Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 31.
Published in final edited form as: Arterioscler Thromb Vasc Biol. 2015 Feb;35(2):253–257. doi: 10.1161/ATVBAHA.114.305144

Lymphocytes in Atherosclerosis

Catherine C Hedrick 1
PMCID: PMC4327776  NIHMSID: NIHMS650544  PMID: 25609772

Introduction

Atherosclerosis is the major cause of cardiovascular disease, and cardiovascular disease remains the primary cause of death in the US. The primary complication for patients with Type 2 diabetes is cardiovascular disease18. As the incidences of Type 2 diabetes and obesity are drastically increasing in the US and now world-wide, understanding how atherosclerosis is accelerated in these diseases becomes even more important to discern.

While many scientific advances in research on atherosclerosis have been made by multiple groups over the last thirty years, particularly regarding the function of endothelial and smooth muscle biology and macrophage foam cell formation in atherosclerotic plaque development, the roles of immune cells in regulating atherosclerosis onset and progression are still being elucidated. It was widely believed that immune cells played little part in atherogenesis until 1986, when Hansson and colleagues reported the presence of lymphocytes within atherosclerotic lesions 912; yet it was many years later until it was widely accepted that immune cells play an important role in atherogenesis.

Since 1986, the viewpoint has changed significantly, but details on lymphocyte subsets and mechanisms are still emerging. Moreover, controversy still exists regarding the roles that some lymphocyte subsets play in atherosclerosis. These discrepancies in findings are likely due, at least partly, by differences in mouse models utilized, diet composition, and length of time on diet for atherosclerosis measurements. However, despite a few ongoing controversies, there are many studies on immune cells in atherogenesis in mice that show very clear findings10, 1223. For example, CD4+ T lymphocytes have been shown to accelerate atherosclerosis18, 2429. Activated CD4 T cells can be distinguished based on their phenotype, and the predominant CD4+ effector T cells studied to date in the context of atherosclerosis are Th1, Th2, and Th17 cells. The Th1 cell, a CD4+ T effector cell that produces IFNγ, is proatherogenic and very abundant in atherosclerotic lesions30. In contrast, very few Th2 cells are found in atherosclerotic plaques30 and their role in atherosclerosis remains unclear. Th17 cells produce IL-17A and IL-17F cytokines and have been localized to atherosclerotic lesions. However, the role of IL-17 in atherosclerosis remains somewhat controversial, as there are published studies that support both a pro-inflammatory as well as anti-inflammatory role for this cytokine in atherogenesis3136. In contrast, CD4+Foxp3+ Tregulatory cells (or ‘Tregs’), a suppressive subset of T cells, have been shown to be highly atheroprotective in mice and this has been confirmed in multiple studies17, 37,16, 19, 20, 38.

CD8 cells are much less studied in the context of atherosclerosis, but recent studies suggest that they may be proatherogenic39, although one very recent study found that a CD8+CD25+ subset of CD8 cells may be atheroprotective40. Expect to see more studies emerging on CD8 cells in atherosclerosis in the next few years as interest in this topic is expanding. Of note for further reading, a very nice detailed review by Li et al on T cells in atherosclerosis with a focus on lymphocyte homing to aortic tissue was published this year in ATVB41.

Moreover, little is known about the role of innate-like lymphocyte subsets (natural killer cells, γδ (gammadelta) cells, and NKT cells) in atherosclerosis. This article highlights recent studies published in ATVB on T and B lymphocyte subsets and innate-like lymphocyte subsets and their roles in atherosclerosis and related vascular diseases.

T lymphocytes

As noted above, T lymphocyte subsets have been found to play differing roles in atherosclerosis. Lichtman and colleagues found that numbers of atheroprotective Tregs are reduced in atherosclerotic mice during high cholesterol diet feeding, and that this was a reversible process: upon normalization of plasma cholesterol levels in the mice, Treg numbers were restored 42. Mechanisms for this are likely to be several, but these investigators found that high cholesterol diet feeding influenced Treg migration to aorta. Earlier studies have shown that the cholesterol content of lymphocytes impacts their proliferation. Wilhelm et al. reported in ATVB in 2009 that mice lacking apoA-I and functional HDL, had increased atherosclerosis and increased cholesterol-associated CD4 T lymphocyte proliferation and activation43. Studies by Bensinger44 and also by our own group45 found that sterol content of lymphocytes increased their proliferation, and this was primarily regulated by the ATP binding cassette transporter ABCG1. Whether the cholesterol content of Tregs directly impacts their proliferation and/or function is not known, and the role that HDL may play in this process warrants further investigation.

T regs may be also protective in other vascular diseases. In a recent study in ATVB, T regs were shown to protect against abdominal aortic aneurysm. In a well-established inducible mouse model of aneurysm using angiotensin-II (Ang-II), selective depletion of Tregs using CD25 antibodies enhanced susceptibility of mice to aneurysm and promoted aortic rupture46. Further, these authors found that IL-10 played an important role in the protection against Ang-II-induced aneurysm in this model. To address whether there is a link between T2D or obesity and lymphocyte function, recent work in ATVB shows that T cell frequencies are changed in the adipose tissue of obese subjects. Engleman and colleagues found that both visceral and subcutaneous fat depots in overweight and obese human subjects contained elevated numbers of both CD4 and CD8 T cells47. Furthermore, these investigators found that Th2 frequencies in both fat pad depots correlated with reduced incidence of insulin resistance. Thus, adipose tissue of obese subjects likely contains cytokines and antigenic stimuli for modulation of T cell numbers and activation. One possible mediator of increased T cell recruitment to adipose tissue is the chemokine receptor CXCR3. In obese mice, CXCR3 expression was higher on stromal vascular cells of adipose tissue48. CXCR3-deficient mice possessed fewer T cells in adipose depots and showed reductions in proinflammatory cytokines. A second novel mediator of T cell infiltration and accumulation in adipose is CD11a, a β2 integrin. Jiang et al showed that CD11a is upregulated on CD8 cells from obese mice, and that CD8 T cells from CD11a-defcient mice failed to migrate to adipose tissue in vivo49. This study is of interest as it supports a novel role for this integrin in lymphocyte homing to adipose tissue, which will be important for immunity associated with T2D and obesity.

Despite accepted knowledge that lymphocytes significantly impact lesion development, we know very little about the detailed mechanisms involved, nor do we know great detail about the cross-talk that occurs between myeloid cells and lymphocytes in the artery wall. Several studies in ATVB this year reported how novel cytokines and other novel T cell modulators impacted atherogenesis. For example, Interleukin-19 (IL-19) is an anti-inflammatory cytokine produced by Th2 lymphocytes. Atherosclerotic-susceptible LDLR−/− mice treated with recombinant IL-19 developed less aortic atherosclerosis, and this was accompanied by polarization of CD4+ lymphocytes to a Th2 phenotype, with decreased IFNγ and IL1β expression, and increased expression of GATA3 and Foxp3 transcription factors50. This study suggested that Il-19 was a potent inhibitor of atherosclerosis through its effect on T cell polarization. Another novel molecule, T-cell immunoglobulin and mucin domain-3 (Tim-3), acts as a negative regulator of immune responses. In a recent report in ATVB, treatment of mice with anti-Tim3 antibody depleted Tim-3 and increased lesion development 51. This was accompanied by increased numbers of activated CD4+ effector T cells and reduced numbers of Tregs. Related to Th2 polarization is a recent study in humans that indicated that increased numbers of Th2 lymphocytes in blood was associated with reduced risk of myocardial infarction52, suggesting that Th2 bias may indeed be atheroprotective.

Macrophages and dendritic cells are known to present antigen to T cells and also to secrete cytokines which participate in T cell phenotypic switching in the vascular microenvironment. Three studies published recently in ATVB cite new mechanisms by which myeloid cells influence T cell activation in atherosclerosis. One interesting paper in particular relayed findings where oxidized LDL, but not native LDL, stimulated DC activation53. These oxidized LDL-stimulated DC were able to induce T cell proliferation and activation and these DC polarized the CD4 cells to a Th1 or Th17 bias. Mechanistically, this was shown to be caused in part by action of heat shock proteins. Interestingly, treatment of oxidized LDL-stimulated DC with the anticoagulant Annexin A5 caused the DC to produce both IL-10 and TGFβ, which promoted differentiation of naive T cells to Treg cells in vitro. A second paper explored how TLR9 signaling in myeloid cells protected mice against atherosclerosis54. Although the exact mechanisms for the increased atherosclerosis in TLR9−/−apoE−/− mice are unclear, the data support the notion that TLR9 signaling may be atheroprotective: in the absence of TLR9 signaling, there is an accumulation of DC in atherosclerotic lesions which cause recruitment and activation of CD4+ T cells. Related to the above work on Tim-3, these investigators also found an increase in Tim-3 in the atherosclerotic lesions of TLR9-deficient mice, supporting the pro-atherogenic role for Tim-3 noted above.

In sum, new work published in ATVB showing a protective role for Tregs in other vascular diseases, including aneurysm, support the notion that Tregs serve a protective role in vascular disease. New studies in mice and humans with T2D and/or obesity suggest that adipose depots are hotspots for immune regulation, with increased myeloid cell content and supporting active CD4 and CD8 lymphocyte recruitment to these fat depots. Understanding the functions of lymphocytes in adipose tissue may aid in preventing complications associated with obesity and T2D. Finally, new mechanisms for how lymphocytes may impact atherosclerosis are emerging, including myeloid cell-lymphocyte crosstalk in the artery wall, thus, shedding light on possible new therapies for cardiovascular disease.

Innate lymphocytes

The ‘other lymphocytes’, or ‘unusual suspects’, as classified by Reardon and colleagues in 200555, include γδ (gammadelta) lymphocytes, NKT cells and NK cells. These cells make up a small percentage of lymphocytes, but they seem to pack a powerful punch, and as such, should not be underestimated in disease causation or protection. Gammadelta lymphocytes possess the γδ TCR and are enriched in skin and in tissues during inflammation. Many γδ cells do not require antigen processing and presentation in the context of MHC molecules to respond to antigen and many recognize non-peptide antigens, including phosphorylated nucleotides. In the context of atherosclerosis, γδ cells producing IL-17 were shown to be significantly elevated by almost 3-fold in aortas of apoE−/− mice fed a Western diet for 15 weeks33. However, we studied early atherosclerosis development in TCRδ−/−apoE−/− double KO mice and found no differences in atherosclerosis in mice lacking γδ cells, suggesting that they play a minor role, at least in early atherogenesis56. NKT cells, or natural killer T cells, possess an invariant T cell receptor (TCR) that recognizes self and foreign lipid antigens presented by the Class-I like molecule CD1d. The unique ability of NKT cells to recognize lipid antigens presented by CD1d suggests that they may play an important role in atherosclerosis, which is considered to be a lipid-driven disease. Most studies have reported that NKT cells are proatherogenic5764, including a recent study by Li et al65. A recent review on NKT cells in atherosclerosis by Bondarenko and colleagues was just published66, and interested readers are encouraged to study this article. Recently in ATVB, the absence of NKT cells in liver of Jα18-deficient mice which lack NKT cells, was associated with fewer and smaller adipocytes and increased lipolysis in adipose tissue, suggesting that NKT cells in liver can function to influence lipid metabolism in adipose67. However, CD1d-deficient mice (which also lack NKT cells) had increased adiposity68, suggesting that NKT cells may prevent metabolic dysfunction. More studies are needed to sort out these differences. Finally, natural killer (NK) cells, which are innate lymphocytes that do not possess a TCR and function early in the immune response to kill viruses and other pathogens, may also be important in atherosclerosis. A recent study by Kyaw and colleagues reports that depletion of NK cells in mice using anti-Asialo-GM1 antibodies attenuated atherosclerosis in ApoE−/− mice 69. In support of this notion is a recent human study in ATVB that shows that a subset of NK cells expressing CD94 was associated with increased risk of plaque rupture70. Kossmann et al71 also showed in mice that NK cells were involved in angiotensin II-mediated vascular dysfunction. These unusual lymphocyte suspects are being studied more and more in the context of vascular diseases, and it is becoming quite clear that these minor lymphocyte subsets play an important role in regulating vascular immunity.

B lymphocytes

The roles that B lymphocytes play in atherogenesis have been studied less than the roles of T lymphocytes to date, but the last few years have been clearly the time of the B cell. There are 2 main families of B cells: B1 and B2 cells. A review article on targeting B cells in atherosclerosis was very recently published in ATVB72, and the reader is referred to that paper for detailed information. Early on, B2 cells were thought to be atheroprotective73, 74 and this has been confirmed recently in an apoE−/− mouse model that examined resident B cells in aorta75. However, B2 cells have also been shown to be proatherogenic74, 76, so the role of B cells in atherosclerosis remains somewhat controversial. However, multiple studies support the concept that B1 cells are atheroprotective due to their production of natural IgM antibodies. A recent study in ATVB showed that mice lacking Id3 had fewer B1-a cells77 and reduced serum levels of the natural antibody E0678 that recognizes the phosphocholine (PC) headgroup of oxidized phospholipids77. B1-a cell proliferation was found to be caused by Id3-mediated regulation of IL-5 production by natural helper cells, another innate lymphoid cell77. In humans CD19+ B cells subsets have now been associated with risk of stroke. CD19+CD86+ B cells in blood were associated with higher incidence of developing stroke and CD19+CD40+ B cells were associated with lower incidence of stroke risk in a cardiovascular cohort of the Malmo Diet and Cancer Study79. Finally, Quax and colleagues have found a link between TLR4 signaling and B cell numbers and activation in atherosclerosis80. Taken together, these studies suggest that B cell subsets play a critical role in atherosclerosis and mechanisms that influence B cell function, including transcription factors and receptors, including TLRs, are likely important mediators of atherosclerosis.

Summary

In summary, new studies this year in ATVB continue to expand our knowledge of lymphocytes in atherosclerosis and other vascular diseases. Also, several studies reported new mechanisms for how lymphocytes, including B cells and innate lymphocyte subsets, impact atherosclerosis and vascular disease.

Acknowledgments

Research in the Hedrick laboratory is supported by NIH grants P01 HL055798, R01 HL112276, R01 HL085790, R01 HL097368, the American Heart Association, and the American Diabetes Association.

References

  • 1.Stratton IM, Adler AI, Neil HA, Matthews DR, Manley SE, Cull CA, Hadden D, Turner RC, Holman RR. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (ukpds 35): Prospective observational study. BMJ. 2000;321:405–412. doi: 10.1136/bmj.321.7258.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gu K, Cowie CC, Harris MI. Diabetes and decline in heart disease mortality in us adults. JAMA. 1999;281:1291–1297. doi: 10.1001/jama.281.14.1291. [DOI] [PubMed] [Google Scholar]
  • 3.Gregg EW, Gu Q, Cheng YJ, Narayan KM, Cowie CC. Mortality trends in men and women with diabetes, 1971 to 2000. Ann Intern Med. 2007;147:149–155. doi: 10.7326/0003-4819-147-3-200708070-00167. [DOI] [PubMed] [Google Scholar]
  • 4.Bertoni AG, Wong ND, Shea S, Ma S, Liu K, Preethi S, Jacobs DR, Jr, Wu C, Saad MF, Szklo M. Insulin resistance, metabolic syndrome, and subclinical atherosclerosis: The multi-ethnic study of atherosclerosis (mesa) Diabetes Care. 2007;30:2951–2956. doi: 10.2337/dc07-1042. [DOI] [PubMed] [Google Scholar]
  • 5.Kannel WB, McGee DL. Diabetes and cardiovascular disease. The framingham study. JAMA. 1979;241:2035–2038. doi: 10.1001/jama.241.19.2035. [DOI] [PubMed] [Google Scholar]
  • 6.Kannel WB, McGee DL. Diabetes and glucose tolerance as risk factors for cardiovascular disease: The framingham study. Diabetes Care. 1979;2:120–126. doi: 10.2337/diacare.2.2.120. [DOI] [PubMed] [Google Scholar]
  • 7.Stern MP. Diabetes and cardiovascular disease. The “common soil” hypothesis. Diabetes. 1995;44:369–374. doi: 10.2337/diab.44.4.369. [DOI] [PubMed] [Google Scholar]
  • 8.Gleissner CA, Galkina E, Nadler JL, Ley K. Mechanisms by which diabetes increases cardiovascular disease. Drug Discov Today Dis Mech. 2007;4:131–140. doi: 10.1016/j.ddmec.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jonasson L, Holm J, Skalli O, Bondjers G, Hansson GK. Regional accumulations of t cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis. 1986;6:131–138. doi: 10.1161/01.atv.6.2.131. [DOI] [PubMed] [Google Scholar]
  • 10.Hansson GK, Jonasson L, Lojsthed B, Stemme S, Kocher O, Gabbiani G. Localization of t lymphocytes and macrophages in fibrous and complicated human atherosclerotic plaques. Atherosclerosis. 1988;72:135–141. doi: 10.1016/0021-9150(88)90074-3. [DOI] [PubMed] [Google Scholar]
  • 11.Hansson GK, Holm J, Jonasson L. Detection of activated t lymphocytes in the human atherosclerotic plaque. Am J Pathol. 1989;135:169–175. [PMC free article] [PubMed] [Google Scholar]
  • 12.Hansson GK. Atherosclerosis--an immune disease: The anitschkov lecture 2007. Atherosclerosis. 2009;202:2–10. doi: 10.1016/j.atherosclerosis.2008.08.039. [DOI] [PubMed] [Google Scholar]
  • 13.Hansson GK. Regulation of immune mechanisms in atherosclerosis. Ann NY Acad Sci. 2001;947:157–165. [PubMed] [Google Scholar]
  • 14.Tobias P, Curtiss LK. Thematic review series: The immune system and atherogenesis. Paying the price for pathogen protection: Toll receptors in atherogenesis. J Lipid Res. 2005;46:404–411. doi: 10.1194/jlr.R400015-JLR200. [DOI] [PubMed] [Google Scholar]
  • 15.Reardon CA, Blachowicz L, White T, Cabana V, Wang Y, Lukens J, Bluestone J, Getz GS. Effect of immune deficiency on lipoproteins and atherosclerosis in male apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2001;21:1011–1016. doi: 10.1161/01.atv.21.6.1011. [DOI] [PubMed] [Google Scholar]
  • 16.Mallat Z, Gojova A, Brun V, Esposito B, Fournier N, Cottrez F, Tedgui A, Groux H. Induction of a regulatory t cell type 1 response reduces the development of atherosclerosis in apolipoprotein e-knockout mice. Circulation. 2003;108:1232–1237. doi: 10.1161/01.CIR.0000089083.61317.A1. [DOI] [PubMed] [Google Scholar]
  • 17.Ait-Oufella H, Salomon BL, Potteaux S, Robertson AK, Gourdy P, Zoll J, Merval R, Esposito B, Cohen JL, Fisson S, Flavell RA, Hansson GK, Klatzmann D, Tedgui A, Mallat Z. Natural regulatory t cells control the development of atherosclerosis in mice. Nat Med. 2006;12:178–180. doi: 10.1038/nm1343. [DOI] [PubMed] [Google Scholar]
  • 18.Buono C, Binder CJ, Stavrakis G, Witztum JL, Glimcher LH, Lichtman AH. T-bet deficiency reduces atherosclerosis and alters plaque antigen-specific immune responses. Proc Natl Acad Sci U S A. 2005;102:1596–1601. doi: 10.1073/pnas.0409015102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gotsman I, Grabie N, Gupta R, Dacosta R, MacConmara M, Lederer J, Sukhova G, Witztum JL, Sharpe AH, Lichtman AH. Impaired regulatory t-cell response and enhanced atherosclerosis in the absence of inducible costimulatory molecule. Circulation. 2006;114:2047–2055. doi: 10.1161/CIRCULATIONAHA.106.633263. [DOI] [PubMed] [Google Scholar]
  • 20.Gotsman I, Gupta R, Lichtman AH. The influence of the regulatory t lymphocytes on atherosclerosis. Arterioscler Thromb Vasc Biol. 2007;27:2493–2495. doi: 10.1161/ATVBAHA.107.153064. [DOI] [PubMed] [Google Scholar]
  • 21.Pinderski LJ, Fischbein MP, Subbanagounder G, Fishbein MC, Kubo N, Cheroutre H, Curtiss LK, Berliner JA, Boisvert WA. Overexpression of interleukin-10 by activated t lymphocytes inhibits atherosclerosis in ldl receptor-deficient mice by altering lymphocyte and macrophage phenotypes. Circ Res. 2002;90:1064–1071. doi: 10.1161/01.res.0000018941.10726.fa. [DOI] [PubMed] [Google Scholar]
  • 22.Schiller NK, Boisvert WA, Curtiss LK. Inflammation in atherosclerosis: Lesion formation in ldl receptor-deficient mice with perforin and lyst(beige) mutations. Arterioscler Thromb Vasc Biol. 2002;22:1341–1346. doi: 10.1161/01.atv.0000024082.46387.38. [DOI] [PubMed] [Google Scholar]
  • 23.Andersson J, Libby P, Hansson GK. Adaptive immunity and atherosclerosis. Clin Immunol. 2010;134:33–46. doi: 10.1016/j.clim.2009.07.002. [DOI] [PubMed] [Google Scholar]
  • 24.Stemme S, Faber B, Holm J, Wiklund O, Witztum JL, Hansson GK. T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc Natl Acad Sci US A. 1995;92:3893–3897. doi: 10.1073/pnas.92.9.3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Emeson EE, Shen ML, Bell CG, Qureshi A. Inhibition of atherosclerosis in cd4 t-cell-ablated and nude (nu/nu) c57bl/6 hyperlipidemic mice. Am J Pathol. 1996;149:675–685. [PMC free article] [PubMed] [Google Scholar]
  • 26.Zhou X, Nicoletti A, Elhage R, Hansson GK. Transfer of cd4(+) t cells aggravates atherosclerosis in immunodeficient apolipoprotein e knockout mice. Circulation. 2000;102:2919–2922. doi: 10.1161/01.cir.102.24.2919. [DOI] [PubMed] [Google Scholar]
  • 27.Zhou X, Robertson AK, Hjerpe C, Hansson GK. Adoptive transfer of cd4+ t cells reactive to modified low-density lipoprotein aggravates atherosclerosis. Arterioscler Thromb Vasc Biol. 2006;26:864–870. doi: 10.1161/01.ATV.0000206122.61591.ff. [DOI] [PubMed] [Google Scholar]
  • 28.Zhou X. Cd4+ t cells in atherosclerosis. Biomed Pharmacother. 2003;57:287–291. doi: 10.1016/s0753-3322(03)00082-9. [DOI] [PubMed] [Google Scholar]
  • 29.Huber SA, Sakkinen P, David C, Newell MK, Tracy RP. T helper-cell phenotype regulates atherosclerosis in mice under conditions of mild hypercholesterolemia. Circulation. 2001;103:2610–2616. doi: 10.1161/01.cir.103.21.2610. [DOI] [PubMed] [Google Scholar]
  • 30.Frostegard J, Ulfgren AK, Nyberg P, Hedin U, Swedenborg J, Andersson U, Hansson GK. Cytokine expression in advanced human atherosclerotic plaques: Dominance of pro-inflammatory (th1) and macrophage-stimulating cytokines. Atherosclerosis. 1999;145:33–43. doi: 10.1016/s0021-9150(99)00011-8. [DOI] [PubMed] [Google Scholar]
  • 31.Chen S, Crother TR, Arditi M. Emerging role of il-17 in atherosclerosis. J Innate Immun. 2010;2:325–333. doi: 10.1159/000314626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Erbel C, Chen L, Bea F, Wangler S, Celik S, Lasitschka F, Wang Y, Bockler D, Katus HA, Dengler TJ. Inhibition of il-17a attenuates atherosclerotic lesion development in apoe-deficient mice. Journal of immunology. 2009;183:8167–8175. doi: 10.4049/jimmunol.0901126. [DOI] [PubMed] [Google Scholar]
  • 33.Smith E, Prasad KM, Butcher M, Dobrian A, Kolls JK, Ley K, Galkina E. Blockade of interleukin-17a results in reduced atherosclerosis in apolipoprotein e-deficient mice. Circulation. 2010;121:1746–1755. doi: 10.1161/CIRCULATIONAHA.109.924886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Taleb S, Tedgui A, Mallat Z. Interleukin-17: Friend or foe in atherosclerosis? Curr Opin Lipidol. 2010;21:404–408. doi: 10.1097/MOL.0b013e32833dc7f9. [DOI] [PubMed] [Google Scholar]
  • 35.Taleb S, Tedgui A, Mallat Z. Il-17 and th17 cells in atherosclerosis: Subtle and contextual roles. Arterioscler Thromb Vasc Biol. 2014 doi: 10.1161/ATVBAHA.114.303567. [DOI] [PubMed] [Google Scholar]
  • 36.van Es T, van Puijvelde GH, Ramos OH, Segers FM, Joosten LA, van den Berg WB, Michon IM, de Vos P, van Berkel TJ, Kuiper J. Attenuated atherosclerosis upon il-17r signaling disruption in ldlr deficient mice. Biochemical and biophysical research communications. 2009;388:261–265. doi: 10.1016/j.bbrc.2009.07.152. [DOI] [PubMed] [Google Scholar]
  • 37.Entin-Meer M, Afek A, George J. Regulatory t-cells, foxp3 and atherosclerosis. Adv Exp Med Biol. 2009;665:106–114. doi: 10.1007/978-1-4419-1599-3_8. [DOI] [PubMed] [Google Scholar]
  • 38.Taleb S, Tedgui A, Mallat Z. Regulatory t-cell immunity and its relevance to atherosclerosis. J Intern Med. 2008;263:489–499. doi: 10.1111/j.1365-2796.2008.01944.x. [DOI] [PubMed] [Google Scholar]
  • 39.Kyaw T, Winship A, Tay C, Kanellakis P, Hosseini H, Cao A, Li P, Tipping P, Bobik A, Toh BH. Cytotoxic and proinflammatory cd8+ t lymphocytes promote development of vulnerable atherosclerotic plaques in apoe-deficient mice. Circulation. 2013;127:1028–1039. doi: 10.1161/CIRCULATIONAHA.112.001347. [DOI] [PubMed] [Google Scholar]
  • 40.Zhou J, Dimayuga PC, Zhao X, Yano J, Lio WM, Trinidad P, Honjo T, Cercek B, Shah PK, Chyu KY. Cd8(+)cd25(+) t cells reduce atherosclerosis in apoe(−/−) mice. Biochem Biophys Res Commun. 2014;443:864–870. doi: 10.1016/j.bbrc.2013.12.057. [DOI] [PubMed] [Google Scholar]
  • 41.Li J, Ley K. Lymphocyte migration into atherosclerotic plaque. Arterioscler Thromb Vasc Biol. 2014 doi: 10.1161/ATVBAHA.114.303227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Maganto-Garcia E, Tarrio ML, Grabie N, Bu DX, Lichtman AH. Dynamic changes in regulatory t cells are linked to levels of diet-induced hypercholesterolemia. Circulation. 2011;124:185–195. doi: 10.1161/CIRCULATIONAHA.110.006411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wilhelm AJ, Zabalawi M, Grayson JM, Weant AE, Major AS, Owen J, Bharadwaj M, Walzem R, Chan L, Oka K, Thomas MJ, Sorci-Thomas MG. Apolipoprotein a-i and its role in lymphocyte cholesterol homeostasis and autoimmunity. Arterioscler Thromb Vasc Biol. 2009;29:843–849. doi: 10.1161/ATVBAHA.108.183442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bensinger SJ, Bradley MN, Joseph SB, Zelcer N, Janssen EM, Hausner MA, Shih R, Parks JS, Edwards PA, Jamieson BD, Tontonoz P. Lxr signaling couples sterol metabolism to proliferation in the acquired immune response. Cell. 2008;134:97–111. doi: 10.1016/j.cell.2008.04.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Armstrong AJ, Gebre AK, Parks JS, Hedrick CC. Atp-binding cassette transporter g1 negatively regulates thymocyte and peripheral lymphocyte proliferation. Journal of immunology. 2010;184:173–183. doi: 10.4049/jimmunol.0902372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ait-Oufella H, Wang Y, Herbin O, Bourcier S, Potteaux S, Joffre J, Loyer X, Ponnuswamy P, Esposito B, Dalloz M, Laurans L, Tedgui A, Mallat Z. Natural regulatory t cells limit angiotensin ii-induced aneurysm formation and rupture in mice. Arterioscler Thromb Vasc Biol. 2013;33:2374–2379. doi: 10.1161/ATVBAHA.113.301280. [DOI] [PubMed] [Google Scholar]
  • 47.McLaughlin T, Liu LF, Lamendola C, Shen L, Morton J, Rivas H, Winer D, Tolentino L, Choi O, Zhang H, Hui Yen Chng M, Engleman E. T-cell profile in adipose tissue is associated with insulin resistance and systemic inflammation in humans. Arterioscler Thromb Vasc Biol. 2014;34:2637–2643. doi: 10.1161/ATVBAHA.114.304636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Rocha VZ, Folco EJ, Ozdemir C, Sheikine Y, Christen T, Sukhova GK, Tang EH, Bittencourt MS, Santos RD, Luster AD, Cohen DE, Libby P. Cxcr3 controls t-cell accumulation in fat inflammation. Arterioscler Thromb Vasc Biol. 2014;34:1374–1381. doi: 10.1161/ATVBAHA.113.303133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Jiang E, Perrard XD, Yang D, Khan IM, Perrard JL, Smith CW, Ballantyne CM, Wu H. Essential role of cd11a in cd8+ t-cell accumulation and activation in adipose tissue. Arterioscler Thromb Vasc Biol. 2014;34:34–43. doi: 10.1161/ATVBAHA.113.302077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ellison S, Gabunia K, Kelemen SE, England RN, Scalia R, Richards JM, Orr AW, Traylor JG, Jr, Rogers T, Cornwell W, Berglund LM, Goncalves I, Gomez MF, Autieri MV. Attenuation of experimental atherosclerosis by interleukin-19. Arterioscler Thromb Vasc Biol. 2013;33:2316–2324. doi: 10.1161/ATVBAHA.113.301521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Foks AC, Ran IA, Wasserman L, Frodermann V, Ter Borg MN, de Jager SC, van Santbrink PJ, Yagita H, Akiba H, Bot I, Kuiper J, van Puijvelde GH. T-cell immunoglobulin and mucin domain 3 acts as a negative regulator of atherosclerosis. Arterioscler Thromb Vasc Biol. 2013;33:2558–2565. doi: 10.1161/ATVBAHA.113.301879. [DOI] [PubMed] [Google Scholar]
  • 52.Engelbertsen D, Andersson L, Ljungcrantz I, Wigren M, Hedblad B, Nilsson J, Bjorkbacka H. T-helper 2 immunity is associated with reduced risk of myocardial infarction and stroke. Arterioscler Thromb Vasc Biol. 2013;33:637–644. doi: 10.1161/ATVBAHA.112.300871. [DOI] [PubMed] [Google Scholar]
  • 53.Liu A, Ming JY, Fiskesund R, Ninio E, Karabina SA, Bergmark C, Frostegard AG, Frostegard J. Induction of dendritic cell-mediated t-cell activation by modified but not native low-density lipoprotein in humans and inhibition by annexin a5: Involvement of heat shock proteins. Arterioscler Thromb Vasc Biol. 2014 doi: 10.1161/ATVBAHA.114.304342. [DOI] [PubMed] [Google Scholar]
  • 54.Koulis C, Chen YC, Hausding C, Ahrens I, Kyaw TS, Tay C, Allen T, Jandeleit-Dahm K, Sweet MJ, Akira S, Bobik A, Peter K, Agrotis A. Protective role for toll-like receptor-9 in the development of atherosclerosis in apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2014;34:516–525. doi: 10.1161/ATVBAHA.113.302407. [DOI] [PubMed] [Google Scholar]
  • 55.Vanderlaan PA, Reardon CA. Thematic review series: The immune system and atherogenesis. The unusual suspects:An overview of the minor leukocyte populations in atherosclerosis. J Lipid Res. 2005;46:829–838. doi: 10.1194/jlr.R500003-JLR200. [DOI] [PubMed] [Google Scholar]
  • 56.Cheng HY, Wu R, Hedrick CC. Gammadelta (gammadelta) t lymphocytes do not impact the development of early atherosclerosis. Atherosclerosis. 2014;234:265–269. doi: 10.1016/j.atherosclerosis.2014.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Aslanian AM, Chapman HA, Charo IF. Transient role for cd1d-restricted natural killer t cells in the formation of atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2005;25:628–632. doi: 10.1161/01.ATV.0000153046.59370.13. [DOI] [PubMed] [Google Scholar]
  • 58.Major AS, Singh RR, Joyce S, Van Kaer L. The role of invariant natural killer t cells in lupus and atherogenesis. Immunol Res. 2006;34:49–66. doi: 10.1385/ir:34:1:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Major AS, Wilson MT, McCaleb JL, Ru Su Y, Stanic AK, Joyce S, Van Kaer L, Fazio S, Linton MF. Quantitative and qualitative differences in proatherogenic nkt cells in apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2004;24:2351–2357. doi: 10.1161/01.ATV.0000147112.84168.87. [DOI] [PubMed] [Google Scholar]
  • 60.Nakai Y, Iwabuchi K, Fujii S, Ishimori N, Dashtsoodol N, Watano K, Mishima T, Iwabuchi C, Tanaka S, Bezbradica JS, Nakayama T, Taniguchi M, Miyake S, Yamamura T, Kitabatake A, Joyce S, Van Kaer L, Onoe K. Natural killer t cells accelerate atherogenesis in mice. Blood. 2004;104:2051–2059. doi: 10.1182/blood-2003-10-3485. [DOI] [PubMed] [Google Scholar]
  • 61.Rogers L, Burchat S, Gage J, Hasu M, Thabet M, Willcox L, Ramsamy TA, Whitman SC. Deficiency of invariant v alpha 14 natural killer t cells decreases atherosclerosis in ldl receptor null mice. Cardiovasc Res. 2008;78:167–174. doi: 10.1093/cvr/cvn005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tupin E, Nicoletti A, Elhage R, Rudling M, Ljunggren HG, Hansson GK, Berne GP. Cd1d-dependent activation of nkt cells aggravates atherosclerosis. J Exp Med. 2004;199:417–422. doi: 10.1084/jem.20030997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.van Puijvelde GH, van Wanrooij EJ, Hauer AD, de Vos P, van Berkel TJ, Kuiper J. Effect of natural killer t cell activation on the initiation of atherosclerosis. Thromb Haemost. 2009;102:223–230. doi: 10.1160/TH09-01-0020. [DOI] [PubMed] [Google Scholar]
  • 64.VanderLaan PA, Reardon CA, Sagiv Y, Blachowicz L, Lukens J, Nissenbaum M, Wang CR, Getz GS. Characterization of the natural killer t-cell response in an adoptive transfer model of atherosclerosis. Am J Pathol. 2007;170:1100–1107. doi: 10.2353/ajpath.2007.060188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Li Y, To K, Kanellakis P, Hosseini H, Deswaerte V, Tipping P, Smyth MJ, Toh BH, Bobik A, Kyaw T. Cd4+ natural killer t cells potently augment aortic root atherosclerosis by perforin- and granzyme b-dependent cytotoxicity. Circ Res. 2014 doi: 10.1161/CIRCRESAHA.116.304734. [DOI] [PubMed] [Google Scholar]
  • 66.Bondarenko S, Catapano AL, Norata GD. The cd1d-natural killer t cell axis in atherosclerosis. J Innate Immun. 2014;6:3–12. doi: 10.1159/000351034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Strodthoff D, Lundberg AM, Agardh HE, Ketelhuth DF, Paulsson-Berne G, Arner P, Hansson GK, Gerdes N. Lack of invariant natural killer t cells affects lipid metabolism in adipose tissue of diet-induced obese mice. Arterioscler Thromb Vasc Biol. 2013;33:1189–1196. doi: 10.1161/ATVBAHA.112.301105. [DOI] [PubMed] [Google Scholar]
  • 68.Martin-Murphy BV, You Q, Wang H, De La Houssaye BA, Reilly TP, Friedman JE, Ju C. Mice lacking natural killer t cells are more susceptible to metabolic alterations following high fat diet feeding. PLoS One. 2014;9:e80949. doi: 10.1371/journal.pone.0080949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Selathurai A, Deswaerte V, Kanellakis P, Tipping P, Toh BH, Bobik A, Kyaw T. Natural killer (nk) cells augment atherosclerosis by cytotoxic-dependent mechanisms. Cardiovasc Res. 2014;102:128–137. doi: 10.1093/cvr/cvu016. [DOI] [PubMed] [Google Scholar]
  • 70.Martinez-Rodriguez JE, Munne-Collado J, Rasal R, Cuadrado E, Roig L, Ois A, Muntasell A, Baro T, Alameda F, Roquer J, Lopez-Botet M. Expansion of the nkg2c+ natural killer-cell subset is associated with high-risk carotid atherosclerotic plaques in seropositive patients for human cytomegalovirus. Arterioscler Thromb Vasc Biol. 2013;33:2653–2659. doi: 10.1161/ATVBAHA.113.302163. [DOI] [PubMed] [Google Scholar]
  • 71.Kossmann S, Schwenk M, Hausding M, Karbach SH, Schmidgen MI, Brandt M, Knorr M, Hu H, Kroller-Schon S, Schonfelder T, Grabbe S, Oelze M, Daiber A, Munzel T, Becker C, Wenzel P. Angiotensin ii-induced vascular dysfunction depends on interferon-gamma-driven immune cell recruitment and mutual activation of monocytes and nk-cells. Arterioscler Thromb Vasc Biol. 2013;33:1313–1319. doi: 10.1161/ATVBAHA.113.301437. [DOI] [PubMed] [Google Scholar]
  • 72.Tsiantoulas D, Sage AP, Mallat Z, Binder CJ. Targeting b cells in atherosclerosis: Closing the gap from bench to bedside. Arterioscler Thromb Vasc Biol. 2014 doi: 10.1161/ATVBAHA.114.303569. [DOI] [PubMed] [Google Scholar]
  • 73.Caligiuri G, Nicoletti A, Poirier B, Hansson GK. Protective immunity against atherosclerosis carried by b cells of hypercholesterolemic mice. J Clin Invest. 2002;109:745–753. doi: 10.1172/JCI07272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ait-Oufella H, Herbin O, Bouaziz JD, Binder CJ, Uyttenhove C, Laurans L, Taleb S, Van Vre E, Esposito B, Vilar J, Sirvent J, Van Snick J, Tedgui A, Tedder TF, Mallat Z. B cell depletion reduces the development of atherosclerosis in mice. J Exp Med. 2010;207:1579–1587. doi: 10.1084/jem.20100155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Doran AC, Lipinski MJ, Oldham SN, Garmey JC, Campbell KA, Skaflen MD, Cutchins A, Lee DJ, Glover DK, Kelly KA, Galkina EV, Ley K, Witztum JL, Tsimikas S, Bender TP, McNamara CA. B-cell aortic homing and atheroprotection depend on id3. Circ Res. 2012;110:e1–12. doi: 10.1161/CIRCRESAHA.111.256438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kyaw T, Tay C, Khan A, Dumouchel V, Cao A, To K, Kehry M, Dunn R, Agrotis A, Tipping P, Bobik A, Toh BH. Conventional b2 b cell depletion ameliorates whereas its adoptive transfer aggravates atherosclerosis. J Immunol. 2010;185:4410–4419. doi: 10.4049/jimmunol.1000033. [DOI] [PubMed] [Google Scholar]
  • 77.Perry HM, Oldham SN, Fahl SP, Que X, Gonen A, Harmon DB, Tsimikas S, Witztum JL, Bender TP, McNamara CA. Helix-loop-helix factor inhibitor of differentiation 3 regulates interleukin-5 expression and b-1a b cell proliferation. Arterioscler Thromb Vasc Biol. 2013;33:2771–2779. doi: 10.1161/ATVBAHA.113.302571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chou MY, Hartvigsen K, Hansen LF, Fogelstrand L, Shaw PX, Boullier A, Binder CJ, Witztum JL. Oxidation-specific epitopes are important targets of innate immunity. J Intern Med. 2008;263:479–488. doi: 10.1111/j.1365-2796.2008.01968.x. [DOI] [PubMed] [Google Scholar]
  • 79.Mantani PT, Ljungcrantz I, Andersson L, Alm R, Hedblad B, Bjorkbacka H, Nilsson J, Fredrikson GN. Circulating cd40+ and cd86+ b cell subsets demonstrate opposing associations with risk of stroke. Arterioscler Thromb Vasc Biol. 2014;34:211–218. doi: 10.1161/ATVBAHA.113.302667. [DOI] [PubMed] [Google Scholar]
  • 80.Karper JC, de Jager SC, Ewing MM, de Vries MR, Bot I, van Santbrink PJ, Redeker A, Mallat Z, Binder CJ, Arens R, Jukema JW, Kuiper J, Quax PH. An unexpected intriguing effect of toll-like receptor regulator rp105 (cd180) on atherosclerosis formation with alterations on b-cell activation. Arterioscler Thromb Vasc Biol. 2013;33:2810–2817. doi: 10.1161/ATVBAHA.113.301882. [DOI] [PubMed] [Google Scholar]

RESOURCES