

10.1056/NEJM199901143400207

10.1038/35025203

10.1038/nature01323

10.1038/nm.2538

10.1016/j.immuni.2013.06.009

10.1002/med.20118

10.1080/07853890701771823

10.1016/j.cell.2004.12.035

Mattes J, Collison A, Foster PS. Emerging role of microRNAs in disease pathogenesis and strategies for therapeutic modulation. Curr Opin Mol Ther. 2008;10:150–157.

10.1016/S0960-9822(02)00809-6

10.1016/j.cell.2007.04.040

10.1126/science.1139253

10.1126/science.1141229

10.1016/j.immuni.2008.03.015

10.1073/pnas.0610731104

10.1093/cvr/cvp121

10.1073/pnas.0811073106

10.1038/cmi.2009.45

10.1073/pnas.0902636106

10.1016/j.immuni.2009.06.024

10.1093/nar/gkp577

10.1371/journal.pone.0035877

10.1172/JCI61716

10.1073/pnas.0602266103

10.1016/j.cmet.2011.03.023

10.1161/atvbaha.111.240705

10.1016/S0378-1119(01)00612-6

10.1128/MCB.17.3.1490

10.1128/JVI.76.9.4275-4286.2002

10.1038/nm1008

10.1161/circulationaha.111.032268

10.1016/j.immuni.2008.11.010

10.1016/j.gde.2008.07.016

10.1016/j.cmet.2012.01.022

10.1016/j.ajpath.2012.03.013

10.4161/auto.7.10.16636

10.1161/circulationaha.112.000736

10.1371/journal.ppat.1003697

Holla S, Kurowska-Stolarska M, Bayry J, Balaji KN. Selective inhibition of IFNG-induced autophagy by—and—responsive WNT5A and SHH signaling. Autophagy. 2014;1:1–20.

10.1172/JCI63891

10.1016/j.atherosclerosis.2013.06.014

10.1172/JCI29950

10.1093/jmcb/mjs044

10.1073/pnas.1119038109

10.1158/1541-7786.MCR-12-0686

10.1161/hypertensionaha.112.197301

10.3892/ijmm.2012.1182

10.1016/j.hlc.2013.05.651