Abstract
The diverse leukocyte infiltrate in atherosclerotic mouse aortas was recently analyzed in 9 single cell RNA-Seq (scRNA-Seq) and 2 mass cytometry (CyTOF) studies. In a comprehensive meta-analysis, we confirm four known macrophage subsets: resident, inflammatory, IFNIC and Trem2 foamy macrophages and identify a new macrophage subset resembling cavity macrophages. We also find that monocytes, neutrophils, dendritic cells, natural killer cells, innate lymphoid cells-2 (ILC2) and CD8 T cells form prominent and separate immune cell populations in atherosclerotic aortas. Many CD4 T cells express interleukin (IL)-17 and the chemokine receptor CXCR6. A small number of Tregs and Th1 cells is also identified. Immature and naïve T cells are present in both healthy and atherosclerotic aortas. Our meta-analysis overcomes limitations of individual studies that, because of their experimental approach, over- or under-represent certain cell populations. CyTOF studies demonstrate that cell surface phenotype provides valuable information beyond the cell transcriptomes. The present analysis helps resolve some long-standing controversies in the field. First, Trem2+ foamy macrophages are not pro-inflammatory, but interferon-inducible cell (IFNIC) and inflammatory macrophages are. Second, about half of all foam cells are smooth muscle cell-derived, retaining smooth muscle cell transcripts rather than transdifferentiating to macrophages. Third, Pf4, which had been considered specific for platelets and megakaryocytes, is also prominently expressed in the main population of resident vascular macrophages. Fourth, a new type of resident macrophage shares transcripts with cavity macrophages. Finally, the discovery of a prominent ILC2 cluster links the scRNA-Seq work to recent flow cytometry data suggesting a strong atheroprotective role of ILC2 cells. This resolves apparent discrepancies regarding the role of Th2 cells in atherosclerosis based on studies that pre-dated the discovery of ILC2 cells.
Keywords: atherosclerosis, scRNA-seq, CyTOF, macrophages, T cells, Vascular Biology
1. Introduction
Atherosclerosis is a disease of large and mid-sized arteries with devastating consequences. The disease process is initiated by low density lipoprotein (LDL) accumulating in the subendothelial space of arteries. Rapidly, resident vascular macrophages and lymphocytes expand in number and differentiate in phenotype. Early on, neutrophils and monocytes are recruited, along with all known cell types of the adaptive immune system. Mature atherosclerotic lesions therefore contain a wide variety of immune cells.
In early studies, the immune cell infiltrate was characterized by immunohistochemistry1. Later, methods were developed to generate single cell suspensions from mouse aorta2 and human atherosclerotic lesions3. Recently, high dimensional methods of analysis became available that generated a plethora of data allowing fine-grained and comprehensive characterization of the immune cell content of atherosclerotic lesions. Mass cytometry4, 5 allows assessment of up to 42 cell surface and intracellular markers. Single cell RNA-sequencing (scRNA-Seq) yields expression matrices of thousands of genes per cell, depending on the single cell sequencing method and platform6. The technical aspects of these high dimensional methods in vascular biology, including quality controls and validation, have recently been reviewed6.
This resource article is focused on leukocyte diversity in atherosclerosis. We assembled a team of specialists in the field to provide comprehensive and, where possible, authoritative information of the immune cell phenotypes as defined by cell surface phenotype, intracellular proteins and gene expression. We will only discuss mouse data, because there are too few studies of leukocytes in human atherosclerosis7, 8. Since genetic models of atherosclerosis (Apoe−/−, Ldr−/−) were generated in 19929–11, the mouse has become the de facto workhorse of atherosclerosis research. Later, more sophisticated mouse models of atherosclerosis were generated12, 13, but these are not as widely used.
One goal of this review is to assemble lists of surface markers and genes (“gene signature”) that can be used to delineate the various immune cell types. Of note, we are not concerned with the ontogeny of the immune cells in atherosclerosis. It is known that tissue resident vascular macrophages in mice are not monocyte-derived, are largely seeded before birth and can proliferate locally14. Other macrophages in atherosclerosis are derived from infiltrating monocytes15. We are also not concerned with cell lineages. In immunology, the relationship between cell types and their precursors, and whether cell types can transdifferentiate into other cell types, is often quite controversial. As a case in point, there is excellent evidence for16 and against17 conversion of regulatory T cells to other T cell types. Thus, we remain agnostic with respect to ontogeny and lineage. Rather, we are focusing on describing the cell types and states in atherosclerotic lesions (“what is”). The goal is to create a resource that will promote systematic and mechanistic investigations. Excellent reviews exist on immune cells in the heart18, 19. Neither heart nor veins or other blood vessels will be considered here. Another goal is to promote clarity in nomenclature, which will enable the field to move forward more rapidly. A distant goal is to translate findings in experimental atherosclerosis into prevention and therapy strategies for people with cardiovascular disease.
This review was spawned at a meeting that Andreas Zirlik organized in Graz, Austria in 2019. This meeting was focused on single cell methods in atherosclerosis research. Several of the authors were present at this meeting. Klaus Ley approached the editors of Circulation Research with the idea of putting together a broad-based review. Bioinformatics re-analysis of existing data resulted in this review becoming a resource paper. Computational biologists sifted through the data to identify transcriptomes, gene signatures and surface markers across labs, approaches and platforms. Most authors who published on mouse atherosclerosis using high dimensional single cell methods accepted the invitation to contribute.
This review is focused on CD4 and CD8 αβ T cells, γδ T cells, NK and NKT cells, B cells, neutrophils, macrophages, monocytes and dendritic cells. In addition, eosinophils, mast cells and innate lymphocyte-like cells (ILCs) are found in atherosclerotic lesions. Certainly, other cell types not mentioned here will emerge as important players in atherosclerosis. In fact, single cell technologies have a unique potential to discover new cell types.
2. Introduction of Immune Cell Types
T cells are cells of the adaptive immune system that develop in the thymus, where they find niche conditions conducive to rearrangement, under the control of the recombinase activating genes (RAG)-1 and 2, of their T cell receptor (TCR) γδ or αβ subunits, expression of their unique TCRs, positive and negative selection, and maturation20. CD4 T cells recognize antigenic peptides bound to major histocompatibility complex (MHC)-II and are either regulatory or helper T cells. CD8 T cells recognize peptide antigens bound to MHC-I and become cytotoxic T lymphocytes (CTLs). Some αβ T cells recognize glycolipids bound to the MHC-like molecule CD1d and become NKT cells21.
CD4 T cells recognizing self antigens are mostly regulatory T cells. Regulatory CD4+ T cells (Treg) represent immunosuppressive T cells that are defined by the transcription factor Foxp3 and IL-2 receptor α chain (CD25) expression22, 23. Natural Tregs (nTregs) are thymus-derived, whereas peripherally induced Tregs (iTregs) are generated in response to antigen recognition in the presence of transforming growth factor beta (TGF-β)24. Tregs dampen inflammation by limiting the proliferation and function of effector CD4 and CD8 T cells.
Co-stimulation and antigen presentation by antigen-presenting cells induces T cell activation and differentiation into functionally and phenotypically distinct T-helper (TH) types that contribute to atherosclerosis in a subset-dependent manner25. Follicular helper (TFH) cells express ICOS and the chemokine receptor CXCR5. They stay in lymphoid organs and have important helper functions for B cell maturation.
T-helper1 (TH1) CD4+ T cells express the lineage-defining transcription factor T-bet (gene name Tbx21) and the pro-atherogenic cytokine IFN-γ26, 27. The deletion of either Tbx21 or Ifng markedly reduce atherosclerosis28, 29. TH2 cells express the lineage-defining transcription factor GATA3 and secrete interleukins (IL)-4, IL-5, IL-10, and IL-1330. Dependent on the context and model analyzed, TH2 cells have been reported to contribute to atheroprogression or -protection31, 32. TH17 cells are characterized by the transcription factor RORγt and secrete the cytokines IL-17A, IL-17F, and IL-21. The contribution of TH17 to atherosclerosis is controversial, as the hallmark cytokine IL-17 can hamper or enhance atherosclerosis. This is likely related to different functions of different types of IL-17-expressing cells: Th17-Tregs have regulatory function (atheroprotective), whereas true Th17 cells and γδ T cells expressing IL-17 are pro-atherogenic. TH9 T cells express the cytokine IL-9 and the transcription factor PU.1 and accelerate atherosclerosis33.
Tregs prevent the progression of atherosclerosis by multiple mechanisms, including the secretion of IL-10 and TGF-β34, 35. At later stages of atherosclerosis, Tregs vanish36 and may obtain a TH1-like phenotype that includes expression of IFN-γ37, 38. While it was proposed that differentiation of T cells to distinct effector TH cell lineages is final and irreversible, accumulating evidence suggests that TH cells, and in particular inducible Tregs and TH17 cells, are flexible in nature and can be reprogrammed to unique mixed phenotypes or can re-differentiate to other subset of TH cells. TH1-like Tregs expressing IFN-γ and Foxp3 are found in tissues and the peripheral blood of patients with type 1 diabetes, multiple sclerosis, and arthritis39. IL-17+ Tregs are detected in rheumatoid arthritis, atopic asthma, and colorectal cancer, whereas GATA3+Foxp3+ cells are found in patients with food allergies. These findings suggest that the microenvironment promotes reprogramming of TH cells under pathological conditions.
CD8 T cells express inflammatory cytokines, cytotoxins and death-inducing proteins including IFNγ, TNF, granzymes A and B, perforin, FasL and TRAIL. Although CD8 T cells are generally considered pro-inflammatory and cytotoxic, CD25-expressing CD8 regulatory T cells, present in humans and mice, may modulate immune responses42. Once they are activated by antigen-presenting cells in an MHC-I-dependent manner, CD8 T cells can potently induce target cell death via either cytotoxins, cytokines or death-inducing proteins43. In healthy arteries, CD8 T cells are found in the adventitia. CD8 T cell abundance increases in atherosclerosis, where they populate the plaque shoulder and the area around necrotic cores44. In human rupture-prone lesions, cytotoxic CD8 T cells constitute up to 50% of CD45+ leukocytes45. CD8 T cells progressively accumulate but abruptly decline after plaque rupture46, 47. CD8 T cells promote atherosclerosis by increasing vascular inflammation and apoptotic cell numbers in lesions48, 49. CD8 T cells can also promote atherosclerosis by controlling monopoiesis and circulating monocyte levels44, 50. Some CD8 T cells recognize epitopes in ApoB, but most CD8 T cells in arteries have unknown specificities.
NKT cells are a minor T cell subset. They are considered a bridge between innate and adaptive immunity. NKT cells do not have immunological memory51. Besides innate receptor-dependent activation, NKT cells are activated by CD1d-bound glycolipid antigens51. Upon activation, they rapidly release large amounts of cytokines and cytotoxins52, which can aggravate atherosclerosis53–55. Among different subsets, NKT cell research has been focused on invariant NKT (iNKT) cells, also known as type I NKT cells expressing an invariant TCR α chain (Vα14-Jα18 in mouse). Phenotypically, NKT cells can be identified by using combinations of the NK cell marker, killer cell lectin-like receptor subfamily B member 1, also known as KLRB1, NK1.1 or CD161 and the T cell-defining marker CD3. Human and mouse work using lineage tracking or double immunostaining have identified the presence of NKT cells in atherosclerotic lesions54, 56, 57. Recent single-cell immunophenotyping methods have identified NKT cells in human and mouse atherosclerotic lesions7.
γδ T cells were first reported in human atherosclerotic lesions in 199358, yet their exact role in atherosclerosis remains unclear. γδ T cells are a minor subset (1–10% of T cells)59. They express TCRs composed of γ- and δ-chains that are activated independent of MHC. Upon activation through innate immune receptors γδ T cells can produce large amounts of inflammatory cytokines, chemokines and cytotoxins60. CD27+ γδ T cells secrete IFN-γ and CD27− γδ T cells IL-17 61, 62. Another subset of γδ T cells expressing the Vγ9Vδ2 TCR are reported to be cytotoxic63.
NK cells are cytotoxic cells that kill cells that lack expression of MHC-I. NK cells have no direct effect on atherosclerosis or lesion phenotype64. However, NK cells may contribute to atherosclerotic lesion formation when activated, such as under conditions of chronic viral infection64, 65.
B cells are adaptive immune cells, but unlike T cells, do not require the thymus for development. Like the TCR, the B cell receptor (BCR) must undergo V(D)J rearrangement under the Rag genes. These “germline-encoded” BCRs have low affinity to antigen. Immature B cells display IgM and IgD with the same sequence as their BCR. In the presence of follicular helper T cells (TFH), B cells can switch their isotype to various IgG isotypes, IgE or IgA. In germinal centers, their BCR undergoes many cycles of affinity maturation. This is under the control of the AID enzyme and results in high affinity antibodies. B cells express CD19 and CD20.
Many IgM-producing B cells are B1 cells, further divided into B1a and B1b based on the expression of CD5. B2 cells are much more abundant than B1 cells in spleen, bone marrow and blood and make IgG, IgE and IgA. Both B1 and B2 cells express MHC-II and can present antigenic peptides to CD4 T cells. One specialized type of B1 cells are Innate Response Activator (IRA) B cells, which are strongly pro-inflammatory by expressing Csf2 (GM-CSF)66. IRA B cells aggravate atherosclerosis by stimulating TH-1 adaptive immunity67.
In healthy arteries, B cells are found in the adventitia2 and perivascular adipose tissue (PVAT)68. B cells in the artery wall and adventitia of normal arteries probably do not secrete antibodies, but can traffic to other sites and become antibody-secreting plasma cells. Antibody production in the artery appears localized to PVAT68 and arterial tertiary lymphoid organs (ATLOs) adjacent to advanced plaques69.
Neutrophils are short-lived cells of the innate immune system. They are the main defenders against bacterial and fungal infections. A few neutrophils are present in normal arteries. Under conditions of atherosclerosis, some neutrophils are found on the endothelial surface and others in the plaque. Although their numbers are small, neutrophils have important roles in atherosclerosis and thrombosis70. Neutrophils are highly granular cells with a distinct lobed nuclear morphology. They are typically produced in the bone marrow in a process called granulopoiesis. Under conditions of metabolic stress such has hypercholesterolemia, however, neutrophil production also occurs in extramedullary tissues including the spleen, thus contributing to heightened circulating neutrophil counts71. Neutrophils can be identified in mouse blood as well as in single cell suspensions of tissues including aortic tissue as CD45+CD11b+Ly6G+CD11572.
Monocytes are innate immune cells that express CD115 (Csf1r) and CD11b. In mice, they come in two main flavors, classical and non-classical. Classical monocytes express Ly-6C and the chemokine receptor CCR273. Non-classical monocytes highly express the transcription factor Nr4a174 and the chemokine receptor CX3CR175. In healthy arteries, monocytes are rare. However, in atherosclerosis-prone regions (such as the lesser curvature of the ascending aortic arch), classical monocytes can be recruited through VCAM1-dependent interactions on vascular endothelium76, 77. Monocytes rapidly infiltrate atherosclerotic lesions. Some differentiate into macrophages, others to monocyte-derived dendritic cells, and some stay monocytes. Some monocytes express high levels of MHC-II and can present antigenic peptides to CD4 T cells. Non-classical monocytes patrol the endothelial surface and promote endothelial integrity78. Patrolling is highly intensified under conditions of atherosclerosis79, 80. Nonclassical monocytes patrol in response to oxidized LDL and ingest greater amounts of oxidized LDL than classical monocytes, processes that are dependent upon the scavenger receptor CD3680.
Dendritic cells (DCs) are found in secondary lymphoid organs, where they present antigens to naive T cells and start a specific immune response. DCs can be divided into conventional DCs (cDCs, CD11c+) and plasmacytoid DCs (pDCs, CD123+). Among the conventional DCs, lymphoid organ resident CD8α+ and non-lymphoid organ CD103+ type 1 cDC1 expressing Xcr1 and Clec9a (DNGR1) can be discriminated from cDC281. Plasmacytoid DCs produce type I interferons (IFNα and β)82.
Macrophages are the most common and probably the most important cell type in atherosclerosis. They express F4/80 and CD64, which is an activating Fcγ receptor. In healthy arteries, macrophages are in the adventitia, where they contribute to the physiology and diameter of the vessel wall via cross-talk with smooth muscle cells83. These vascular macrophages are largely derived from embryonal precursor cells and self-renew in situ14. A small subset of macrophages is located below the endothelium in certain areas of the arterial circulation such as the aortic arch. Atherosclerosis is characterized by an accumulation of a very large number of macrophages, derived from infiltrating monocytes and proliferating vascular macrophages15. Macrophages function to clear apoptotic cells (efferocytosis), to phagocytose debris, to produce inflammatory and inflammation-resolving cytokines and lipid meditators, and to present antigens84, 85.
3. Meta-Analysis of scRNA-Seq Data
This review and meta-analysis is based on scRNA-Seq and CyTOF data from mouse aortas (figure 1). scRNA-Seq data from 9 data sets (Online Data Set) were analyzed using the latest bioinformatics integration method Harmony86. Harmony allows to simultaneously account for multiple experimental and biological batches across data from different labs. After quality control, a total of 15,288 cells were projected into a shared embedding space in which cells were assigned and corrected for dataset-specificity using fuzzy clustering. Then, all cells were visualized using the non-linear high-dimensionality deduction method, UMAP (Uniform Manifold Approximation and Projection)87. Louvain-based clustering was then used with the Seurat method88 that yielded 17 clusters (figure 2; differentially expressed genes for each of the clusters in Online table I).
Figure 1. Overview.

This analysis is based on published studies where atherosclerosis was induced in mouse models using genetic knockouts Ldlr−/− 91, 96, Apoe−/− 5, diet (chow, western diet, high fat diet, refs) or AAV-PCSK9 induced lipoprotein changes92, resulting in atherosclerotic aortas. In some studies, lineage tracking (Cx3cr1-Cre)92 or chemical labeling (Bodipy)91 labeled specific cell types. All studies used enzymatic digestion with the attendant problems of cell death and loss of surface markers6. Single cells were phenotyped by RNA-Seq91, 92, 96, CyTOF106 or both5. Dimensionality reduction and clustering identified cell types and gene signatures. Matching CyTOF with scRNA-Seq data is challenging. Based on gene signatures, genetic labeling was used to visualize, image and sort some cell types to gain functional insights and deep transcriptomes129.
Figure 2. Integration of scRNA-Seq data from 9 atherosclerosis studies on mouse aortas.

scRNA-Seq data was retrieved from NCBI GEO for Harmony integration and visualized using UMAP86. 17 clusters were identified by Louvain clustering. One cluster (lilac) was dominated by non-leukocyte genes including Acta2 and one cluster (dark yellow) was dominated by proliferation genes including Top2a and Tuba1b, leaving 15 bone fide leukocyte clusters: 2 B cell clusters (red and pink), one CD8 T cell cluster (light green), 4 macrophage clusters (inflammatory, blue, IFNIC, light purple, resident, purple and Trem2, light blue), 2 CD4 T cell clusters (Th17, brown and Th2, teal), CD4+CD8+ cells (turquoise), 2 mixed monocyte/macrophage/DC clusters (orange), Xcr1+ DCs (hot pink), neutrophils (light pink), NK cells (dark green). Total n= 15,288 cells
Overall, macrophages are the most abundant cell type in atherosclerotic mouse aortas. The Louvain clustering algorithm implemented in Harmony clearly identified four subsets. We also identified clusters of two monocyte/DC subsets and one cluster of Xcr1+ cDC1 cells (Xcr1, Irf8). Xcr1 is a chemokine receptor characteristic of cDC1. One of the monocyte/DC clusters has a gene signature suggesting it may contain classical monocytes (Ccr2), the other a mixture of monocyte-derived DCs (Cd209a), and yet unidentified cells. The B cell compartment shows 2 clusters. The B1-like cluster is enriched in B1 cell genes and the B2-like cluster shares gene expression with germinal center and marginal zone B cells. The three T cell clusters include the expected CD8 T cells, CD4+ Th17-like cells and an unexpected subset of CD4+CD8+ T cells. The fourth cluster, although located right next to the T cells, mainly contains ILC2 cells based on marker genes like Klrg1, Il1rl1 and Areg and the absence of Cd3 expression. It is striking that Th1 cells, shown to be abundant in atherosclerotic lesions in mice89 and humans90 by immunostaining, do not appear to form an identifiable cluster. However, Tbx21+ T cells are found in the T cell and NK cell clusters (Online figure I). Similarly, Tregs are not easily identified in scRNA-Seq experiments, because low-expressed transcription factors like FoxP3 are often missed by scRNA-Seq. Also, FoxP3 expression can be pulsatile. Although they do not form a separate cluster, we find a few FoxP3+ CD4 T cells on the edge of the Th17-like cluster (Online figure I). Th1 Tregs37, a type of T cells with features of Tregs and Th1 cells, were not found in this analysis.
The cellular composition of mouse aortic leukocytes varied widely among the 9 data sets (figure 3). This is likely due to differences in the experimental protocols used (Online Data Set) and to biological differences between the different atherosclerosis models. Some datasets show a bias towards or directly focus on macrophages91, 92, while others are more biased towards lymphoid cells, especially Winkels et al.5. A very interesting finding is that the foam cells, identified by Bodipy staining in Kim91, are exclusively Trem2 macrophages93. Thus, based on this meta-analysis, we propose to call this cell type foamy Trem2 macrophages, a classic cell type in atherosclerosis84. About half of the foam cells as defined by Bodipy staining are non-leukocytes, probably smooth muscle cells, as expected94, 95.
Figure 3. Different abundance of aortic leukocyte subsets in 9 scRNA-Seq mouse atherosclerosis studies.
The UMAP from figure 2 was separated into the 9 different studies. Studies identified by first author5, 91, 92, 96 and abbreviated conditions (see online data set for full conditions). Cell subsets as in figure 2.
CD8 T cells are found in all data sets except in the sorted foam cells91 and in the regression study that focuses on flow-sorted CX3CR1+ CD11b+ myeloid cells92. Th17-like T cells are probably a mixture of Th17 and γδ T cells. The ILC2 cluster was only found in the Cochain96 and Winkels5 datasets. Interestingly, these cell types are already present in the healthy aorta, prior to onset of atherosclerosis, whereas the CD8 T cells are rare in healthy aortas96. CD4+CD8+ T cells are present in healthy and atherosclerotic aortas.
Among the B cells, B2-like cells are found in healthy aortas. They become less abundant after 11 or 20 weeks of high fat diet96. B1-like cells are only found in the Winkels dataset5, which contains data from Apoe−/− mice. When comparing the different models of atherosclerosis, it seems that the Apoe−/− model shows preferential accumulation of B2 cells and the appearance of a small cluster of B1 cells.
As expected14, the healthy aorta contains only resident macrophages. Also expected, macrophage frequencies increase with the duration of high fat diet in Ldlr−/− mice96 and with western diet in Apoe−/− mice5. Interestingly, macrophage diversity increases with atherosclerosis progression. Four clear subsets are seen in all datasets: Trem2+ foamy macrophages, resident macrophages, inflammatory macrophages and IFNIC macrophages.
The gene expression driving the various cell types is presented as a dot plot (figure 4). Inflammatory macrophages highly express the inflammatory chemokines Ccl4, Cxcl2, Ccl3 and Ccl2. They also express Cd14 at a higher level than the other macrophage subsets. Many express Il1b and Tnf and a few express Cxcl1. The Trem2 foamy macrophages have a gene expression profile that partially overlaps with the inflammatory macrophages. They highly express Lgals3, the gene encoding galectin-3, a known biomarker for atherosclerosis97, the tetraspanin Cd9 and Ctsd, the gene encoding cathepsin D. Some express the fatty acid binding protein Fabp5. Resident macrophages are very different from the other subsets. They express the chemokine Ccl8, the chemokine-like molecule CXCL4 (Pf4), which was previously thought to be platelet-specific98, coagulation factor XIII (F13a1) and Wfdc17. Some express Lyve1, a marker associated with tissue resident perivascular macrophages in the aorta14, 83, 99. The IFNIC macrophages have a type I interferon signature, prominently expressing Isg15, Irf7, Ifit 1 and Ifit 3. They express the chemokine Ccl12, which encodes an important ligand for CCR2. A few neutrophils are found in all data sets except the sorted foam cells, even in healthy aorta. This is somewhat surprising and unlikely to be due to blood contamination, as Cochain96 used intravascular CD45 staining to exclude blood contamination. Thus, this finding suggests that a few neutrophils may be present in the mouse aorta even under control conditions. Neutrophils share many genes with monocytes and macrophages, but characteristically express Cd7, S100a9 and S100a8.
Figure 4. Top gene signatures for 15 aortic leukocyte subsets.

The top 10 up-regulated genes in each subset compared to all other subsets are shown by Dot plot. Expression level indicated by saturation of blue (dark blue is highest expression, log2 scale where 0 is global average). Dot diameter represents percentage of cells in cluster expressing corresponding genes (largest circle is 100%).
The B cell clusters express typical B cell genes like CD79a and b, Ccr7 and Mzb1. The expression profile between B1-like cells express Tppp3, S100a6 and Cd9, whereas B2-like cells express Fcer2a and Cd23.
Two mixed monocyte/DC subsets are well represented in all datasets with atherosclerosis. As mentioned above, few monocytes are present in healthy aortas, and monocytes are not found among foam cells. The cluster denominated mixed monocytes/ DCs/Ccr2 contains cells expressing genes found in classical monocytes, i.e. Lyz1, and the chemokine receptor Ccr2, but also Fn1, encoding fibronectin, Retnla encoding Relm-α, and the chemokine Ccl6. The other monocyte cluster (mixed monocytes/DC/Cd209a) expresses aspartic peptidase Napsa1 and Cd209a, encoding DC-SIGN, which may suggests the presence of monocyte-derived DCs and cDC2. This cluster also shows expression of interferon genes Ifi30 and Ifitm1. The Xcr1+ DCs highly express the transcription factor Irf8, Cst3 encoding cystatin 3, the N-acylethanolamine acid amidase Naaa, the N-acylgalactosaminidase Naga and the phospholipase Plbd1.
NK cells express Nkgh7, Klre1, Klra7, Klrk1 and granzyme B (Gzmb). Interestingly, they also express the chemokine Xcl1, the only known ligand for dendritic cell receptor Xcr1, and the chemokine Ccl5, also known as RANTES.
ILC2 cells are negative for CD3-encoding transcripts (Cd3d, Cd3g) and express genes associated with ILC2 (Rora, Gata3, Areg, Il1rl1 encoding ST2). The Th17-like T cells express CD3-encoding genes, but are negative for Cd4, Cd8a or Cd8b1 transcripts. These cells are enriched for Il17a expression. This cluster likely contains γδT cells also expressing lymphotoxin B (Ltb), Il7r and Icos.
One cluster had an overwhelming proliferation signature. When regressing out cell cycle genes, this cell cluster primarily expressed myeloid markers such as Cd11b, Adgre1, Cd68, Trem2, and Cxcr1. Thus, they are likely proliferating macrophages. Lesional macrophages that have differentiated from monocytes proliferate to locally expand the macrophage population within plaques. Macrophage turnover becomes increasingly dependent on local proliferation as atherosclerosis progresses15. In a normal mouse aorta, resident adventitial macrophages comprise the vast majority of arterial macrophages. This population expresses Lyve1. Adventitial macrophages arise from CX3CR1+ yolk sac progenitors and fetal liver monocytes. Local proliferation maintains stable cell numbers throughout adulthood in the steady state. Upon exposure to a potent systemic inflammatory stimulus, the number of Lyve1+ adventitial macrophages falls dramatically and gradual recovery is dependent on a local proliferative response. Thus, a tightly regulated proliferative response is critical for maintaining resident arterial macrophages within the adventitial niche both during homeostasis and post-inflammation14. Whether resident aortic macrophages proliferate in atherosclerosis and contribute to this proliferating macrophage cluster is unclear.
One cluster contained Acta2+ cells that were not leukocytes, but smooth muscle cell-derived foam cells. The remaining 15 clusters distributed into 4 macrophage clusters, 2 monocyte/DC clusters, one DC cluster, one neutrophil cluster, one NK cell cluster, 2 B cell clusters, 3 T cell clusters and one innate lymphoid cell (ILC) cluster.
To improve clustering resolution, we selected myeloid cells (except neutrophils), the T/NK cells/ILCs and the B cells each separately for reclustering (figure 5). This reclustering confirmed the four macrophage subsets, resident, inflammatory, Trem2-foamy and IFNIC, and revealed a new fifth subset of macrophages. Leading differentially expressed genes (DEGs) of cells in this subset included Fn1, Clec4b1, Sept11, and Ear2. A full representation of the representative genes (with less than 30% expressed in the rest) is shown as a dot plot in Online figure II. Comparing the top DEGs of the new macrophage subset with the ImmGen database placed it most closely to peritoneal macrophages. However, these cells could also come from the pleural or pericardial cavities. Since the exact location of origin is unknown, we labeled them “cavity macrophages”. The monocytes and DCs were also better identified, revealing clusters of mature DCs, which express CCR7 and fascin-1, known markers of mature DCs100, 101. These cells were previously identified in 96. Reclustering confirmed cDC1 cells and putative pDCs. A rather large cluster contained cells with transcriptomes consistent with monocyte-derived DCs102 and cDC2s. Leading genes included CD209a, Ifitm1 and Klrd1 (Online figure II).
Figure 5. Reclustering of myeloid cells and T cells.

We selected myeloid cells (Macrophages, Mixed monocyte/macrophage/DC clusters except neutrophils, left panel), and T cells (T cell, IL-17+, NK and ILC cell clusters, right panel) from the UMAP from figure 2 for reclustering. The following myeloid clusters were retrieved: 5 macrophage clusters (inflammatory, dark green, IFNIC, light green, resident, dark pink, Trem2, light pink, and cavity, red), monocytes (blue), MoDC/cDC2 (yellow), and 3 DC clusters (Mature DC, turquoise, cDC1, yellow green, and pDC, purple). T cells clustered into naïve T cells (turquoise), CD4+Cd8+ (light pink), IL17+Cxcr6+ (light green), Treg (dark pink), CD8 (yellow), NK cells (purple) and ILC2s (dark green).
We formally compared the prevalence of these 10 myeloid cell types among the 9 data sets (Online figure III), projecting each dataset onto the same UMAP of myeloid cells and constructing stacked bar graphs (Online figure IV). The myeloid cell composition of the Cochain et al. 11 weeks HFD and the Kim et al. Ldlr−/− 12 weeks HFD datasets are almost identical. Thus, we have confidence in the myeloid cell composition of aortas harvested from HFD-fed Ldlr−/− mice, because the two studies confirm each other. The data from Winkels et al. 12 weeks HFD-fed Apoe−/− is also similar. However, the Winkels et al. studies are systematically missing foam cells, which could be caused by the harsher digestion procedure used (see Online Data Set). The Lin et al. progression and regression show no difference in myeloid cell composition. The data also show that foam cell enrichment as used in the Kim et al. study (Apoe−/− intimal foam cells) works well. Effectively, more than 80% of the foam cell-enriched preparation are actually Trem2+ foamy macrophages based on this meta-analysis. The other myeloid cell types are removed in about equal proportions. Finally, as expected, myeloid cells in the healthy mouse aorta are dominated by resident macrophages with almost no foam cells in healthy aortas (Cochain et al. Ldlr−/− mice chow). Although Winkels et al. did not investigate healthy aortas, the myeloid composition of their chow-fed Apoe−/− mouse aorta is similar, with the exception of a higher proportion of monocytes and monocyte-derived DCs.
Reclustering the T cells, NK cells, and ILCs revealed a new cluster of regulatory T cells, which express GITR (Tnfrsf18), Cd134, also known as Ox40L or Tnfrsf4, Nt5e (CD73), Ctla4 and Nrp1 (figure 5, Online figure V). In some of these cells, the hallmark transcription factor Foxp3 is also detectable (Online figure VI). Regulatory T cells are known to be important in limiting atherosclerosis34. The CD8 T cluster cells express Cd8b, Cd8a and the transcription factor Eomes. Part of the CD4+CD8+ T cells expressed the transcription factor Tox (Online figure VI). Tox is associated with exhaustion103, but is also involved in beta selection104. All of the Cd4+Cd8+ cells exclusively expressed Sox4 (Online figure VI), a transcription factor known in stem cells and early progenitors in different tissues105. Thus, this finding suggests that CD4+CD8+ lymphocytes in the aorta may be immature. The nature of the ILC2 cells was confirmed by overlaying expression of Il1rl1, which encodes the IL-32 receptor ST-2 (Online figure VI). The IL-17-expressing cells were also confirmed; many of them express the chemokine receptor CXCR6. Reclustering the B cells did not reveal any new subset with statistically significantly DEGs, and confirmed a small subset of B1 cells and a larger subset of B2 cells (data not shown).
Lymphocyte populations are similar in HFD-fed Ldlr−/− (Cochain et al. 11 weeks HFD and Kim et al. Ldlr−/− 12 weeks HFD) and Apoe−/− mice (Winkels et al. 12 weeks HFD) (Online figure VII). An expansion of CD8+ T cells is notable in all datasets in atherosclerosis. In the healthy aorta, lymphocytes dominate (figure 3), because there are so few myeloid cells2. This meta-analysis suggests that proportions of potentially immature CD4+CD8+ T cells are increased and Tregs are absent in healthy aortas (Cochain et al. Ldlr−/− mice chow).
Meta-Analysis of CyTOF Data
Two of the studies of mouse aortic leukocytes5, 106 used CyTOF to characterize the immune cell landscape of Apoe−/− mouse aortas. Each study used 35 markers, 33 of which were conjugated to monoclonal antibodies recognizing cell surface and intracellular molecules. Although the number of markers (33) is much smaller than in scRNA-Seq (~2,000), these markers are very informative, because they were picked based on 30 years of experience with flow cytometry and 15 years of experience with flow cytometry of mouse aortas2.
Cole106 used 19 myeloid markers, 5 T cell markers, 2 B cell markers and 8 others. They identified 4 macrophage subsets (table 1). All macrophages expressed CD11b, CD64 and CD68107. High levels of CD64 distinguish macrophages from other mononuclear phagocytes including monocytes Markers that differed among subsets were CD11c+CD44+ for Mac 1, CD11clow CD44+ CCR2+ CD206med CD169− for Mac 2, CD11c− CD44− CD206+ CD169+ CD209b− for Mac 3 and CD11c− CD44− CD206+ CD169+ CD209b+ for Mac 4.
Table 1. Identification of immune cells from atherosclerotic mouse aortas by CyTOF.
Data compiled from Winkels et al. and Cole et al.
| Author | Winkels et al. | Cole et al. |
|---|---|---|
| Macrophages (CD11b+) |
• Cl. 14 (CD11cmed F4/80low CD64low CD43high) • Cl. 3 (F4/80med CD64high CD103med FcεRIhigh) • Cl. 9 (F4/80high CD64med CD103low CD160high CD138high) |
• Mac 1 (CD68+ CD64+ F4/80− CD11c+ CD44+ CCR2low) • Mac 2 (CD68+ CD64+ F4/80+ CD11clow CD44+ CCR2+ CD206med CD169−) • Mac 3 (CD68+ CD64+ F4/80+ CD11c− CD44low CD206+ CD169+ CD209b−) • Mac 4 (CD68+ CD64+ F4/80+ CD11c− CD44− CD206+ CD169+ CD209b+) |
| Monocytes (CD11b+ F4/80−) |
• Cl. 24 Ly6C+ Monocytes (CD43+ Ly6Chigh MHC-IIlow CD64low CD11c−) • Cl. 6 Ly6C- Monocytes (CD43+ Ly6Clow MHC-IImed CD64low CD11clow) |
• Ly6C+ Monocytes (CD68+ Ly6C+ CCR2+ CD44+ CD64med MHC-IIlow CD11c−) • Ly6C- Monocytes (CD68+ Ly6C− CCR2− CD44+ CD64low MHC-II− CD11c+) |
| Granulocytes (CD11b+) |
• Cl.11 Neutrophils (Ly6C+ Ly6G+) • Cl.28 Eosinophils (Ly6C− SiglecFhigh F4/80high) |
• Neutrophils (Ly6C+ Ly6G/C+) • Eosinophils (Ly6C− SiglecF+ F4/80+) |
| DCs (CD11c+) |
• Cl. 16 (CD11bmed MHC-IImed CD117low CD4low CD103high) • Cl. 22 (CD11bmed MHC-IIhigh CD117high CD4med CD64low) • Cl. 10 (CD11blow MHC-IImed CD117med CD4neg) |
• cDC1 (CD68+ MHC-II+ CD103+ CD11b− CD24+ XCR1high CD172a−) • cDC2 (CD68+ MHC-II+ CD103− CD11b+ CD24low XCR1+ CD172a+) • pDC (CD68+ MHC-II+ SiglecH+ B220med CD4med) |
| B cells (CD19+) |
• Cl. 7 (B220high MHC-IImed CD117med IgMhigh CD43−) • Cl. 4 (B220high MHC-IIhigh CD117high IgMmed CD43med) • Cl. 1 (B220med MHC-IImed CD117− IgMlow CD43−) • Cl. 13 (B220low MHC-IImed CD117low CD11bhigh CD43high) |
• B220+ MHC-II+ • B220− MHC-II+ |
| T cells | • Cl. 15 (CD4high CD5med Ly6Chigh FR4high) • Cl. 17 (CD4high CD5high Ly6C− FR4high) • Cl. 25 (CD4low CD5low CD8high FR4−) • Cl. 21 (CD8high CD5med Ly6Chigh FR4high) • Cl. 23 (CD8high CD5med Ly6C− FR4high) • Cl. 20 (TCRβlow IgMlow CD11clow) • Cl. 27 (CD4low CD25high CD43high) • Cl. 19 (TCRγδhigh CD4low CD25high CD43high) |
• CD3ε+ TCRβ+ CD4+ Ly6C+ • CD3ε+ TCRβ+ CD4+ Ly6C− • CD3ε+ TCRβ+ CD8+ Ly6C+ • CD3ε+ TCRβ+ CD4+ Ly6C− • CD3ε+ TCRγδ+ |
| ILCs + NK cells | • Cl. 26 (NKp46high NK1.1high CD11bmed CD43high) |
• NKp46+ NK1.1+ CD11b+ CD11c+ CD90.2+ • CD3ε− TCRβ− CD90.2+ IL7Rα+ |
Three of these four subsets were also identified in Winkels et al5, who used 7 myeloid, 7 T cell, 7 B cell and 10 other markers. Again, all macrophages expressed CD11b and CD64; CD68 was not in the panel of Winkels et al. CD68 in Cole et al. was expressed by all clusters of monocytes, macrophages and dendritic cells, including pDCs106. Winkels’ Mac cluster 14 was CD11cmed and CD43+ and likely corresponds to Mac 1 in Cole106. Winkels’ Mac cluster 3 was CD11c− and expressed CD103 and FcεR1. It might correspond to Cole’s Mac 3 or Mac 4, which are CD11c− CD206+ CD169+ CD209b±. Winkels’ Mac cluster 9 was also CD11c- but expressed CD160 and CD138. Cole et. al.106 identified a shift in macrophage populations induced by Western diet, with a reduction in resident-like macrophages (CD11c− CD206+ CD169+) and an increase in the CD11c+ CD44+ population.
Both Cole106 and Winkels5 found the two known subsets of mouse monocytes, classical and non-classical. Both subsets express CD11b and CD68. Ly6C+ classical monocytes expressed CD68, CCR2, CD44, CD43 but not MHC-II or CD11c (table 1). Ly6C− nonclassical monocytes expressed CD68, CD43, CD44 and some MHC-II and CD11c. It is interesting that the CyTOF studies both identify classical and non-classical monocytes in atherosclerotic mouse aortas, but the scRNA-Seq data (figures 2 and 5) do not yield such clear signatures. This suggests that the monocytes may rapidly change their transcriptome upon entering the arterial wall.
Both Winkels and Cole found cDC1 cells, called cluster 16 in Winkels et al.5. They expressed CD68, CD11c, MHC-II, CD103 and XCR1, but little CD11b. cDC2 (cluster 22 in Winkels) also expressed CD68, CD11c and MHC-II, but not CD103. Instead, they expressed CD11b, CD41 and CD172a. Winkels found a third DC subset (cluster 10) that differed from cluster 22 by lower expression of CD11b and absence of CD41, while Cole identified pDCs defined by expression of B220 and SiglecH.
CyTOF identified two granulocyte populations, neutrophils (CD11b+Ly6C+Ly6G+) and eosinophils (CD11b+ SiglecF+ F4/80+). B lymphocytes (CD19+) were clustered into 4 clusters in Winkels et al.5 based on the expression of B220, CD117, IgM and CD43, and 2 clusters (B220+ and B220−) in Cole et al.106.
The two studies showed 5 common T lymphocyte populations including CD4+ Ly6C−, CD4+ Ly6C+, CD8+ Ly6C−, CD8+ Ly6C+ and TCRβ− TCRγδ+ cells5, 106. Additionally, the Winkels panel identified CD4low CD5low CD8high FR4− and TCRβlow IgMlow CD11clow T cell clusters5. NK cells that express NKp64 and NK1.1 were identified in both studies5, 106 and innate lymphoid cells defined as CD3ε− TCRβ− CD90.2+ IL7Rα+ were detected in Cole et al.’s study106.
Overall, the CyTOF studies, albeit more limited in scope when compared to the scRNASeq date, offer a solid steer on an extensive panel of antibodies that are useful to disambiguate the leukocyte populations within the atherosclerotic aorta (e.g. macrophages from dendritic cells), leading the way towards more effective and multi-analyte lesion phenotyping. Further integration of CyTOF with scRNASeq-led markers will further refine our capture of other subsets that emerge from the scRNASeq, including Trem2 foamy macrophages.
Macrophage Subsets in Atherosclerosis
As revealed by our meta-analysis, five subsets of macrophages, namely resident-like macrophages, foamy Trem2 macrophages, inflammatory macrophages, IFNIC macrophages and cavity macrophages are distinguishable in atherosclerosis.
Resident-like macrophages expressed genes previously associated with resident aortic macrophages, such as Lyve1 and Mrc114, 83. These marker genes have been described in yolk sac-derived and embryonic precursor-derived tissue resident macrophages in the adventitia of the aorta and other organs. GSEA analysis of the transcriptome of resident aortic macrophages, obtained by Affimetryx14, and our integrated scRNA-seq data set indeed showed that vascular macrophages of healthy mouse arteries were most similar to the resident-like macrophages found in healthy and atherosclerotic mouse aortas96. Resident-like macrophages are not found in the normal arterial intima, but it is possible that these cells may also be located in the atherosclerotic plaque.
Within the population of resident-like macrophages, Cochain et al. identified a subset of macrophages that showed higher expression of Ccr2 (but lower expression of Lyve1)96, suggesting that atherosclerotic aortas may contain monocyte-derived macrophages that adopt a phenotype similar to bone fide resident aortic macrophages, which self-renew independently of CCR2-dependent monocyte recruitment14.
Among the genes expressed in this cell cluster (figure 4), resident-like macrophages also showed enrichment in Pf4. Pf4 was originally thought to be specific for platelets and megakaryocytes. Pf4 expression was proposed to reflect platelet/macrophage conjugates. However, several studies in reporter mice driven by Pf4-Cre have demonstrated widespread macrophage expression of this gene, notably in peritoneal macrophages108 and (peri)vascular macrophages109. Thus, Pf4 can no longer be considered platelet-specific. If Pf4-Cre mice are used to drive a gene modification, the intended gene modification will occur in megakaryocytes, platelets and resident macrophages of the peritoneum and artery wall110.
The foamy Trem2 macrophage population predominantly found in atherosclerotic aortas and almost absent in healthy aortas was identified by Cochain et al96. This macrophage population displays high expression of Mmp12, Mmp14, Itgax (CD11c) and markers of lipid loading (Abcg1, Trem2, Fabp4), lysosomal cathepsins (Ctsd, Ctsl) Cd9, and Spp1 (osteopontin). Establishing a flow cytometry-based method of lipid staining of macrophages, Kim et al. detected lipid-laden foam cells in the intima of atherosclerotic aortas91. Cross-comparison of data from Kim and Cochain et al. indicated that foamy macrophages and Trem2hi macrophages are identical93, 111. Gene ontology or KEGG pathway analysis assigned foamy Trem2 macrophages putative functions in lipid metabolism, cholesterol efflux and lysosome function91, 96. Foamy Trem2 macrophages had low inflammatory gene expression, in line with prior findings showing that foam cells are characterized by low expression of inflammatory-response genes112. This resolves an important controversy: atherosclerotic arteries show a pro-inflammatory milieu113, 114, yet foam cells are not pro-inflammatory112. With the improved resolution of macrophage subsets provided by scRNA-Seq and CyTOF, it is now clear that the Trem2 macrophages96 are the foam cells91, but do not supply the inflammatory cytokines and chemokines so characteristic of atherosclerotic arteries.
Mononuclear phagocytes sharing transcriptomic features with aortic foamy Trem2 macrophages have been observed across various disease contexts in mice. In particular, this signature is associated with disease-associated microglia in neurodegeneration115, 116, and demyelinating disease117, non-alcoholic hepatosteatosis- and fibrosis-associated macrophages in the liver118, 119, and lipid-associated macrophages in obese adipose tissue120. How these cells acquire this specific transcriptomic signature remains to be fully determined. Recent evidence implicates TREM2 in downstream control of cholesterol metabolism in phagocytes115–117, 120, and suggests a TREM2-ApoE pathway121. Insights from other diseased tissues suggest that macrophage ontogeny may have little impact on acquisition of the foamy Trem2 signature. Both yolk sac-derived microglia115 and monocyte-derived macrophages in adipose tissue120 can acquire the foamy Trem2 macrophage state. Importantly, results so far indicate that ApoE deficiency does not affect acquisition of the foamy Trem2 macrophage transcriptional signature in atherosclerosis (Cochain et al.96 and the meta-analysis here).
Inflammatory macrophages96 primarily evidenced in atherosclerotic aortas by scRNA-seq, show enrichment in inflammatory transcripts, including the chemokines Cxcl1, Cxcl2, Ccl2, Ccl3, Ccl4, and the inflammatory cytokines Il1b, and Tnf. Inflammatory macrophages, also designated as chemokinehigh macrophages92 or non-foamy macrophages91, show a strong proinflammatory gene profile. They are not foam cells and express CCR2, suggesting they are likely derived from blood monocytes. How Trem2 macrophages and inflammatory-macrophages are interrelated is currently unclear. An algorithm to predict differentiation pathways122 from scRNA-seq data suggested that differentiation pathways into these macrophage subsets may be distinct and independent from each other114
Interferon-inducible macrophages (IFNIC) form a small cluster with a signature characteristic of a type 1 interferon response. IFNIC are enriched for numerous interferon-inducible genes including Ifit3, Irf7 and Isg15. IFNIC were found in the two studies with large numbers of macrophages91, 92, but not by Cochain et al.96 or Winkels et al.5 Identification of the IFNIC cluster was likely favored by the large number of macrophages sequenced in91, 92. After the meta-analysis (figure 3), small numbers of IFNIC macrophages became visible in the Winkels5 and Cochain96 studies. IFNIC macrophages are reminiscent of macrophages described in the ischemic heart by King et al.123. IFNIC may originate from remote type I IFN-mediated priming of monocytes and their progenitors in the bone marrow after tissue injury124. However, macrophages with a similar gene expression signature have been found at steady state in the heart125. Thus, it is not clear whether IFNIC macrophages serve a homeostatic role or whether they are strictly disease-induced. Given the pro-atherogenic role of type I IFN signaling126, this cell cluster could be relevant to disease progression.
Cavity macrophages represent a small cell cluster expressing Cd226, Itgax (CD11c), Ccr2, Retnla and MHCII encoding genes, thus showing similarities to previously identified monocyte-derived CD226+CD11c+MHCII+ small peritoneal macrophages that also populate other serous cavities (pleura)127. The origin and function of these macrophages, never observed in the aorta previously, is unclear.
These five aortic macrophage populations can be observed across studies and in three distinct models of atherosclerosis in mice: Ldlr−/− mice, Apoe−/− mice5, 91, 96, and in progressing and regressing atherosclerotic lesions of C57Bl/6 mice treated with PCSK9-AAV92. Thus, the present meta-analysis resolves the number of macrophage subsets in atherosclerotic arteries.
Relationship between Macrophage Subsets Identified by scRNA-Seq and CyTOF
How the 5 macrophage subsets identified by scRNA-Seq correspond to the 4 subsets identified by CyTOF is not completely clear. Future work with CITE-Seq where the cell surface phenotype and the transcriptome is available for the same cells7 may provide a definitive answer. However, at this point, the evidence suggests that the resident-like, foamy Trem2 and inflammatory macrophage populations may correspond to the main macrophage populations identified by mass cytometry. The largest macrophage cluster displayed a CD11b+ CD64+ CD206+ CD169+ cell surface phenotype, and could further be discriminated by expression of the C-type lectin receptor CD209b (SIGNR1) into CD209b− and CD209b+ cell clusters106. A large majority of these CD206+ macrophages also expressed other resident macrophage markers such as Lyve1 and Tim4 (Monaco et al., unpublished observation). Thus, CD11b+ CD64+ CD206+ CD169+ macrophages, encompassing Mac 3 and Mac4 in Cole et al.106 likely correspond to the resident-like macrophages identified by scRNA-seq.
Another cluster of vascular macrophages expressed CD11c, CD44, CD11b and CD64 on the cell surface but lacked resident markers such as CD206 and CD169106. MHCII expression appeared to subdivide this subset further into two clusters, however, it is yet to be determined whether these two are functionally or ontogenetically distinct. Given high expression of CD11c in foamy intimal macrophages91, this cell cluster may correspond to foamy Trem2 macrophages. Mass cytometry also identified a small macrophage subset expressing monocyte chemokine receptor CCR2 and low/intermediate levels of CD206106, which may map to inflammatory macrophages.
Macrophage and Smooth Muscle Foam Cells
It has been postulated that vascular smooth muscle cells can transdifferentiate to macrophage-like cells in murine and human atherosclerotic lesions94, 95, 128. These cells acquire some macrophage markers like CD68. However, scRNA-Seq of fate-mapped SMCs demonstrated that smooth muscle cells transform into unique fibroblast-like cells, termed ‘fibromyocytes’, rather than into macrophages. Their portfolio of macrophage genes is limited8. Such ‘fibromyocytes’ are not recovered by cell isolation strategies focusing on leukocytes or cells derived from Cx3cr1+ precursors5, 92, 96. However, in a study focused on foam cells, almost half of the foam cells were of non-leukocytic origin and contained mostly smooth muscle cells (Acta2) and few endothelial cells (Pecam1)91. In our meta-analysis, intimal foam cells (figure 3, dataset “Apoe−/− intimal foam cells (Kim et al.)”) clustered with foamy Trem2 macrophages, as expected, and cells enriched in SMC genes (Acta2, Tagln). Thus, the present meta-analysis corroborates that smooth muscle cells significantly contribute to foam cells. In all data sets, single cells were recovered that clustered with these SMC-derived foam cells (figure 3), suggesting contaminating SMCs.
Comparing Macrophage Subsets Defined by scRNA-Seq, bulk RNA-Seq and Gene Chips
It is also not clear how the four macrophage subsets identified by scRNA-Seq correspond to the four macrophage subsets GFP+, YFP+, GFP+YFP+ and unlabeled in Cx3cr1-GFP+/CD11c-YFP Apoe−/− mice129. McArdle et al. re-analyzed the macrophage data from the Winkels5 study. Using genomic gating, they found 4 macrophage subsets (rather than the 3 originally reported in Winkels). One of the macrophage subsets identified by scRNA-Seq expressed Cx3cr1, one expressed Itgax, one expressed both and one expressed neither. Thus, it is possible that the GFP+ macrophages correspond to the Cx3cr1+, the YFP+ to the Itgax+, the GFP+YFP+ to the Cx3cr1+Itgax+ and the unlabeled to the Cx3cr1−Itgax−. To address this question, we formally mapped the four subsets from the McArdle study to the 4 subsets from the Cochain and Winkels studies5, 96. We compared the genes that were significantly up- or downregulated in one macrophage subset versus the three others in McArdle et al. with the genes that were significantly up- or downregulated in one macrophage subset versus the three others in the present meta-analysis (figure 6). Cx3cr1-GFP macrophages were most similar to resident macrophages as suggested by 36 genes upregulated in common (figure 6) and 10 genes downregulated in common. CD11c-YFP macrophages were most similar to Trem2 foamy macrophages suggested by 21 genes upregulated in common (figure 6) and 24 genes downregulated in common. GFP+YFP+ cells did not map clearly. They showed 11 and 12 genes downregulated in common with inflammatory and Trem2 foamy macrophages, respectively. Thus, based on the intravital 2-photon microscopy analysis in 129 the Trem2 foamy macrophages may be motile and the resident macrophages not.
Figure 6. Relationship between Cx3cr1-GFP+ and CD11c-YFP+ macrophages and the resident, IFNIC, inflammatory and Trem2 foamy macrophages from the scRNA-Seq studies.

Significantly differentially expressed (DE) genes were determined for Cx3cr1−GFP+, CD11c−YFP+, GFP+YFP+ and unlabeled macrophages against the other 3 subsets. The same was done for resident, IFNIC, inflammatory and Trem2 foamy macrophages. The DE gene lists were intersected. Only the genes that were upregulated in common are shown.
Monocytes in Atherosclerosis
Monocyte recruitment intensifies in the setting of vascular inflammation. Parabiosis studies have shown that recruited monocytes persist within the tissue or become tissue macrophages in early lesions15, 130, 131. This is supported by observing the integrated data from Ldlr−/− and Apoe−/− mice under normo- and hypercholesterolemic conditions. This meta-analysis revealed a distinct monocyte cell cluster (Ly6c2). Monocytes have been characterized in the bone marrow and blood using RNA sequencing of sorted monocyte subsets. These studies revealed the enrichment of several genes including Lgals3, Mmp8, Ccr2, Ly6c2, and Cebpd in classical Ly6C+ monocytes. Nonclassical Ly6Cint/Ly6C− monocytes expressed elevated levels of Nr4a1, Cebpb, Mef2a, Pparg, Cd209a, and Itgal transcripts132. The monocyte population in the atherosclerotic aorta does follow a classical Ly6c+ (Ly6c2a+Ccr2+) or non-classical Ly6c− (Ccr2−Nr4a1hiCx3cr1hi) transcriptomic profile as defined by Thomas and colleagues133 and Mildner and colleagues132. Thus, aortic monocytes possess characteristics of both classical and non-classical monocytes. It remains to be determined how soon and how completely they differentiate into macrophages or monocyte-derived DCs. Monocyte differentiation programs may initiate soon upon entry to the atherosclerotic lesion or adventitia. However, in a model of skin inflammation, monocytes have been shown to retain transcriptomes similar to blood monocytes during their migration to draining lymph nodes134. Indeed, the meta-analysis conducted on scRNA-Seq data from atherosclerotic aortas shows gene signatures compatible with classical and non-classical monocytes.
The present meta-analysis shows that the vast majority of monocytes were detected in aortas of WD or HFD-fed mice. Furthermore, when comparing cell populations from CD-fed Ldlr−/− and Apoe−/− mice, the small number of monocytes contributed from “healthy” mice are from the Apoe−/− contributed cells. This is likely due to the 2-fold elevated circulating plasma cholesterol in Apoe−/− mice that occurs under basal conditions, which is known to induce spontaneous lesion formation in the absence of diet-induced hypercholesterolemia135. This is in keeping with previous functional studies reporting that the recruitment of classical monocytes to atherosclerotic lesions begins to increase approximately 2 weeks after the initiation of diet-induced hypercholesterolemia136 and that monocyte recruitment to the ascending aorta is elevated in 10 week-old Apoe−/− mice77. It remains to be fully understood why there is a delay between the recruitment of monocytes to nascent lesions (immediately) and the detection of macrophage foam cells (detectable within a few days of initiating hypercholesterolemia). Although recruitment of monocyte persists in advanced lesions77, 130, 131, parabiosis experiments showed that recruited monocytes contribute minimally to lesion macrophages15.
Hypercholesterolemic mice also display monocytosis in the bone marrow131. Hypercholesterolemia in mice expands monopoiesis, resulting in increased numbers of Ly6C+ monocytes in blood137. High levels of hypercholesterolemia in mice induce the formation of foamy monocytes138. The cytoplasmic lipid droplets result in a high side scatter when analyzed by flow cytometry. Monocyte lipid uptake is associated with upregulated expression of CD11c, chemokine receptors, and activation of α4 integrin (CD49d), which mediates adhesion to vascular cell adhesion molecule-180, 139. Studies designed to detect monocyte fate-differentiation during atherosclerotic lesion development will require the utilization of peripheral monocyte-inducible systems (driven by Ccr2, Ms4a3140 or Cxcr4141) with periodic retrieval to map differentiation programs from these cells. These approaches will answer long-standing questions regarding kinetics of fate-specification and heterogeneity of differentiation programs of monocytes entering inflamed lesions.
B Cells in Atherosclerosis
Although some B cells are found, especially in the Apoe−/− data set5 (figure 3) but also in the healthy aorta96 (figure 3), single cell analyses of atherosclerotic plaques show that B cells are not a predominant cell type in atherosclerotic aortas5, 7, 68. Resting B cells do not produce antibodies, but can differentiate to antibody-producing plasma cells in supportive niches of the spleen and bone marrow. Hence, it is not surprising that antibody production in the artery wall itself is minimal68. However, antibodies can be produced in aortic tertiary lymphoid organs (ATLO)142, 143 located in the adventitia adjacent to atherosclerotic plaques and in perivascular adipose tissue (PVAT) of aged mice. In 1 year-old mice, B cells are abundant in ATLOs5, 68 and may be more differentiated. Earlier studies2, 144 suggesting a greater abundance of B cells in atherosclerotic vessels likely included the adventitial and PVAT compartments. Flow cytometry68, CyTOF and RNAseq5 confirm the presence of both B1 and B2-like cells in atherosclerotic vessels (figure 2). Although the exact location is not known, many B cells may reside in the adventitia and PVAT. These findings underscore the need to further study adventitia and PVAT as vascular compartments important in immune regulation of atherosclerosis. Moreover, future characterization of the unique BCR sequences associated with different B cell subsets and functions will provide important insights into the specific antigens that are recognized and enhance our understanding of B cell immunity in the context of atherogenesis.
Neutrophils in Atherosclerosis
While neutrophils are consistently found in atherosclerotic lesions and aortic tissue when using antibody-based detection methods, some studies employing scRNAseq of both human and mouse atherosclerotic tissue have failed to present a neutrophil cluster5, 7. This discrepancy is likely explained by the very low mRNA content of neutrophils when compared to other leukocyte subsets. In addition, neutrophils are rich in easily releasable ribonucleases that rapidly and potently degrade endogenous RNA145. Regardless of such intrinsic obstacles, neutrophils have been detected in healthy and atherosclerotic arteries by scRNAseq96, and in this meta-analysis (figure 2). In atherosclerosis, these neutrophils have recently been proposed to segregate into Siglecfhi and Siglecflow neutrophil subsets, similar to the neutrophil subpopulations infiltrating the heart after myocardial infarction146.
T cells in Atherosclerosis
Four studies employed single cell RNA-sequencing of flow-sorted CD45+ leukocytes in murine and human atherosclerosis5, 7, 96, 147. In these studies, T cells were identified as cells expressing the mRNA coding for CD3d or CD3e (Cd3d, Cd3e). Coding genes for the T cell receptor (TCR) as frequently used in flow cytometry are less reliably found in scRNAseq data sets. In these studies, T cells were present at all stages of atherosclerosis development. Depending on models, diets and time points, Cd3e+ cells accounted for ~46 to 65% of leukocytes enzymatically released from mouse aortas. The presence of CD3d/e+ cells in different layers of atherosclerotic arteries has been inferred from a genetic deconvolution strategy of microdissected tissues148 with highest absolute cell numbers in atherosclerotic lesions and the highest fraction among all leukocytes in the adventitial layer of established atherosclerosis. In aged mice, the adventitia may contain arterial tertiary lymphoid organs (ATLOs)5, 148, 149. Direct scRNAseq of adventitial leukocytes confirmed the presence of T cells in WT and Apoe−/− mice in the adventitia of aortas from 12-week-old mice on a chow diet147. Notably, scRNAseq identified T cells even in healthy arteries from Apoe−/− mice5. Fractions of Cd3d/e+ cells among all lesional leukocytes seem to be relatively higher in Apoe−/− and Ldlr−/− mice fed with a chow diet (61 and 54%, respectively), while the fraction of monocytes and macrophages increases during WD and HFD feeding in Apoe−/− and Ldlr−/− mice. This suggests that in the setting of aggravated atherosclerosis, the contribution of myeloid cells increases relatively to T cells. In humans, CD4+ and CD8+ T cells account for ~65% of all leukocytes in carotid endarteriectomy specimens as measured by CyTOF7. These results are at odds with the predominance of macrophages and the low percentage of T cells (5 to 20%) in immunohistochemistry of atherosclerotic plaques1, 150, suggesting that enzymatic isolation, which is required before flow cytometry and scRNAseq, may overestimate relative T cell content due to a loss of fragile myeloid cells including macrophages during tissue digestion. Genetic deconvolution of human carotid plaque gene expression data sets151 suggests that T cells may account for about 1/8 of all leukocytes, while macrophages dominate in atherosclerotic plaques.
The role of T cells in atherosclerosis was recently reviewed152. As introduced above, phenotypes of T cells are traditionally attributed to lineages that are defined by the expression of specific transcription factors and cytokines153. In CD4 T cells, the expression of adhesion and chemokine receptors correlates with TH-lineage assignment and may be used as surrogates of TH phenotypes154. One technical limitation of scRNAseq, in particular of commercial drop-sequencing approaches, is the only incomplete coverage of transcription factor expression at a single cell level. Transcription factors are not highly expressed. Thus, scRNA-seq may reveal a transcription factor-coding mRNA or not, depending whether transcripts were present at the time of sampling155. In addition, expression of TH-specific cytokines at the mRNA and protein levels may only be detectable after cell stimulation, e.g. by PMA and ionomycin, for several hours. The latter considerations make scRNAseq a less reliable tool for detecting expression of TH-defining transcription factors or cytokines that would identify TH-lineage commitment. Dimensionality reduction tools may overcome this limitation by clustering non-transcription factor and non-cytokine coding TH-specific genes.
So far, T cell heterogeneity in atherosclerosis has only been characterized in single cell suspensions of aortic leukocytes. This provides sufficient discrimination between major principal leukocyte lineages, e.g. T- vs myeloid cells, but underestimates heterogeneity and differentiating genes within principal lineages. For example, one macrophage cluster in the study of Winkels et al.5 was re-clustered and revealed three distinct subsets129. Only one study focusing on Treg plasticity has employed scRNAseq of flow sorted CD4+ T cells37. In the available studies, several distinct T cell clusters were identified: Winkels et al. detected 5 T cell populations in Apoe−/− mice5, Cochain et al. 4 in Ldlr−/− mice96, and Gu et al. 3 populations in adventitial cell preparations from Apoe−/− and Ldlr−/− mice147.
In the atherosclerotic plaque, one of the T cell clusters across studies represents CD8+ (Cd8b1+) cytotoxic T cells5, 96. Differentially expressed genes in the CD8 T cell cluster included Nkg7 (Natural Killer Cell Granule Protein 7), Ms4a4b, the CD20 homologue in T cells, Ccl5, and Gzmk (Granzyme K). Notably, this cluster was also present in healthy arteries of 8-week-old Apoe−/− mice, indicating the existence of tissue-resident CD8+ T cells. Another cluster of potentially immature T cells expresses both CD4 and CD8 (previously termed ‘mixed’5, 96).
The most discrete cluster among non-CD8+ T cells identified in two independent studies expresses the chemokine receptor Cxcr6 and the transcription factor Rora+ (ROR-α). These cells accounts for ~9 % of cells (Ldlr−/−) and ~15 % (Apoe−/−) of non-CD8 T cells, respectively. CXCR6 is a chemokine receptor that is expressed by CD4+ T cells, Natural Killer T (NKT) cells and γδ T cells. Global deficiency of CXCR6 reduced atherosclerosis and CD4+ T cell accumulation156, particularly of the pro-inflammatory IL-17A-producing CD4+ T cell subset157, consistent with the concomitant expression of Rora in this subset. In this cluster of Th17-like cells, Cd4 was not detected, arguing that γδ T cells and not Th17 cells dominate.
Our meta-analysis identified another cell cluster displaying an ILC2 gene signature5. This cluster was mostly negative for Cd3 and Cd4, but showed enrichment in Areg encoding amphiregulin158 and Il1rl1 (encoding the IL-33 receptior ST2), consistent with type 2 innate lymphoid cells (ILC2s) that require RORA and GATA3 for their development159, 160. This cell cluster may also contain a few bona fide Th2 cells. Further analysis of larger cell numbers combined with CITE-seq detection of surface markers and 5’-seq of T cell receptor encoding transcripts will likely help resolve the full spectrum of T cell heterogeneity in atherosclerotic vessels.
CyTOF and scRNA-Seq data inherently do not contain spatial information. Thus, it remains unknown in which regions of the atherosclerotic plaque T cells accumulate. Immunohistochemistry has indicated that T cells are mostly found in the fibrous cap regions of the plaque1. Spatial transcriptomics could precisely address this question by combining single cell gene expression and locations. No such studies are available in mouse or human atherosclerosis.
Association Between Clinical Outcome and Plaque T cell Phenotypes
A recent study describes the T cell landscape of carotid artery endarteriectomy specimens from patients with symptomatic (recent stroke or transient ischemic attack) or asymptomatic disease7. Single-cell proteomic and transcriptomic analyses revealed a distinct distribution of leukocytes between blood and atherosclerotic plaques with a population of CD4+ and CD8+ cells being the most abundant in the plaques. CyTOF analysis identified 13 clusters of aortic CD4+ T cells that include central memory, effector memory, terminally differentiated effector memory, and regulatory T cells. One of the most striking phenotypic features of plaque-derived T cells is elevated expression of the activation marker CD69, the chemokine receptor CCR5, and PD-1, a negative regulator of T cell activation and a marker of T cell exhaustion, within CD4+ T cells. These data further support the idea that continuous and repeated activation of T cells in the aorta results in dysfunctional T cells that correlate with inflammation. Based on additional CD4+ subsets discovered by CyTOF and scRNA-Seq, it will be important to elucidate transcriptional regulation of TH cell subsets and to identify molecules that drive phenotypes and functions of Th cells in atherosclerosis. In a genetic deconvolution strategy, a gene signature of a scRNAseq-derived T cell cluster from mouse plaques negatively correlated with the clinical outcome of carotid stenosis, corroborating the concept that T cell phenotypes may predict plaque outcomes161.
Limitations of this study
scRNA-Seq preferentially yields highly expressed genes, because genes with low expression or periodic expression have many dropouts and thus are less likely to be significantly different between cell types. This is particularly obvious for transcription factors. One way around this issue is to define gene signatures of abundantly expressed genes that correlate with the low-expressed genes of interest.
Although a total of over 15,000 cells were analyzed in this meta-analysis, this number may still be too low to truly capture all cell subsets and their intermediate states. Rapidly evolving scRNA-Seq technology makes the analysis of larger numbers of cells feasible, approaching the numbers of cells analyzed by FACS or CyTOF.
The cellular stress response during enzymatic processing can induce gene expression artifacts6. Expression of immediate early genes induced during enzymatic digestion could induce artifactual clustering of macrophages. This is of particular concern, as many of these genes encode transcripts involved in immune responses, such as heat shock proteins (Hspa1b) that have been used to identify some clusters in atherosclerosis92. New protocols e.g. employing cold active protease digestion coupled with inhibition of new RNA synthesis during cell processing may help avoid such biases in future studies6.
mRNA levels for cell surface markers are poorly correlated with protein expression on the cell surface. This is because cell surface proteins must undergo glycosylation, cleavage of the signal peptide, vesicular trafficking and sometimes enzymatic modifications to appear at the cell surface. Experience teaches that it is useful to measure the cell surface phenotype in addition to single cell transcriptomes. Although 35 CyTOF markers provide a fine-grained picture of leukocyte heterogeneity in atherosclerosis5, it remains to be determined how the CyTOF clusters relate to transcriptional cell states and unique cellular functions. CITE-Seq allows linking cell surface phenotype to transcriptomes by using oligonucleotide-tagged antibodies162, combining scRNA-Seq with quantitative measurement of 50-200 cell surface markers, which will help to further resolve cluster phenotypes. There is no CITE-Seq dataset available for mouse aortas.
Enzymatic tissue digestion methods may lead to enrichment or loss of specific cell types, which may explain the high variability in aortic immune cell composition observed across the different studies. Macrophages are likely much more sensitive to isolation and sorting procedures than T cells and thus may be lost preferentially. In particular, it is unclear if large, lipid-laden foam cells that are embedded deep within lesions can be efficiently isolated and sorted. The overall low proportion of foamy Trem2 macrophages enumerated in scRNA-seq analysis may thus underestimate the contribution of this cell subset to lesion cellularity. Alternative tissue preparation techniques6 may overcome this technical limitation.
Because of differences in cell isolation procedures, different animal models and experimental variations, a direct comparison of cell types in healthy and atherosclerotic arteries is still challenging. Additional studies directly comparing cell subset distributions during atherosclerotic lesion formation, e.g. using cell hashing techniques at multiple time points of disease progression are warranted.
scRNA-Seq and CyTOF retain no information on cell position within the vessel wall or relative to other immune cells. Spatial transcriptomics methods163, 164 are on the horizon, but we currently still lack information on spatial gene expression, including the exact localization of immune cells in the plaque versus the adventitia.
scRNA-Seq is a discovery tool. As such, it is great at identifying new cell types. For example, scRNA-Seq provided clarity with respect to the macrophage types in atherosclerotic mouse aortas. Once the phenotype of the cells of interest is known, with the cell surface phenotype defined by CITE-Seq, flow cytometry-based cell sorting can be used to sort homogeneous cell populations, extract RNA and perform bulk RNA-Seq. This yields much better, deeper transcriptomes than scRNA-Seq129.
Conclusions
The modern single cell methods scRNA-Seq and CyTOF consistently defined 3-5 different macrophage subsets, two monocyte subsets, 3-5 T cell subsets, 2 B cell subsets, one NK cell subset, neutrophils, eosinophils and dendritic cells in atherosclerotic mouse aortas. This approach, albeit in its infancy, has promise in terms of disambiguation of leukocyte cell populations and states within lesions giving us the tools for multi-dimensional atherosclerotic lesion phenotyping.
By integrating 9 different datasets, we identified the cell subsets found in healthy and atherosclerotic aortas at unprecedented resolution. Macrophages are the predominant cell type in atherosclerotic aortas and can be distributed into 5 different subsets. In addition, neutrophils, monocytes, moDCs, mature DCs and pDC-like cell clusters can be discriminated. Among the T cells, a cluster of CD4+CD8+ T cells exclusively expressing Sox4 was also found in healthy aortas and may represent T cells in an immature state. In addition, naive T cells were found in both healthy and atherosclerotic aortas, in line with their constitutive migration into the aorta2. Some pro-atherogenic Th1 T cells expressing Tbx21 were dispersed among T and NK cells but did not form an identifiable cell cluster. Reclustering T cells revealed a cluster of Tregs. Il17+ Cxcr6+ T cells were consistently found. Some B1-like and mostly B2-like cells were also present in both healthy and atherosclerotic aortas.
This meta- resolved several apparent controversies. First, foam cells in atherosclerotic aortas are macrophage-derived and smooth muscle cell-derived. Although these smooth muscle foam cells acquire some markers including CD68, they do not become macrophages. Second, macrophage-derived foamy Trem2 cells do not express a pro-inflammatory gene signature. This reconciles a study by Glass et al.112 with many studies showing pro-inflammatory functions of atherosclerotic macrophages. This is resolved by the discovery of two inflammatory macrophage subsets, called IFNIC and inflammatory macrophages. Third, resident-like macrophages are found in the atherosclerotic aorta and express Pf4. Pf4 is not platelet-specific, because Pf4 is consistently expressed in these resident vascular macrophages. Reclustering aortic myeloid cells revealed a fifth macrophage subset resembling cavity macrophages. Their origin and function are unclear. We also identified proliferating macrophages, a process known to dominate macrophage accumulation in atherosclerosis15.
The discovery of an ILC2 cluster links the scRNA-Seq work to recent work by Mallat’s group165, showing that ILC2 cells are strongly atheroprotective. This could resolve the controversy about the role of Th2 cells in atherosclerosis: Some of these Th2 cells in earlier studies may have been ILC2 cells instead. Finally, the observation of SiglecF+ neutrophils in atherosclerosis146 is striking. Such neutrophils had previously been observed in cancer166, and in the infarcted mouse heart124, 146.
This meta-analysis also generated new hypotheses that can be tested in future work. The Treg switch hypothesis has received much support from the recent single cell studies, but it remains a hypothesis that needs to be tested. In particular, the switch mechanism is not known, neither is the switch direction. There is evidence for a switch to Th137, 38, Th25, Th17167 and TFH168. Other hypotheses flowing from this work include the NK cell hypothesis. In mouse models, NK cells do not accelerate or curb atherosclerosis64, but are more like bystanders. The reason for this cannot be gleaned from the NK cell transcriptomes consistently found in the scRNA-Seq studies and awaits further experimental testing.
The single cell interrogation techniques have ushered in a new wealth of information that will eventually lead to new depths of understanding. Here, we identify 19 types of leukocytes in mouse aortas: 5 types of macrophages, 5 types of T cells, 2 types of monocytes, 2 types of DCs, B1, B2, NK and ILC2 cells (figure 7). Future work will combine the power of cell surface phenotype assessment by CITE-Seq with single cell transcriptome analysis. Highly detailed analysis of the immune cell infiltrate in atherosclerotic lesions in experimental model systems is needed to translate the insights from the immunology of atherosclerosis into therapies that ultimately can benefit patients.
Figure 7. Summary of cell clusters detectable in the atherosclerotic aorta.

The leukocyte infiltrate in atherosclerotic mouse aortas as analyzed in the comprehensive meta-analysis of 9 single cell RNA-Seq studies identified 19 types of leukocytes: neutrophils, monocytes and DCs, 5 types of macrophages, 5 types of T cells, B1, B2, NK and ILC2 cells. Top expressed and characteristic genes are indicated in italics.
Supplementary Material
Acknowledgements
We thank Andreas Zirlik, University of Graz, Austria, for organizing the symposium that spawned the idea for this meta-analysis. We thank Georg Gasteiger (Institute of Systems Immunology, University of Würzburg, Würzburg, Germany) for discussion of ILC2 biology, and Florentina Porsch (Medical University of Vienna, Vienna, Austria) for help with the analyses of B cell data. Illustrations of cell types in the summary figure are based on graphics from Servier Medical Art.
Sources of Funding
A.Z. was supported by the Interdisciplinary Center for Clinical Research (IZKF [Interdisziplinäres Zentrum für Klinische Forschung]), University Hospital Würzburg (E-352 and A-384), and the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation, 374031971 - TRR 240, 324392634 - TR221, ZE827/13-1, 14-1, 15-1, and 16-1). H.W. was supported by the Deutsche Forschungsgemeinschaft (DFG, WI 4811/1-1) C.C. was supported by the Interdisciplinary Center for Clinical Research (IZKF), University Hospital Würzburg (E-353), the German Ministry of Research and Education within the Comprehensive Heart Failure Centre Würzburg (BMBF 01EO1504), the Deutsche Forschungsgemeinschaft (DFG, CO1220/1-1). H.Q.D and C.C.H. were supported by NIH P01 HL136275, NIH P01 HL055798, NIH R01 HL134236, NIH R01 CA202987. D.W. has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement No 853425). O.S. was supported by the Else-Kröner-Fresenius Stiftung, the Leducq foundation, the Vetenskapsrådet (2017-01762), and the Deutsche Forschungsgemeinschaft (DFG, SO876/11-1, SFB914 TP B8, SFB1123 TP A6 and TP B5). C.A.McN. was supported by 1R01HL 136098-01 and P01 HL136275-01. C.B. was supported by the Austrian Science Fund (FWF, SFB InThro F54). M.I.C. was supported by Canadian Institutes of Health Research (FDN-154299). E.V.G. was supported by NIH R01HL139000. K.L. was supported by NIH HL 136275, 140976, 145241, 146134 and 148094.
Disclosures
O.S. has consulted for Novo Nordisk and Astra Zeneca, has received a grant from Novo Nordisk to study the effect of circadian rhythms on atherosclerosis, and holds a patent on targeting histones in cardiovascular inflammation. K.L. has received research grants from Novo Nordisk and Kirin Pharmaceuticals. All other authors have nothing to disclose.
Nonstandard Abbreviations and Acronyms:
- AAV
adeno-associated virus
- Acta2
actin alpha 2
- ATLO
artery tertiary lymphoid organ
- Apoe
apolipoprotein E
- CC
C-C chemokine
- CXC
C-X-C chemokine
- CX3C
C-X3-C chemokine
- CD
Cluster of differentiation
- CITE-seq
cellular indexing of transcriptomes and epitopes by sequencing
- CTL
cytotoxic T lymphocytes
- CyTOF
mass cytometry
- DC
dendritic cell
- DEG
differentially expressed gene
- GFP
green fluorescent protein
- GSEA
gene set enrichment analysis
- GWAS
genome-wide association study
- HFD
high-fat diet
- IFNIC
interferon-inducible cells
- ILC
innate lymphocyte-like cells
- LDL
low density lipoprotein
- Ldlr
low density lipoprotein receptor
- MHC
major histocompatibility complex
- NK cell
natural killer cell
- NKT
natural killer T cell
- RAG
recombinase activating gene
- scRNA-seq
single cells RNA sequencing
- TCR
T cell receptor
- TFH cell
T follicular helper cell
- TH cell
T helper cell
- Treg
regulatory T cell
- TREM2
triggering receptor expressed on myeloid cells 2
- TRM
tissue resident memory
- UMAP
uniform manifold approximation and projection
- pDC
plasmacytoid dendritic cell
- PCSK9
protein convertase subtilisin/kexin type 9
- PVAT
perivascular adipose tissue
- VCAM
vascular cell adhesion molecule
- WD
Western diet
- YFP
yellow fluorescent protein
References
- 1.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] [PubMed] [Google Scholar]
- 2.Galkina E, Kadl A, Sanders J, Varughese D, Sarembock IJ, Ley K. Lymphocyte recruitment into the aortic wall before and during development of atherosclerosis is partially l-selectin dependent. J Exp Med. 2006;203:1273–1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Bonanno E, Mauriello A, Partenzi A, Anemona L, Spagnoli LG. Flow cytometry analysis of atherosclerotic plaque cells from human carotids: A validation study. Cytometry. 2000;39:158–165. [DOI] [PubMed] [Google Scholar]
- 4.Bandura DR, Baranov VI, Ornatsky OI, et al. Mass cytometry: Technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Analytical chemistry. 2009;81:6813–6822. [DOI] [PubMed] [Google Scholar]
- 5.Winkels H, Ehinger E, Vassallo M, et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell rna-sequencing and mass cytometry. Circ Res. 2018;122:1675–1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Williams JW, Winkels H, Durant C, Zaitsev K, Ghosheh Y, Ley K. Single cell rna sequencing in atherosclerosis research. Circ Res. 2020;In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fernandez DM, Rahman AH, Fernandez NF, et al. Single-cell immune landscape of human atherosclerotic plaques. Nat Med. 2019;25:1576–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wirka RC, Wagh D, Paik DT, et al. Atheroprotective roles of smooth muscle cell phenotypic modulation and the tcf21 disease gene as revealed by single-cell analysis. Nat Med. 2019;25:1280–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein e. Science. 1992;258:468–471. [DOI] [PubMed] [Google Scholar]
- 10.Plump AS, Smith JD, Hayek T, Aalto-Setala K, Walsh A, Verstuyft JG, Rubin EM, Breslow JL. Severe hypercholesterolemia and atherosclerosis in apolipoprotein e-deficient mice created by homologous recombination in es cells. Cell. 1992;71:343–353. [DOI] [PubMed] [Google Scholar]
- 11.Ishibashi S, Goldstein JL, Brown MS, Herz J, Burns DK. Massive xanthomatosis and atherosclerosis in cholesterol-fed low density lipoprotein receptor-negative mice. J Clin Invest. 1994;93:1885–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dutta R, Singh U, Li TB, Fornage M, Teng BB. Hepatic gene expression profiling reveals perturbed calcium signaling in a mouse model lacking both ldl receptor and apobec1 genes. Atherosclerosis. 2003;169:51–62. [DOI] [PubMed] [Google Scholar]
- 13.Braun A, Trigatti BL, Post MJ, Sato K, Simons M, Edelberg JM, Rosenberg RD, Schrenzel M, Krieger M. Loss of sr-bi expression leads to the early onset of occlusive atherosclerotic coronary artery disease, spontaneous myocardial infarctions, severe cardiac dysfunction, and premature death in apolipoprotein e-deficient mice. Circ Res. 2002;90:270–276. [DOI] [PubMed] [Google Scholar]
- 14.Ensan S, Li A, Besla R, et al. Self-renewing resident arterial macrophages arise from embryonic cx3cr1(+) precursors and circulating monocytes immediately after birth. Nat Immunol. 2016;17:159–168. [DOI] [PubMed] [Google Scholar]
- 15.Robbins CS, Hilgendorf I, Weber GF, et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med. 2013;19:1166–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bailey-Bucktrout SL, Martinez-Llordella M, Zhou X, Anthony B, Rosenthal W, Luche H, Fehling HJ, Bluestone JA. Self-antigen-driven activation induces instability of regulatory t cells during an inflammatory autoimmune response. Immunity. 2013;39:949–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sakaguchi S, Vignali DA, Rudensky AY, Niec RE, Waldmann H. The plasticity and stability of regulatory t cells. Nature reviews. Immunology. 2013;13:461–467. [DOI] [PubMed] [Google Scholar]
- 18.Swirski FK, Nahrendorf M. Cardioimmunology: The immune system in cardiac homeostasis and disease. Nature reviews. Immunology. 2018;18:733–744. [DOI] [PubMed] [Google Scholar]
- 19.Hofmann U, Frantz S. Role of lymphocytes in myocardial injury, healing, and remodeling after myocardial infarction. Circ Res. 2015;116:354–367. [DOI] [PubMed] [Google Scholar]
- 20.Yui MA, Rothenberg EV. Developmental gene networks: A triathlon on the course to t cell identity. Nature reviews. Immunology. 2014;14:529–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Nikolich-Zugich J, Slifka MK, Messaoudi I. The many important facets of t-cell repertoire diversity. Nature reviews. Immunology. 2004;4:123–132. [DOI] [PubMed] [Google Scholar]
- 22.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of cd4+cd25+ regulatory t cells. Nat Immunol. 2003;4:330–336. [DOI] [PubMed] [Google Scholar]
- 23.Hori S, Nomura T, Sakaguchi S. Control of regulatory t cell development by the transcription factor foxp3. Science. 2003;299:1057–1061. [PubMed] [Google Scholar]
- 24.Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady G, Wahl SM. Conversion of peripheral cd4+cd25- naive t cells to cd4+cd25+ regulatory t cells by tgf-beta induction of transcription factor foxp3. J Exp Med. 2003;198:1875–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wolf D, Ley K. Immunity and inflammation in atherosclerosis. Circ Res. 2019;124:315–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Koltsova EK, Garcia Z, Chodaczek G, et al. Dynamic t cell-apc interactions sustain chronic inflammation in atherosclerosis. J Clin Invest. 2012;122:3114–3126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Stemme S, Faber B, Holm J, Wiklund O, Witztum JL, Hansson GK. T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:3893–3897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Buono C, Binder CJ, Stavrakis G, Witztum JL, Glimcher LH, Lichtman AH. T-bet deficiency reduces atherosclerosis and alters plaque antigen-specific immune responses. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:1596–1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C. Ifn-gamma potentiates atherosclerosis in apoe knock-out mice. J Clin Invest. 1997;99:2752–2761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper t cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol. 1986;136:2348–2357. [PubMed] [Google Scholar]
- 31.Cardilo-Reis L, Gruber S, Schreier SM, et al. Interleukin-13 protects from atherosclerosis and modulates plaque composition by skewing the macrophage phenotype. EMBO Mol Med. 2012;4:1072–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.King VL, Szilvassy SJ, Daugherty A. Interleukin-4 deficiency decreases atherosclerotic lesion formation in a site-specific manner in female ldl receptor−/− mice. Arterioscler Thromb Vasc Biol. 2002;22:456–461. [DOI] [PubMed] [Google Scholar]
- 33.Li Q, Ming T, Wang Y, Ding S, Hu C, Zhang C, Cao Q, Wang Y. Increased th9 cells and il-9 levels accelerate disease progression in experimental atherosclerosis. Am J Transl Res. 2017;9:1335–1343. [PMC free article] [PubMed] [Google Scholar]
- 34.Ait-Oufella H, Salomon BL, Potteaux S, et al. Natural regulatory t cells control the development of atherosclerosis in mice. Nat Med. 2006;12:178–180. [DOI] [PubMed] [Google Scholar]
- 35.Klingenberg R, Gerdes N, Badeau RM, et al. Depletion of foxp3+ regulatory t cells promotes hypercholesterolemia and atherosclerosis. J Clin Invest. 2013;123:1323–1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.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] [PMC free article] [PubMed] [Google Scholar]
- 37.Butcher MJ, Filipowicz AR, Waseem TC, McGary CM, Crow KJ, Magilnick N, Boldin M, Lundberg PS, Galkina EV. Atherosclerosis-driven treg plasticity results in formation of a dysfunctional subset of plastic ifngamma+ th1/tregs. Circ Res. 2016;119:1190–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Li J, McArdle S, Gholami A, Kimura T, Wolf D, Gerhardt T, Miller J, Weber C, Ley K. Ccr5+t-bet+foxp3+ effector cd4 t cells drive atherosclerosis. Circ Res. 2016;118:1540–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.DuPage M, Bluestone JA. Harnessing the plasticity of cd4(+) t cells to treat immune-mediated disease. Nature reviews. Immunology. 2016;16:149–163. [DOI] [PubMed] [Google Scholar]
- 40.Eid RE, Rao DA, Zhou J, et al. Interleukin-17 and interferon-gamma are produced concomitantly by human coronary artery-infiltrating t cells and act synergistically on vascular smooth muscle cells. Circulation. 2009;119:1424–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yazdani MR, Khosropanah S, Doroudchi M. Interleukin-17 production by cd4+cd45ro+foxp3+ t cells in peripheral blood of patients with atherosclerosis. Archives of medical sciences. Atherosclerotic diseases. 2019;4:e215–e224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bisikirska B, Colgan J, Luban J, Bluestone JA, Herold KC. Tcr stimulation with modified anti-cd3 mab expands cd8+ t cell population and induces cd8+cd25+ tregs. J Clin Invest. 2005;115:2904–2913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Andersen MH, Schrama D, Thor Straten P, Becker JC. Cytotoxic t cells. The Journal of investigative dermatology. 2006;126:32–41. [DOI] [PubMed] [Google Scholar]
- 44.Cochain C, Koch M, Chaudhari SM, Busch M, Pelisek J, Boon L, Zernecke A. Cd8+ t cells regulate monopoiesis and circulating ly6c-high monocyte levels in atherosclerosis in mice. Circ Res. 2015;117:244–253. [DOI] [PubMed] [Google Scholar]
- 45.Gewaltig J, Kummer M, Koella C, Cathomas G, Biedermann BC. Requirements for cd8 t-cell migration into the human arterial wall. Human pathology. 2008;39:1756–1762. [DOI] [PubMed] [Google Scholar]
- 46.van Dijk RA, Duinisveld AJ, Schaapherder AF, et al. A change in inflammatory footprint precedes plaque instability: A systematic evaluation of cellular aspects of the adaptive immune response in human atherosclerosis. Journal of the American Heart Association. 2015;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Biessen EA, Sluimer JC. Staging lymphocyte presence in human atherosclerosis: A tale told by numbers. Journal of the American Heart Association. 2015;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kyaw T, Winship A, Tay C, et al. Cytotoxic and proinflammatory cd8+ t lymphocytes promote development of vulnerable atherosclerotic plaques in apoe-deficient mice. Circulation. 2013;127:1028–1039. [DOI] [PubMed] [Google Scholar]
- 49.Kolbus D, Ramos OH, Berg KE, Persson J, Wigren M, Bjorkbacka H, Fredrikson GN, Nilsson J. Cd8+ t cell activation predominate early immune responses to hypercholesterolemia in apoe(−)(/)(−) mice. BMC immunology. 2010;11:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Schurch CM, Riether C, Ochsenbein AF. Cytotoxic cd8+ t cells stimulate hematopoietic progenitors by promoting cytokine release from bone marrow mesenchymal stromal cells. Cell stem cell. 2014;14:460–472. [DOI] [PubMed] [Google Scholar]
- 51.Getz GS, Reardon CA. Natural killer t cells in atherosclerosis. Nat Rev Cardiol. 2017;14:304–314. [DOI] [PubMed] [Google Scholar]
- 52.van Puijvelde GHM, Kuiper J. Nkt cells in cardiovascular diseases. Eur J Pharmacol. 2017;816:47–57. [DOI] [PubMed] [Google Scholar]
- 53.Ito S, Iwaki S, Kondo R, et al. Tnf-alpha production in nkt cell hybridoma is regulated by sphingosine-1-phosphate: Implications for inflammation in atherosclerosis. Coron Artery Dis. 2014;25:311–320. [DOI] [PubMed] [Google Scholar]
- 54.Li Y, To K, Kanellakis P, et al. Cd4+ natural killer t cells potently augment aortic root atherosclerosis by perforin- and granzyme b-dependent cytotoxicity. Circ Res. 2015;116:245–254. [DOI] [PubMed] [Google Scholar]
- 55.To K, Agrotis A, Besra G, Bobik A, Toh BH. Nkt cell subsets mediate differential proatherogenic effects in apoe−/− mice. Arterioscler Thromb Vasc Biol. 2009;29:671–677. [DOI] [PubMed] [Google Scholar]
- 56.Bobryshev YV, Lord RS. Co-accumulation of dendritic cells and natural killer t cells within rupture-prone regions in human atherosclerotic plaques. J Histochem Cytochem. 2005;53:781–785. [DOI] [PubMed] [Google Scholar]
- 57.Kyriakakis E, Cavallari M, Andert J, et al. Invariant natural killer t cells: Linking inflammation and neovascularization in human atherosclerosis. Eur J Immunol. 2010;40:3268–3279. [DOI] [PubMed] [Google Scholar]
- 58.Kleindienst R, Xu Q, Willeit J, Waldenberger FR, Weimann S, Wick G. Immunology of atherosclerosis. Demonstration of heat shock protein 60 expression and t lymphocytes bearing alpha/beta or gamma/delta receptor in human atherosclerotic lesions. The American journal of pathology. 1993;142:1927–1937. [PMC free article] [PubMed] [Google Scholar]
- 59.Garcillan B, Marin AV, Jimenez-Reinoso A, et al. Gammadelta t lymphocytes in the diagnosis of human t cell receptor immunodeficiencies. Front Immunol. 2015;6:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lawand M, Dechanet-Merville J, Dieu-Nosjean MC. Key features of gamma-delta t-cell subsets in human diseases and their immunotherapeutic implications. Front Immunol. 2017;8:761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Prinz I, Silva-Santos B, Pennington DJ. Functional development of gammadelta t cells. Eur J Immunol. 2013;43:1988–1994. [DOI] [PubMed] [Google Scholar]
- 62.Ribot JC, deBarros A, Pang DJ, et al. Cd27 is a thymic determinant of the balance between interferon-gamma- and interleukin 17-producing gammadelta t cell subsets. Nat Immunol. 2009;10:427–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zaridze DG, Chevchenko VE, Levtshuk AA, Lifanova YE, Maximovitch DM. Fatty acid composition of phospholipids in erythrocyte membranes and risk of breast cancer. International journal of cancer. 1990;45:807–810. [DOI] [PubMed] [Google Scholar]
- 64.Nour-Eldine W, Joffre J, Zibara K, et al. Genetic depletion or hyperresponsiveness of natural killer cells do not affect atherosclerosis development. Circ Res. 2018;122:47–57. [DOI] [PubMed] [Google Scholar]
- 65.Winkels H, Ley K. Natural killer cells at ease: Atherosclerosis is not affected by genetic depletion or hyperactivation of natural killer cells. Circ Res. 2018;122:6–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rauch PJ, Chudnovskiy A, Robbins CS, et al. Innate response activator b cells protect against microbial sepsis. Science. 2012;335:597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Hilgendorf I, Theurl I, Gerhardt LM, et al. Innate response activator b cells aggravate atherosclerosis by stimulating t helper-1 adaptive immunity. Circulation. 2014;129:1677–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Srikakulapu P, Upadhye A, Rosenfeld SM, et al. Perivascular adipose tissue harbors atheroprotective igm-producing b cells. Frontiers in Physiology. 2017;8:719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Srikakulapu P, Hu D, Yin C, et al. Artery tertiary lymphoid organs control multilayered territorialized atherosclerosis b-cell responses in aged apoe−/− mice. Arterioscler Thromb Vasc Biol. 2016;36:1174–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Silvestre-Roig C, Braster Q, Wichapong K, et al. Externalized histone h4 orchestrates chronic inflammation by inducing lytic cell death. Nature. 2019;569:236–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Westerterp M, Gourion-Arsiquaud S, Murphy AJ, Shih A, Cremers S, Levine RL, Tall AR, Yvan-Charvet L. Regulation of hematopoietic stem and progenitor cell mobilization by cholesterol efflux pathways. Cell stem cell. 2012;11:195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Cossarizza A, Chang HD, Radbruch A, et al. Guidelines for the use of flow cytometry and cell sorting in immunological studies (second edition). Eur J Immunol. 2019;49:1457–1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in ccr2−/− mice reveals a role for chemokines in the initiation of atherosclerosis. Nature. 1998;394:894–897. [DOI] [PubMed] [Google Scholar]
- 74.Hanna RN, Carlin LM, Hubbeling HG, Nackiewicz D, Green AM, Punt JA, Geissmann F, Hedrick CC. The transcription factor nr4a1 (nur77) controls bone marrow differentiation and the survival of ly6c- monocytes. Nat Immunol. 2011;12:778–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003;19:71–82. [DOI] [PubMed] [Google Scholar]
- 76.Jongstra-Bilen J, Haidari M, Zhu SN, Chen M, Guha D, Cybulsky MI. Low-grade chronic inflammation in regions of the normal mouse arterial intima predisposed to atherosclerosis. J Exp Med. 2006;203:2073–2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Swirski FK, Pittet MJ, Kircher MF, Aikawa E, Jaffer FA, Libby P, Weissleder R. Monocyte accumulation in mouse atherogenesis is progressive and proportional to extent of disease. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:10340–10345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Carlin LM, Stamatiades EG, Auffray C, et al. Nr4a1-dependent ly6c(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell. 2013;153:362–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Quintar A, McArdle S, Wolf D, et al. Endothelial protective monocyte patrolling in large arteries intensified by western diet and atherosclerosis. Circ Res. 2017;120:1789–1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Marcovecchio PM, Thomas GD, Mikulski Z, et al. Scavenger receptor cd36 directs nonclassical monocyte patrolling along the endothelium during early atherogenesis. Arterioscler Thromb Vasc Biol. 2017;37:2043–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, Segura E, Tussiwand R, Yona S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nature reviews. Immunology. 2014;14:571–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Swiecki M, Colonna M. The multifaceted biology of plasmacytoid dendritic cells. Nature reviews. Immunology. 2015;15:471–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Lim HY, Lim SY, Tan CK, et al. Hyaluronan receptor lyve-1-expressing macrophages maintain arterial tone through hyaluronan-mediated regulation of smooth muscle cell collagen. Immunity. 2018;49:326‖341 e327. [DOI] [PubMed] [Google Scholar]
- 84.Cochain C, Zernecke A. Macrophages in vascular inflammation and atherosclerosis. Pflugers Archiv : European journal of physiology. 2017;469:485–499. [DOI] [PubMed] [Google Scholar]
- 85.Cybulsky MI, Cheong C, Robbins CS. Macrophages and dendritic cells: Partners in atherogenesis. Circ Res. 2016;118:637–652. [DOI] [PubMed] [Google Scholar]
- 86.Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with harmony. Nature methods. 2019;16:1289–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, Ginhoux F, Newell EW. Dimensionality reduction for visualizing single-cell data using umap. Nature biotechnology. 2018 [DOI] [PubMed] [Google Scholar]
- 88.Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888–1902 e1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhou X, Paulsson G, Stemme S, Hansson GK. Hypercholesterolemia is associated with a t helper (th) 1/th2 switch of the autoimmune response in atherosclerotic apo e-knockout mice. J Clin Invest. 1998;101:1717–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.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] [PubMed] [Google Scholar]
- 91.Kim K, Shim D, Lee JS, et al. Transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models. Circ Res. 2018;123:1127–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lin JD, Nishi H, Poles J, et al. Single-cell analysis of fate-mapped macrophages reveals heterogeneity, including stem-like properties, during atherosclerosis progression and regression. JCI insight. 2019;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cochain C, Saliba AE, Zernecke A. Letter by cochain et al regarding article, “transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models”. Circ Res. 2018;123:e48–e49. [DOI] [PubMed] [Google Scholar]
- 94.Feil S, Fehrenbacher B, Lukowski R, Essmann F, Schulze-Osthoff K, Schaller M, Feil R. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ Res. 2014;115:662–667. [DOI] [PubMed] [Google Scholar]
- 95.Shankman LS, Gomez D, Cherepanova OA, et al. Klf4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat Med. 2015;21:628–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Cochain C, Vafadarnejad E, Arampatzi P, Pelisek J, Winkels H, Ley K, Wolf D, Saliba AE, Zernecke A. Single-cell rna-seq reveals the transcriptional landscape and heterogeneity of aortic macrophages in murine atherosclerosis. Circ Res. 2018;122:1661–1674. [DOI] [PubMed] [Google Scholar]
- 97.Gleissner CA, Erbel C, Linden F, et al. Galectin-3 binding protein, coronary artery disease and cardiovascular mortality: Insights from the luric study. Atherosclerosis. 2017;260:121–129. [DOI] [PubMed] [Google Scholar]
- 98.Tiedt R, Schomber T, Hao-Shen H, Skoda RC. Pf4-cre transgenic mice allow the generation of lineage-restricted gene knockouts for studying megakaryocyte and platelet function in vivo. Blood. 2007;109:1503–1506. [DOI] [PubMed] [Google Scholar]
- 99.Chakarov S, Lim HY, Tan L, et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science. 2019;363. [DOI] [PubMed] [Google Scholar]
- 100.Dieu MC, Vanbervliet B, Vicari A, et al. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J Exp Med. 1998;188:373–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yamakita Y, Matsumura F, Lipscomb MW, Chou PC, Werlen G, Burkhardt JK, Yamashiro S. Fascin1 promotes cell migration of mature dendritic cells. J Immunol. 2011;186:2850–2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Menezes S, Melandri D, Anselmi G, et al. The heterogeneity of ly6c(hi) monocytes controls their differentiation into inos(+) macrophages or monocyte-derived dendritic cells. Immunity. 2016;45:1205–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Khan O, Giles JR, McDonald S, et al. Tox transcriptionally and epigenetically programs cd8(+) t cell exhaustion. Nature. 2019;571:211–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Aliahmad P, Seksenyan A, Kaye J. The many roles of tox in the immune system. Current opinion in immunology. 2012;24:173–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Busslinger M Transcriptional control of early b cell development. Annu Rev Immunol. 2004;22:55–79. [DOI] [PubMed] [Google Scholar]
- 106.Cole JE, Park I, Ahern DJ, Kassiteridi C, Danso Abeam D, Goddard ME, Green P, Maffia P, Monaco C. Immune cell census in murine atherosclerosis: Cytometry by time of flight illuminates vascular myeloid cell diversity. Cardiovasc Res. 2018;114:1360–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Tamoutounour S, Henri S, Lelouard H, et al. Cd64 distinguishes macrophages from dendritic cells in the gut and reveals the th1-inducing role of mesenteric lymph node macrophages during colitis. Eur J Immunol. 2012;42:3150–3166. [DOI] [PubMed] [Google Scholar]
- 108.Abram CL, Roberge GL, Hu Y, Lowell CA. Comparative analysis of the efficiency and specificity of myeloid-cre deleting strains using rosa-eyfp reporter mice. Journal of immunological methods. 2014;408:89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.McKinsey GL, Lizama CO, Keown-Lang AE, Niu A, Chee E, Santander N, Arnold TD. New tools for genetically targeting myeloid populations in the central nervous system. bioRxiv. 2019 [Google Scholar]
- 110.Nagy Z, Vogtle T, Geer MJ, et al. The gp1ba-cre transgenic mouse: A new model to delineate platelet and leukocyte functions. Blood. 2019;133:331–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kim K, Choi JH. Response by kim and choi to letter regarding article, “transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models”. Circ Res. 2018;123:e50. [DOI] [PubMed] [Google Scholar]
- 112.Spann NJ, Garmire LX, McDonald JG, et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell. 2012;151:138–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Libby P, Hansson GK. From focal lipid storage to systemic inflammation: Jacc review topic of the week. J Am Coll Cardiol. 2019;74:1594–1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Williams JW, Huang LH, Randolph GJ. Cytokine circuits in cardiovascular disease. Immunity. 2019;50:941–954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Keren-Shaul H, Spinrad A, Weiner A, et al. A unique microglia type associated with restricting development of alzheimer’s disease. Cell. 2017;169:1276–1290 e1217. [DOI] [PubMed] [Google Scholar]
- 116.Zhou Y, Song WM, Andhey PS, et al. Human and mouse single-nucleus transcriptomics reveal trem2-dependent and trem2-independent cellular responses in alzheimer’s disease. Nat Med. 2020;26:131–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Nugent AA, Lin K, van Lengerich B, et al. Trem2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron. 2019 [DOI] [PubMed] [Google Scholar]
- 118.Xiong X, Kuang H, Ansari S, et al. Landscape of intercellular crosstalk in healthy and nash liver revealed by single-cell secretome gene analysis. Molecular cell. 2019;75:644–660 e645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ramachandran P, Dobie R, Wilson-Kanamori JR, et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature. 2019;575:512–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Jaitin DA, Adlung L, Thaiss CA, et al. Lipid-associated macrophages control metabolic homeostasis in a trem2-dependent manner. Cell. 2019;178:686–698 e614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Krasemann S, Madore C, Cialic R, et al. The trem2-apoe pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity. 2017;47:566–581 e569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Trapnell C, Cacchiarelli D, Grimsby J, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nature biotechnology. 2014;32:381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.King KR, Aguirre AD, Ye YX, et al. Irf3 and type i interferons fuel a fatal response to myocardial infarction. Nat Med. 2017;23:1481–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Calcagno DM, Ng RP, Toomu A, et al. Type i interferon responses to ischemic injury begin in the bone marrow of mice and humans and depend on tet2, nrf2, and irf3. bioRxiv 765404 2020 [Google Scholar]
- 125.Dick SA, Macklin JA, Nejat S, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. 2019;20:29–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Chen HJ, Tas SW, de Winther MPJ. Type-i interferons in atherosclerosis. J Exp Med. 2020;217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kim KW, Williams JW, Wang YT, Ivanov S, Gilfillan S, Colonna M, Virgin HW, Gautier EL, Randolph GJ. Mhc ii+ resident peritoneal and pleural macrophages rely on irf4 for development from circulating monocytes. J Exp Med. 2016;213:1951–1959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Allahverdian S, Chehroudi AC, McManus BM, Abraham T, Francis GA. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis. Circulation. 2014;129:1551–1559. [DOI] [PubMed] [Google Scholar]
- 129.McArdle S, Buscher K, Ghosheh Y, Pramod AB, Miller J, Winkels H, Wolf D, Ley K. Migratory and dancing macrophage subsets in atherosclerotic lesions. Circ Res. 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Tacke F, Alvarez D, Kaplan TJ, et al. Monocyte subsets differentially employ ccr2, ccr5, and cx3cr1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117:185–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, Pittet MJ. Ly-6chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007;117:195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Mildner A, Schonheit J, Giladi A, et al. Genomic characterization of murine monocytes reveals c/ebpbeta transcription factor dependence of ly6c(−) cells. Immunity. 2017;46:849–862 e847. [DOI] [PubMed] [Google Scholar]
- 133.Thomas GD, Hanna RN, Vasudevan NT, et al. Deleting an nr4a1 super-enhancer subdomain ablates ly6c(low) monocytes while preserving macrophage gene function. Immunity. 2016;45:975–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Jakubzick C, Gautier EL, Gibbings SL, et al. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity. 2013;39:599–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zadelaar S, Kleemann R, Verschuren L, de Vries-Van der Weij J, van der Hoorn J, Princen HM, Kooistra T. Mouse models for atherosclerosis and pharmaceutical modifiers. Arterioscler Thromb Vasc Biol. 2007;27:1706–1721. [DOI] [PubMed] [Google Scholar]
- 136.Zhu SN, Chen M, Jongstra-Bilen J, Cybulsky MI. Gm-csf regulates intimal cell proliferation in nascent atherosclerotic lesions. J Exp Med. 2009;206:2141–2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yvan-Charvet L, Pagler T, Gautier EL, et al. Atp-binding cassette transporters and hdl suppress hematopoietic stem cell proliferation. Science. 2010;328:1689–1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Wu H, Gower RM, Wang H, et al. Functional role of cd11c+ monocytes in atherogenesis associated with hypercholesterolemia. Circulation. 2009;119:2708–2717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Gower RM, Wu H, Foster GA, Devaraj S, Jialal I, Ballantyne CM, Knowlton AA, Simon SI. Cd11c/cd18 expression is upregulated on blood monocytes during hypertriglyceridemia and enhances adhesion to vascular cell adhesion molecule-1. Arterioscler Thromb Vasc Biol. 2011;31:160–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Liu Z, Gu Y, Chakarov S, et al. Fate mapping via ms4a3-expression history traces monocyte-derived cells. Cell. 2019;178:1509–1525 e1519. [DOI] [PubMed] [Google Scholar]
- 141.Werner Y, Mass E, Ashok Kumar P, et al. Cxcr4 distinguishes hsc-derived monocytes from microglia and reveals monocyte immune responses to experimental stroke. Nature neuroscience. 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Mohanta SK, Yin C, Peng L, et al. Artery tertiary lymphoid organs contribute to innate and adaptive immune responses in advanced mouse atherosclerosis. Circ Res.114:1772–1787. [DOI] [PubMed] [Google Scholar]
- 143.Hu D, Mohanta Sarajo ÂK, Yin C, et al. Artery tertiary lymphoid organs control aorta immunity and protect against atherosclerosis via vascular smooth muscle cell lymphotoxin î² receptors. Immunity.42:1100–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Doran AC, Lipinski MJ, Oldham SN, et al. B-cell aortic homing and atheroprotection depend on id3 Circ Res. 2012;110:e1–e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cassatella MA, Ostberg NK, Tamassia N, Soehnlein O. Biological roles of neutrophil-derived granule proteins and cytokines. Trends in immunology. 2019;40:648–664. [DOI] [PubMed] [Google Scholar]
- 146.Vafadarnejad E, Rizzo G, Krampert L, et al. Time-resolved single-cell transcriptomics uncovers dynamics of cardiac neutrophil diversity in murine myocardial infarction. [DOI] [PubMed] [Google Scholar]
- 147.Gu W, Ni Z, Tan YQ, et al. Adventitial cell atlas of wt (wild type) and apoe (apolipoprotein e)-deficient mice defined by single-cell rna sequencing. Arterioscler Thromb Vasc Biol. 2019;39:1055–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Moos MP, John N, Grabner R, Nossmann S, Gunther B, Vollandt R, Funk CD, Kaiser B, Habenicht AJ. The lamina adventitia is the major site of immune cell accumulation in standard chow-fed apolipoprotein e-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25:2386–2391. [DOI] [PubMed] [Google Scholar]
- 149.Grabner R, Lotzer K, Dopping S, et al. Lymphotoxin beta receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged apoe−/− mice. J Exp Med. 2009;206:233–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Hansson GK, Holm J, Jonasson L. Detection of activated t lymphocytes in the human atherosclerotic plaque. The American journal of pathology. 1989;135:169–175. [PMC free article] [PubMed] [Google Scholar]
- 151.Folkersen L, Persson J, Ekstrand J, Agardh HE, Hansson GK, Gabrielsen A, Hedin U, Paulsson-Berne G. Prediction of ischemic events on the basis of transcriptomic and genomic profiling in patients undergoing carotid endarterectomy. Mol Med. 2012;18:669–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Saigusa R, Winkels H, Ley K. T cell subsets and functions in atherosclerosis. Nat Rev Cardiol. 2020;In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Adler R Janeway’s immunobiology. Choice: Current Reviews for Academic Libraries. 2008;45:1793–1794. [Google Scholar]
- 154.Sallusto F, Mackay CR, Lanzavecchia A. The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol. 2000;18:593–620. [DOI] [PubMed] [Google Scholar]
- 155.Ziegenhain C, Vieth B, Parekh S, et al. Comparative analysis of single-cell rna sequencing methods. Molecular cell. 2017;65:631–643 e634. [DOI] [PubMed] [Google Scholar]
- 156.Galkina E, Harry BL, Ludwig A, Liehn EA, Sanders JM, Bruce A, Weber C, Ley K. Cxcr6 promotes atherosclerosis by supporting t-cell homing, interferon-gamma production, and macrophage accumulation in the aortic wall. Circulation. 2007;116:1801–1811. [DOI] [PubMed] [Google Scholar]
- 157.Butcher MJ, Wu CI, Waseem T, Galkina EV. Cxcr6 regulates the recruitment of pro-inflammatory il-17a-producing t cells into atherosclerotic aortas. Int Immunol. 2016;28:255–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Zaiss DMW, Gause WC, Osborne LC, Artis D. Emerging functions of amphiregulin in orchestrating immunity, inflammation, and tissue repair. Immunity. 2015;42:216–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Wong SH, Walker JA, Jolin HE, et al. Transcription factor roralpha is critical for nuocyte development. Nat Immunol. 2012;13:229–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Hoyler T, Klose CS, Souabni A, Turqueti-Neves A, Pfeifer D, Rawlins EL, Voehringer D, Busslinger M, Diefenbach A. The transcription factor gata-3 controls cell fate and maintenance of type 2 innate lymphoid cells. Immunity. 2012;37:634–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Winkels H, Ehinger E, Ghosheh Y, Wolf D, Ley K. Atherosclerosis in the single-cell era. Current opinion in lipidology. 2018;29:389–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, Satija R, Smibert P. Simultaneous epitope and transcriptome measurement in single cells. Nature methods. 2017;14:865–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Rodriques SG, Stickels RR, Goeva A, et al. Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution. Science. 2019;363:1463–1467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Eng CL, Lawson M, Zhu Q, et al. Transcriptome-scale super-resolved imaging in tissues by rna seqfish. Nature. 2019;568:235–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Newland SA, Mohanta S, Clement M, et al. Type-2 innate lymphoid cells control the development of atherosclerosis in mice. Nature communications. 2017;8:15781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Engblom C, Pfirschke C, Zilionis R, et al. Osteoblasts remotely supply lung tumors with cancer-promoting siglecf(high) neutrophils. Science. 2017;358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Kimura T, Kobiyama K, Winkels H, et al. Regulatory cd4(+) t cells recognize mhc-ii-restricted peptide epitopes of apolipoprotein b. Circulation. 2018;138:1130–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Gaddis DE, Padgett LE, Wu R, et al. Apolipoprotein ai prevents regulatory to follicular helper t cell switching during atherosclerosis. Nature communications. 2018;9:1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

