#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Plasma Cholesterol–Induced Lesion Networks Activated before Regression of Early, Mature, and Advanced Atherosclerosis


Plasma cholesterol lowering (PCL) slows and sometimes prevents progression of atherosclerosis and may even lead to regression. Little is known about how molecular processes in the atherosclerotic arterial wall respond to PCL and modify responses to atherosclerosis regression. We studied atherosclerosis regression and global gene expression responses to PCL (≥80%) and to atherosclerosis regression itself in early, mature, and advanced lesions. In atherosclerotic aortic wall from Ldlr−/−Apob100/100Mttpflox/floxMx1-Cre mice, atherosclerosis regressed after PCL regardless of lesion stage. However, near-complete regression was observed only in mice with early lesions; mice with mature and advanced lesions were left with regression-resistant, relatively unstable plaque remnants. Atherosclerosis genes responding to PCL before regression, unlike those responding to the regression itself, were enriched in inherited risk for coronary artery disease and myocardial infarction, indicating causality. Inference of transcription factor (TF) regulatory networks of these PCL-responsive gene sets revealed largely different networks in early, mature, and advanced lesions. In early lesions, PPARG was identified as a specific master regulator of the PCL-responsive atherosclerosis TF-regulatory network, whereas in mature and advanced lesions, the specific master regulators were MLL5 and SRSF10/XRN2, respectively. In a THP-1 foam cell model of atherosclerosis regression, siRNA targeting of these master regulators activated the time-point-specific TF-regulatory networks and altered the accumulation of cholesterol esters. We conclude that PCL leads to complete atherosclerosis regression only in mice with early lesions. Identified master regulators and related PCL-responsive TF-regulatory networks will be interesting targets to enhance PCL-mediated regression of mature and advanced atherosclerotic lesions.


Vyšlo v časopise: Plasma Cholesterol–Induced Lesion Networks Activated before Regression of Early, Mature, and Advanced Atherosclerosis. PLoS Genet 10(2): e32767. doi:10.1371/journal.pgen.1004201
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004201

Souhrn

Plasma cholesterol lowering (PCL) slows and sometimes prevents progression of atherosclerosis and may even lead to regression. Little is known about how molecular processes in the atherosclerotic arterial wall respond to PCL and modify responses to atherosclerosis regression. We studied atherosclerosis regression and global gene expression responses to PCL (≥80%) and to atherosclerosis regression itself in early, mature, and advanced lesions. In atherosclerotic aortic wall from Ldlr−/−Apob100/100Mttpflox/floxMx1-Cre mice, atherosclerosis regressed after PCL regardless of lesion stage. However, near-complete regression was observed only in mice with early lesions; mice with mature and advanced lesions were left with regression-resistant, relatively unstable plaque remnants. Atherosclerosis genes responding to PCL before regression, unlike those responding to the regression itself, were enriched in inherited risk for coronary artery disease and myocardial infarction, indicating causality. Inference of transcription factor (TF) regulatory networks of these PCL-responsive gene sets revealed largely different networks in early, mature, and advanced lesions. In early lesions, PPARG was identified as a specific master regulator of the PCL-responsive atherosclerosis TF-regulatory network, whereas in mature and advanced lesions, the specific master regulators were MLL5 and SRSF10/XRN2, respectively. In a THP-1 foam cell model of atherosclerosis regression, siRNA targeting of these master regulators activated the time-point-specific TF-regulatory networks and altered the accumulation of cholesterol esters. We conclude that PCL leads to complete atherosclerosis regression only in mice with early lesions. Identified master regulators and related PCL-responsive TF-regulatory networks will be interesting targets to enhance PCL-mediated regression of mature and advanced atherosclerotic lesions.


Zdroje

1. Global Atlas on Cardiovascular Disease Prevention and Control (Mendis S, Puska P, Norrving B, eds). Geneva: World Health Organization, 2011.

2. BrownMS, GoldsteinJL (2006) Biomedicine. Lowering LDL–not only how low, but how long? Science 311: 1721–1723.

3. BrownBG, ZhaoXQ, SaccoDE, AlbersJJ (1993) Lipid lowering and plaque regression. New insights into prevention of plaque disruption and clinical events in coronary disease. Circulation 87: 1781–1791.

4. LaRosaJC, GrundySM, WatersDD, ShearC, BarterP, et al. (2005) Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 352: 1425–1435.

5. OngHT (2005) The statin studies: from targeting hypercholesterolaemia to targeting the high-risk patient. QJM 98: 599–614.

6. NissenSE, NichollsSJ, SipahiI, LibbyP, RaichlenJS, et al. (2006) Effect of very high-intensity statin therapy on regression of coronary atherosclerosis: the ASTEROID trial. JAMA 295: 1556–1565.

7. CrouseJR3rd, RaichlenJS, RileyWA, EvansGW, PalmerMK, et al. (2007) Effect of rosuvastatin on progression of carotid intima-media thickness in low-risk individuals with subclinical atherosclerosis: the METEOR Trial. JAMA 297: 1344–1353.

8. GrinesCL (2006) Transcatheter cardiovascular therapeutics annual meeting. J Interv Cardiol 19: 183–210.

9. ChanAW, BhattDL, ChewDP, QuinnMJ, MoliternoDJ, et al. (2002) Early and sustained survival benefit associated with statin therapy at the time of percutaneous coronary intervention. Circulation 105: 691–696.

10. TaylorF, WardK, MooreTH, BurkeM, Davey SmithG, et al. (2011) Statins for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev CD004816.

11. TaylorF, HuffmanMD, MacedoAF, MooreTH, BurkeM, et al. (2013) Statins for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 1: CD004816.

12. SkogsbergJ, LundstromJ, KovacsA, NilssonR, NooriP, et al. (2008) Transcriptional profiling uncovers a network of cholesterol-responsive atherosclerosis target genes. PLoS Genet 4: e1000036.

13. FeigJE, Pineda-TorraI, SansonM, BradleyMN, VengrenyukY, et al. (2010) LXR promotes the maximal egress of monocyte-derived cells from mouse aortic plaques during atherosclerosis regression. J Clin Invest 120: 4415–4424.

14. ReisED, LiJ, FayadZA, RongJX, HansotyD, et al. (2001) Dramatic remodeling of advanced atherosclerotic plaques of the apolipoprotein E-deficient mouse in a novel transplantation model. J Vasc Surg 34: 541–547.

15. TroganE, FayadZA, ItskovichVV, AguinaldoJG, ManiV, et al. (2004) Serial studies of mouse atherosclerosis by in vivo magnetic resonance imaging detect lesion regression after correction of dyslipidemia. Arterioscler Thromb Vasc Biol 24: 1714–1719.

16. TroganE, FeigJE, DoganS, RothblatGH, AngeliV, et al. (2006) Gene expression changes in foam cells and the role of chemokine receptor CCR7 during atherosclerosis regression in ApoE-deficient mice. Proc Natl Acad Sci U S A 103: 3781–3786.

17. FeigJE, VengrenyukY, ReiserV, WuC, StatnikovA, et al. (2012) Regression of atherosclerosis is characterized by broad changes in the plaque macrophage transcriptome. PLoS One 7: e39790.

18. LlodraJ, AngeliV, LiuJ, TroganE, FisherEA, et al. (2004) Emigration of monocyte-derived cells from atherosclerotic lesions characterizes regressive, but not progressive, plaques. Proc Natl Acad Sci U S A 101: 11779–11784.

19. PotteauxS, GautierEL, HutchisonSB, van RooijenN, RaderDJ, et al. (2011) Suppressed monocyte recruitment drives macrophage removal from atherosclerotic plaques of Apoe−/− mice during disease regression. J Clin Invest 121: 2025–2036.

20. HewingB, ParathathS, MaiCK, FielMI, GuoL, et al. (2013) Rapid regression of atherosclerosis with MTP inhibitor treatment. Atherosclerosis 227: 125–129.

21. FeigJE, ParathathS, RongJX, MickSL, VengrenyukY, et al. (2011) Reversal of hyperlipidemia with a genetic switch favorably affects the content and inflammatory state of macrophages in atherosclerotic plaques. Circulation 123: 989–998.

22. LieuHD, WithycombeSK, WalkerQ, RongJX, WalzemRL, et al. (2003) Eliminating atherogenesis in mice by switching off hepatic lipoprotein secretion. Circulation 107: 1315–1321.

23. ParathathS, GrauerL, HuangLS, SansonM, DistelE, et al. (2011) Diabetes adversely affects macrophages during atherosclerotic plaque regression in mice. Diabetes 60: 1759–1769.

24. SchadtEE (2009) Molecular networks as sensors and drivers of common human diseases. Nature 461: 218–223.

25. SchadtEE, BjorkegrenJL (2012) NEW: network-enabled wisdom in biology, medicine, and health care. Sci Transl Med 4: 115rv111.

26. RaabeM, VeniantMM, SullivanMA, ZlotCH, BjorkegrenJ, et al. (1999) Analysis of the role of microsomal triglyceride transfer protein in the liver of tissue-specific knockout mice. J Clin Invest 103: 1287–1298.

27. NiW, EgashiraK, KitamotoS, KataokaC, KoyanagiM, et al. (2001) New anti-monocyte chemoattractant protein-1 gene therapy attenuates atherosclerosis in apolipoprotein E-knockout mice. Circulation 103: 2096–2101.

28. KathiresanS, VoightBF, PurcellS, MusunuruK, ArdissinoD, et al. (2009) Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat Genet 41: 334–341.

29. HaggS, SkogsbergJ, LundstromJ, NooriP, NilssonR, et al. (2009) Multi-organ expression profiling uncovers a gene module in coronary artery disease involving transendothelial migration of leukocytes and LIM domain binding 2: the Stockholm Atherosclerosis Gene Expression (STAGE) study. PLoS Genet 5: e1000754.

30. RamenskyV, BorkP, SunyaevS (2002) Human non-synonymous SNPs: server and survey. Nucleic Acids Res 30: 3894–3900.

31. HoldtLM, TeupserD (2013) From genotype to phenotype in human atherosclerosis–recent findings. Curr Opin Lipidol 24: 410–418.

32. RobertsR, StewartAF (2012) Genes and coronary artery disease: where are we? J Am Coll Cardiol 60: 1715–1721.

33. FeigJE, ShangY, RotllanN, VengrenyukY, WuC, et al. (2011) Statins promote the regression of atherosclerosis via activation of the CCR7-dependent emigration pathway in macrophages. PLoS One 6: e28534.

34. RobbinsCS, HilgendorfI, WeberGF, TheurlI, IwamotoY, et al. (2013) Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med 19: 1166–1172.

35. GrundySM (1986) Cholesterol and coronary heart disease. A new era. JAMA 256: 2849–2858.

36. BentzonJF, FalkE (2010) Atherosclerotic lesions in mouse and man: is it the same disease? Curr Opin Lipidol 21: 434–440.

37. GoldbourtU, YaariS, MedalieJH (1997) Isolated low HDL cholesterol as a risk factor for coronary heart disease mortality. A 21-year follow-up of 8000 men. Arterioscler Thromb Vasc Biol 17: 107–113.

38. GordonDJ, RifkindBM (1989) High-density lipoprotein–the clinical implications of recent studies. N Engl J Med 321: 1311–1316.

39. ShahPK, YanoJ, ReyesO, ChyuKY, KaulS, et al. (2001) High-dose recombinant apolipoprotein A-I(milano) mobilizes tissue cholesterol and rapidly reduces plaque lipid and macrophage content in apolipoprotein e-deficient mice. Potential implications for acute plaque stabilization. Circulation 103: 3047–3050.

40. LiR, ChaoH, KoKW, CormierS, DiekerC, et al. (2011) Gene therapy targeting LDL cholesterol but not HDL cholesterol induces regression of advanced atherosclerosis in a mouse model of familial hypercholesterolemia. J Genet Syndr Gene Ther 2: 106.

41. TangiralaRK, TsukamotoK, ChunSH, UsherD, PureE, et al. (1999) Regression of atherosclerosis induced by liver-directed gene transfer of apolipoprotein A-I in mice. Circulation 100: 1816–1822.

42. Van CraeyveldE, GordtsSC, NefyodovaE, JacobsF, De GeestB (2011) Regression and stabilization of advanced murine atherosclerotic lesions: a comparison of LDL lowering and HDL raising gene transfer strategies. J Mol Med (Berl) 89: 555–567.

43. FeigJE, RongJX, ShamirR, SansonM, VengrenyukY, et al. (2011) HDL promotes rapid atherosclerosis regression in mice and alters inflammatory properties of plaque monocyte-derived cells. Proc Natl Acad Sci U S A 108: 7166–7171.

44. GomezD, OwensGK (2012) Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res 95: 156–164.

45. TegnerJ, SkogsbergJ, BjorkegrenJ (2007) Thematic review series: systems biology approaches to metabolic and cardiovascular disorders. Multi-organ whole-genome measurements and reverse engineering to uncover gene networks underlying complex traits. J Lipid Res 48: 267–277.

46. VeniantMM, SullivanMA, KimSK, AmbroziakP, ChuA, et al. (2000) Defining the atherogenicity of large and small lipoproteins containing apolipoprotein B100. J Clin Invest 106: 1501–1510.

47. StotzE, SchenkEA, ChurukianC, WillisC (1986) Oil red O: comparison of staining quality and chemical components as determined by thin layer chromatography. Stain Technol 61: 187–190.

48. WagsaterD, ZhuC, BjorkegrenJ, SkogsbergJ, ErikssonP (2011) MMP-2 and MMP-9 are prominent matrix metalloproteinases during atherosclerosis development in the Ldlr−/−Apob100/100 mouse. Int J Mol Med 28: 247–253.

49. IrizarryRA, BolstadBM, CollinF, CopeLM, HobbsB, et al. (2003) Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 31: e15.

50. EfronB, TibshiraniR, StoreyJ, TusherV (2001) Empirical Bayes analysis of a microarray experiment. J Am Stat Assoc 96: 1151–1160.

51. PawitanY, MichielsS, KoscielnyS, GusnantoA, PlonerA (2005) False discovery rate, sensitivity and sample size for microarray studies. Bioinformatics 21: 3017–3024.

52. Huang daW, ShermanBT, LempickiRA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4: 44–57.

53. Huang daW, ShermanBT, LempickiRA (2009) Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res 37: 1–13.

54. SchadtEE, MolonyC, ChudinE, HaoK, YangX, et al. (2008) Mapping the genetic architecture of gene expression in human liver. PLoS Biol 6: e107.

55. FaithJJ, HayeteB, ThadenJT, MognoI, WierzbowskiJ, et al. (2007) Large-scale mapping and validation of Escherichia coli transcriptional regulation from a compendium of expression profiles. PLoS Biol 5: e8.

56. MadarA, GreenfieldA, Vanden-EijndenE, BonneauR (2010) DREAM3: network inference using dynamic context likelihood of relatedness and the inferelator. PLoS One 5: e9803.

57. ShannonP, MarkielA, OzierO, BaligaNS, WangJT, et al. (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13: 2498–2504.

58. BasuSK, GoldsteinJL, AndersonGW, BrownMS (1976) Degradation of cationized low density lipoprotein and regulation of cholesterol metabolism in homozygous familial hypercholesterolemia fibroblasts. Proc Natl Acad Sci U S A 73: 3178–3182.

59. RedgraveTG, CarlsonLA (1979) Changes in plasma very low density and low density lipoprotein content, composition, and size after a fatty meal in normo- and hypertriglyceridemic man. J Lipid Res 20: 217–229.

60. ChristoffersenC, NielsenLB, AxlerO, AnderssonA, JohnsenAH, et al. (2006) Isolation and characterization of human apolipoprotein M-containing lipoproteins. J Lipid Res 47: 1833–1843.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 2
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#