Dickkopf-3 Ablation Attenuates the Development of Atherosclerosis in ApoE-Deficient Mice

Wen-Lin Cheng, Yang Yang, Xiao-Jing Zhang, Junhong Guo, Jun Gong, Fu-Han Gong, Zhi-Gang She, Zan Huang, Hao Xia, Hongliang Li, Wen-Lin Cheng, Yang Yang, Xiao-Jing Zhang, Junhong Guo, Jun Gong, Fu-Han Gong, Zhi-Gang She, Zan Huang, Hao Xia, Hongliang Li

Abstract

Background: Dickkopf-3 (DKK3) is a negative regulator of the Wnt/β-catenin signaling pathway, which is involved in inflammation. However, little is known about the relationship between DKK3 expression and the progression of atherosclerosis. The aim of the present study was to define the role of DKK3 and its potential mechanism in the development of atherosclerosis.

Methods and results: Immunofluorescence analysis showed that DKK3 was strongly expressed in macrophages of atherosclerotic plaques from patients with coronary heart disease and in hyperlipidemic mice. The expression level was significantly increased in atherogenesis. DKK3-/-ApoE-/- mice exhibited a significant decrease in atherosclerotic lesions in the entire aorta, aortic sinus, and brachiocephalic arteries. Transplantation of bone marrow from DKK3-/-ApoE-/- mice into lethally irradiated ApoE-/- recipients resulted in a reduction of atherosclerotic lesions, compared with the lesions in recipients transplanted with ApoE-/- donor cells, suggesting that the effect of DKK3 deficiency was largely mediated by bone marrow-derived cells. A reduction in the necrotic core size, accompanied by increased collagen content and smooth muscle cells and decreased accumulation of macrophages and lipids, contributed to the stability of plaques in DKK3-/-ApoE-/- mice. Furthermore, multiple proinflammatory cytokines exhibited marked decreases in DKK3-/-ApoE-/- mice. Finally, we observed that DKK3 ablation increased β-catenin expression in the nuclei of macrophages both in vivo and in vitro.

Conclusions: DKK3 expression in macrophages is involved in the pathogenesis of atherosclerosis through modulation of inflammation and inactivation of the Wnt/β-catenin pathway.

Keywords: atherosclerosis; dickkopf‐3; inflammation; macrophage; β‐catenin.

© 2017 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley Blackwell.

Figures

Figure 1
Figure 1
Expression of DKK3 in atheromatous lesions of mice. A, Western blotting analysis of DKK3 protein levels in aorta of mice. The expression levels were normalized to GAPDH and quantified. n=4. *P<0.05. B, Representative images showing double immunofluorescence staining of mouse aortic sinus for DKK3 (green) and macrophages (red). Scale bar=50 μm. C, Western blot analysis of DKK3 expression in BMDMs on oxidized‐LDL treatment. *P<0.05. BMDMs indicates bone marrow‐derived macrophages; DKK3, Dickkopf‐3; LDL, low‐density lipoprotein.
Figure 2
Figure 2
Development of atherosclerosis resulted from DKK3 deficiency. A, En face Oil Red O staining of aortas from DKK3−/−ApoE−/− mice and control mice fed NC or a HFD. Lesion occupation was quantified and shown in the right and bottom panel. n=15 to 18. B, Hematoxylin and eosin staining of the lesions in aortic root sections. Scale bar=500 μm, n=8 to 10. C, Brachiocephalic artery sections were stained with hematoxylin and eosin. Scale bar=100 μm, n=11 to 12. D and E, Body weight, triglycerides, total cholesterol, and lipoprotein profiles from DKK3−/−ApoE−/− and ApoE−/− mice fed a HFD. n=20. *P<0.05 in A through C. DKK3 indicates Dickkopf‐3; HDL, high‐density lipoprotein; HFD, high‐fat diet; IDL, intermediate‐density lipoprotein; LDL, low‐density lipoprotein; NC, normal chow; TC, total cholesterol; TG, triglyceride; VLDL, very‐low‐density lipoprotein.
Figure 3
Figure 3
The necrotic area and plaque stability characteristics in DKK3‐deficient mice. A and B, Representative aortic sinus sections (A) and brachiocephalic arteries (B) from DKK3−/−ApoE−/− and ApoE−/− mice were stained with hematoxylin and eosin. The circular region indicates necrotic areas. Below panel: quantification of anuclear, afibrotic, and eosin‐negative necrotic areas. n=8 to 12. C and D, Cross sections of the aortic sinus plaques were stained with picrosirius red for collagen, α‐smooth muscle actin for smooth muscle cells, CD68 for macrophages, and Oil Red O for lipids. The assessment of plaque stability score in DKK3−/−ApoE−/− and ApoE−/− mice. n=5 to 10, Scale bar=100 μm. *P<0.05 in A, B, and D. DKK3 indicates Dickkopf‐3.
Figure 4
Figure 4
Atherogenesis of ApoE−/− mice transplanted with DKK3 deficient bone marrow cells. A and B, The genotype of ApoE (A) and DKK3 (B) in genomic DNA isolated from circulating white blood cells of recipient animals transplanted with DKK3−/−ApoE−/− or ApoE−/− bone marrow. n=4. C, Representative aorta stained with Oil Red O (n=10) (left panel) and quantification of plaque occupation in mice that received bone marrow cells from DKK3−/−ApoE−/− or ApoE−/− mice. D, Aortic root sections stained with hematoxylin and eosin (n=10) and quantification of plaque area. *P<0.05 in (C and D). DKK3 indicates Dickkopf‐3.
Figure 5
Figure 5
Inflammatory cytokine production and activity of the NF‐κB signaling pathway in DKK3 knockout mice. A, mRNA levels of proinflammatory markers in the aortas of ApoE−/− and DKK3−/−ApoE−/− mice were measured by real‐time PCR. n=6. B, Serum TNF‐α, MCP‐1, IL‐6 and IL‐1β levels were measured by ELISA in ApoE−/− and DKK3−/−ApoE−/− mice. n=10. C, Immunofluorescence staining for ICAM‐1, IL‐6, and p65 phosphorylation in the aortic sinuses of ApoE−/− and DKK3−/−ApoE−/− mice. n=5. D, Western blotting analysis of the NF‐κB signaling pathway in the aortas of ApoE−/− and IRF3−/−ApoE−/− mice, as assessed by the levels of IKKβ, IκBα, and p65 phosphorylation. Protein expression levels were normalized to GAPDH. *P<0.05 in A, B, and D. DKK3 indicates Dickkopf‐3; IL‐6, interleukin‐6; IL‐1β, interleukin‐1β; MCP‐1, monocyte chemoattractant protein‐1; TNF‐α, tumor necrosis factor‐α;
Figure 6
Figure 6
Activation of the Wnt/β‐catenin pathway in DKK3 deficiency. A, The total protein levels of β‐catenin in the nucleus and the phosphorylated and total protein levels of β‐catenin in the cytoplasm of DKK3−/−ApoE−/− and ApoE−/− mice. *P<0.05. B, Immunofluorescence costaining of atherosclerotic plaques with β‐catenin (red) and CD68 (green). n=3. Scale bar=20 μm. C, Western blot analysis of β‐catenin in the nucleus and the phosphorylated and total protein levels of β‐catenin in the cytoplasm of peritoneal macrophages isolated from ApoE−/− and DKK3−/− ApoE−/− upon oxidized‐LDL stimulation. *P<0.05. D, mRNA levels of proinflammatory markers in peritoneal macrophages isolated from ApoE−/− and DKK3−/− ApoE−/− treatment with oxidized LDL. *P<0.05. DKK3 indicates Dickkopf‐3; LDL, low‐density lipoprotein.
Figure 7
Figure 7
Expression of DKK3 in human atheromatous lesions. A, Western blotting analysis of DKK3 protein levels in the right coronary artery in humans. The expression levels were normalized to GAPDH and quantified. n=4. *P<0.05. B, Representative images showing double‐immunofluorescence staining of human coronary arteries for DKK3 (green) and macrophages (red). Scale bar=50 μm. DKK3 indicates Dickkopf‐3.

References

    1. Ross R. Atherosclerosis—an inflammatory disease. N Engl J Med. 1999;340:115–126.
    1. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.
    1. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Magid DJ, McGuire DK, Mohler ER III, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, Turner MB; American Heart Association Statistics Committee; Stroke Statistics Subcommittee . Executive summary: heart disease and stroke statistics—2016 update: a report from the American Heart Association. Circulation. 2016;133:447–454.
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007;7:678–689.
    1. Tabas I. Macrophage death and defective inflammation resolution in atherosclerosis. Nat Rev Immunol. 2010;10:36–46.
    1. Virmani R, Burke AP, Kolodgie FD, Farb A. Vulnerable plaque: the pathology of unstable coronary lesions. J Interv Cardiol. 2002;15:439–446.
    1. Nelson WJ, Nusse R. Convergence of Wnt, β‐catenin, and cadherin pathways. Science. 2004;303:1483–1487.
    1. Schaale K, Neumann J, Schneider D, Ehlers S, Reiling N. Wnt signaling in macrophages: augmenting and inhibiting mycobacteria‐induced inflammatory responses. Eur J Cell Biol. 2011;90:553–559.
    1. Ding Y, Shen S, Lino AC, Curotto de Lafaille MA, Lafaille JJ. Beta‐catenin stabilization extends regulatory T cell survival and induces anergy in nonregulatory T cells. Nat Med. 2008;14:162–169.
    1. Niehrs C. Function and biological roles of the Dickkopf family of Wnt modulators. Oncogene. 2006;25:7469–7481.
    1. Nakamura RE, Hunter DD, Yi H, Brunken WJ, Hackam AS. Identification of two novel activities of the Wnt signaling regulator Dickkopf 3 and characterization of its expression in the mouse retina. BMC Cell Biol. 2007;8:52.
    1. Yang ZR, Dong WG, Lei XF, Liu M, Liu QS. Overexpression of Dickkopf‐3 induces apoptosis through mitochondrial pathway in human colon cancer. World J Gastroenterol. 2012;18:1590–1601.
    1. Abarzua F, Sakaguchi M, Takaishi M, Nasu Y, Kurose K, Ebara S, Miyazaki M, Namba M, Kumon H, Huh NH. Adenovirus‐mediated overexpression of REIC/Dkk‐3 selectively induces apoptosis in human prostate cancer cells through activation of c‐Jun‐NH2‐kinase. Cancer Res. 2005;65:9617–9622.
    1. Yu J, Tao Q, Cheng YY, Lee KY, Ng SS, Cheung KF, Tian L, Rha SY, Neumann U, Rocken C, Ebert MP, Chan FK, Sung JJ. Promoter methylation of the Wnt/β‐catenin signaling antagonist Dkk‐3 is associated with poor survival in gastric cancer. Cancer. 2009;115:49–60.
    1. Xiang T, Li L, Yin X, Zhong L, Peng W, Qiu Z, Ren G, Tao Q. Epigenetic silencing of the WNT antagonist Dickkopf 3 disrupts normal Wnt/β‐catenin signalling and apoptosis regulation in breast cancer cells. J Cell Mol Med. 2013;17:1236–1246.
    1. Roman‐Gomez J, Jimenez‐Velasco A, Agirre X, Castillejo JA, Navarro G, Barrios M, Andreu EJ, Prosper F, Heiniger A, Torres A. Transcriptional silencing of the Dickkopfs‐3 (Dkk‐3) gene by CpG hypermethylation in acute lymphoblastic leukaemia. Br J Cancer. 2004;91:707–713.
    1. Kurose K, Sakaguchi M, Nasu Y, Ebara S, Kaku H, Kariyama R, Arao Y, Miyazaki M, Tsushima T, Namba M, Kumon H, Huh NH. Decreased expression of REIC/Dkk‐3 in human renal clear cell carcinoma. J Urol. 2004;171:1314–1318.
    1. Tsuji T, Nozaki I, Miyazaki M, Sakaguchi M, Pu H, Hamazaki Y, Iijima O, Namba M. Antiproliferative activity of REIC/Dkk‐3 and its significant down‐regulation in non‐small‐cell lung carcinomas. Biochem Biophys Res Commun. 2001;289:257–263.
    1. Hsieh SY, Hsieh PS, Chiu CT, Chen WY. Dickkopf‐3/REIC functions as a suppressor gene of tumor growth. Oncogene. 2004;23:9183–9189.
    1. Lu KH, Tounsi A, Shridhar N, Kublbeck G, Klevenz A, Prokosch S, Bald T, Tuting T, Arnold B. Dickkopf‐3 contributes to the regulation of anti‐tumor immune responses by mesenchymal stem cells. Front Immunol. 2015;6:645.
    1. Monaghan AP, Kioschis P, Wu W, Zuniga A, Bock D, Poustka A, Delius H, Niehrs C. Dickkopf genes are co‐ordinately expressed in mesodermal lineages. Mech Dev. 1999;87:45–56.
    1. Zhang Y, Liu Y, Zhu XH, Zhang XD, Jiang DS, Bian ZY, Zhang XF, Chen K, Wei X, Gao L, Zhu LH, Yang Q, Fan GC, Lau WB, Ma X, Li H. Dickkopf‐3 attenuates pressure overload‐induced cardiac remodelling. Cardiovasc Res. 2014;102:35–45.
    1. Bao MW, Cai Z, Zhang XJ, Li L, Liu X, Wan N, Hu G, Wan F, Zhang R, Zhu X, Xia H, Li H. Dickkopf‐3 protects against cardiac dysfunction and ventricular remodelling following myocardial infarction. Basic Res Cardiol. 2015;110:25.
    1. Xie L, Wang PX, Zhang P, Zhang XJ, Zhao GN, Wang A, Guo J, Zhu X, Zhang Q, Li H. DKK3 expression in hepatocytes defines susceptibility to liver steatosis and obesity. J Hepatol. 2016;65:113–124.
    1. Wang PX, Zhang XJ, Luo P, Jiang X, Zhang P, Guo J, Zhao GN, Zhu X, Zhang Y, Yang S, Li H. Hepatocyte TRAF3 promotes liver steatosis and systemic insulin resistance through targeting TAK1‐dependent signalling. Nat Commun. 2016;7:10592.
    1. Li HL, Wang AB, Zhang R, Wei YS, Chen HZ, She ZG, Huang Y, Liu DP, Liang CC. A20 inhibits oxidized low‐density lipoprotein‐induced apoptosis through negative Fas/Fas ligand‐dependent activation of caspase‐8 and mitochondrial pathways in murine RAW264.7 macrophages. J Cell Physiol. 2006;208:307–318.
    1. Ni W, Egashira K, Kitamoto S, Kataoka C, Koyanagi M, Inoue S, Imaizumi K, Akiyama C, Nishida KI, Takeshita A. New anti‐monocyte chemoattractant protein‐1 gene therapy attenuates atherosclerosis in apolipoprotein E‐knockout mice. Circulation. 2001;103:2096–2101.
    1. Ji YX, Zhang P, Zhang XJ, Zhao YC, Deng KQ, Jiang X, Wang PX, Huang Z, Li H. The ubiquitin E3 ligase TRAF6 exacerbates pathological cardiac hypertrophy via TAK1‐dependent signalling. Nat Commun. 2016;7:11267.
    1. Deng KQ, Wang A, Ji YX, Zhang XJ, Fang J, Zhang Y, Zhang P, Jiang X, Gao L, Zhu XY, Zhao Y, Gao L, Yang Q, Zhu XH, Wei X, Pu J, Li H. Suppressor of IKKε is an essential negative regulator of pathological cardiac hypertrophy. Nat Commun. 2016;7:11432.
    1. Jiang DS, Wei X, Zhang XF, Liu Y, Zhang Y, Chen K, Gao L, Zhou H, Zhu XH, Liu PP, Bond Lau W, Ma X, Zou Y, Zhang XD, Fan GC, Li H. IRF8 suppresses pathological cardiac remodelling by inhibiting calcineurin signalling. Nat Commun. 2014;5:3303.
    1. Rosenfeld ME, Carson KG, Johnson JL, Williams H, Jackson CL, Schwartz SM. Animal models of spontaneous plaque rupture: the holy grail of experimental atherosclerosis research. Curr Atheroscler Rep. 2002;4:238–242.
    1. Karamariti E, Margariti A, Winkler B, Wang X, Hong X, Baban D, Ragoussis J, Huang Y, Han JD, Wong MM, Sag CM, Shah AM, Hu Y, Xu Q. Smooth muscle cells differentiated from reprogrammed embryonic lung fibroblasts through DKK3 signaling are potent for tissue engineering of vascular grafts. Circ Res. 2013;112:1433–1443.
    1. Wang X, Karamariti E, Simpson R, Wang W, Xu Q. Dickkopf homolog 3 induces stem cell differentiation into smooth muscle lineage via ATF6 signalling. J Biol Chem. 2015;290:19844–19852.
    1. Angers S, Moon RT. Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol. 2009;10:468–477.
    1. Gordon MD, Nusse R. Wnt signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem. 2006;281:22429–22433.
    1. van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development. 2009;136:3205–3214.
    1. Mikels AJ, Nusse R. Purified Wnt5a protein activates or inhibits β‐catenin–TCF signaling depending on receptor context. PLoS Biol. 2006;4:e115.
    1. Mill C, George SJ. Wnt signalling in smooth muscle cells and its role in cardiovascular disorders. Cardiovasc Res. 2012;95:233–240.
    1. Sarzani R, Salvi F, Bordicchia M, Guerra F, Battistoni I, Pagliariccio G, Carbonari L, Dessi‐Fulgheri P, Rappelli A. Carotid artery atherosclerosis in hypertensive patients with a functional LDL receptor‐related protein 6 gene variant. Nutr Metab Cardiovasc Dis. 2011;21:150–156.
    1. Mani A, Radhakrishnan J, Wang H, Mani A, Mani MA, Nelson‐Williams C, Carew KS, Mane S, Najmabadi H, Wu D, Lifton RP. LRP6 mutation in a family with early coronary disease and metabolic risk factors. Science. 2007;315:1278–1282.
    1. Magoori K, Kang MJ, Ito MR, Kakuuchi H, Ioka RX, Kamataki A, Kim DH, Asaba H, Iwasaki S, Takei YA, Sasaki M, Usui S, Okazaki M, Takahashi S, Ono M, Nose M, Sakai J, Fujino T, Yamamoto TT. Severe hypercholesterolemia, impaired fat tolerance, and advanced atherosclerosis in mice lacking both low density lipoprotein receptor‐related protein 5 and apolipoprotein E. J Biol Chem. 2003;278:11331–11336.
    1. Go GW. Low‐density lipoprotein receptor‐related protein 6 (LRP6) is a novel nutritional therapeutic target for hyperlipidemia, non‐alcoholic fatty liver disease, and atherosclerosis. Nutrients. 2015;7:4453–4464.
    1. Malhotra S, Kincade PW. Canonical Wnt pathway signaling suppresses VCAM‐1 expression by marrow stromal and hematopoietic cells. Exp Hematol. 2009;37:19–30.
    1. Tickenbrock L, Schwable J, Strey A, Sargin B, Hehn S, Baas M, Choudhary C, Gerke V, Berdel WE, Muller‐Tidow C, Serve H. Wnt signaling regulates transendothelial migration of monocytes. J Leukoc Biol. 2006;79:1306–1313.

Source: PubMed

3
구독하다