Abstract
Background
Experimental studies characterize adaptive immune response as a critical factor in the progression and complications of atherosclerosis. Yet, it is unclear whether these observations translate to the human situation. This study systematically evaluates cellular components of the adaptive immune response in a biobank of human aortas covering the full spectrum of atherosclerotic disease.Methods and results
A systematic analysis was performed on 114 well-characterized perirenal aortic specimens with immunostaining for T-cell subsets (CD3/4/8/45RA/45RO/FoxP3) and the Th1/non-Th1/Th17 ratio (CD4(+)T-bet(+)/CD4(+)T-bet(-)/CD4(+)/interleukin-17(+) double staining). CD20 and CD138 were used to identify B cells and plasma cells, while B-cell maturation was evaluated by AID/CD21 staining and expression of lymphoid homeostatic CXCL13. Scattered CD4 and CD8 cells with a T memory subtype were found in normal aorta and early, nonprogressive lesions. The total number of T cells increases in progressive atherosclerotic lesions (≈1:5 CD4/CD8 T-cell ratio). A further increase in medial and adventitial T cells is found upon progression to vulnerable lesions.This critical stage is further hallmarked by de novo formation of adventitial lymphoidlike structures containing B cells and plasma cells, a process accompanied by transient expression of CXCL13. A dramatic reduction of T-cell subsets, disappearance of lymphoid structures, and loss of CXCL13 expression characterize postruptured lesions. FoxP3 and Th17 T cells were minimally present throughout the atherosclerotic process.Conclusions
Transient CXCL13 expression, restricted presence of B cells in human atherosclerosis, along with formation of nonfunctional extranodal lymphoid structures in the phase preceding plaque rupture, indicates a "critical" change in the inflammatory footprint before and during plaque destabilization.Free full text
A Change in Inflammatory Footprint Precedes Plaque Instability: A Systematic Evaluation of Cellular Aspects of the Adaptive Immune Response in Human Atherosclerosis
Abstract
Background
Experimental studies characterize adaptive immune response as a critical factor in the progression and complications of atherosclerosis. Yet, it is unclear whether these observations translate to the human situation. This study systematically evaluates cellular components of the adaptive immune response in a biobank of human aortas covering the full spectrum of atherosclerotic disease.
Methods and Results
A systematic analysis was performed on 114 well‐characterized perirenal aortic specimens with immunostaining for T‐cell subsets (CD3/4/8/45RA/45RO/FoxP3) and the Th1/non‐Th1/Th17 ratio (CD4+T‐bet+/CD4+T‐bet−/CD4+/interleukin‐17+ double staining). CD20 and CD138 were used to identify B cells and plasma cells, while B‐cell maturation was evaluated by AID/CD21 staining and expression of lymphoid homeostatic CXCL13. Scattered CD4 and CD8 cells with a T memory subtype were found in normal aorta and early, nonprogressive lesions. The total number of T cells increases in progressive atherosclerotic lesions (≈1:5 CD4/CD8 T‐cell ratio). A further increase in medial and adventitial T cells is found upon progression to vulnerable lesions.
This critical stage is further hallmarked by de novo formation of adventitial lymphoidlike structures containing B cells and plasma cells, a process accompanied by transient expression of CXCL13. A dramatic reduction of T‐cell subsets, disappearance of lymphoid structures, and loss of CXCL13 expression characterize postruptured lesions. FoxP3 and Th17 T cells were minimally present throughout the atherosclerotic process.
Conclusions
Transient CXCL13 expression, restricted presence of B cells in human atherosclerosis, along with formation of nonfunctional extranodal lymphoid structures in the phase preceding plaque rupture, indicates a “critical” change in the inflammatory footprint before and during plaque destabilization.
Introduction
Atherosclerosis is a highly dynamic1–2 metabolic disease with a strong inflammatory component.3–5 Compelling evidence, largely based on data derived from murine models of atherosclerotic disease, implies extensive involvement of the adaptive immune response in the initiation, progression, and complications of atherosclerotic disease.6 As a consequence, the adaptive immune response is considered a potential target for medical interventions.
An open question is how these observations translate to the human situation. There are fundamental immunological and inflammatory differences between mice and humans.7–9 Moreover, murine models of accelerated atherosclerotic disease require an immunologic background with exaggerated Th1‐cell responsiveness in order for atherosclerosis to develop.8–9 Hence, observations from mouse models may be skewed toward Th1 response. Extrapolation of the murine data is further complicated by absence of vulnerable lesion formation in the current models of atherosclerotic disease. As a result, data on a possible involvement of the adaptive immune response in the advanced stages of atherosclerotic disease are missing.
Available human data, on the other hand, largely rely on surgical specimens. Yet, it is important to note that this material generally represents the final stages of the disease process.10–11 As such, data from these human studies are not representative for the earlier and intermediate phases of atherosclerotic disease; hence, knowledge of the nature of the adaptive immune response in the human atherosclerotic process is limited.
Given the above considerations, we regarded an evaluation of the adaptive immune response within the process of atherosclerotic lesion formation, progression, and stabilization to be relevant. To that end, we performed comprehensive and systematic histological assessment of cellular aspects of the adaptive immune response in tissue samples from a biobank of arterial tissue that covers the full spectrum of human atherosclerotic disease. Results from this explorative study confirm an extensive and dynamic presence of cellular components of the adaptive immunity in the human atherosclerotic process, and reveal profound changes in the inflammatory footprint immediately prior to and during the process of plaque destabilization.
Materials and Methods
Patients and Tissue Sampling
Tissue sections were selected from a tissue bank of aortic wall patches that were obtained during liver, kidney, and pancreas transplantation with grafts derived from cadaveric donors. Details of this bank have been described previously by van Dijk et al12 All patches were from grafts that were eligible for transplantation (ie, all donors met the criteria set by The Eurotransplant Foundation). Due to national regulations, only transplantation‐relevant data for donation are available, as such background information on the specimens is limited. Sample collection and handling was performed in accordance with the guidelines of the Medical and Ethical Committee in Leiden, Netherlands and the code of conduct of the Dutch Federation of Biomedical Scientific Societies (http://www.federa.org/?s=1&m=82&p=0&v=4#827).
Each tissue block in the bank was Movat and H&E stained and classified according to the modified American Heart Association (AHA) classification as proposed by Virmani et al by 2 independent observers with no knowledge of the characteristics of the aortic patch.12–13 The tissue block showing the most advanced plaque was used for further studies. In order to obtain balanced and representative study groups, we selected the first 100 aortic wall samples from the tissue bank. Due to the fact that the vulnerable lesions (ie, thin cap fibroatheroma and plaque ruptures) were under‐represented, we randomly selected several aortic wall samples classified as vulnerable lesions from the remaining 250 patches in order to obtain approximately 10 to 12 samples from each atherosclerotic stage. A total of 114 cases were selected for further examination. Demographic data and the causes of death are summarized in Table 1.
Table 1.
Male | Female | |
---|---|---|
N | 64 | 50 |
Mean age, y (SD) | 49.4 (14.3) | 43.8 (17.3) |
Mean length, cm (SD) | 179.9 (11.9) | 166.6 (12.1) |
Mean weight, kg (SD) | 80.1 (16.3) | 64.9 (15.3) |
Mean BMI, kg/m2 (SD) | 24.8 (3.1) | 22.9 (4.4) |
Number of patients with known history of nicotine abuse | 21 (36.8%) | 17 (39.5%) |
Number of patients with known history of hypertension* | 13 (22.8%) | 10 (23.3%) |
Number of patients with known diabetes | 0 (0.0%) | 1 (2.3%) |
Cause of death | ||
Severe head trauma | 11 (17.1%) | 7 (14.0%) |
Cerebral vascular accident | 8 (12.5%) | 10 (20.0%) |
Subarachnoid bleeding | 15 (23.4%) | 14 (28.0%) |
Cardiac arrest | 7 (10.9%) | 0 (0.0%) |
Trauma | 1 (1.5%) | 1 (2.0%) |
Other | 6 (9.3%) | 3 (6.0%) |
Unknown | 16 (25.0%) | 15 (30%) |
Medication | ||
Antihypertensives | 11 (19.3%) | 6 (14.0%) |
Statins | 1 (1.7%) | 1 (2.3%) |
Anticoagulants | 1 (1.7%) | 2 (4.7%) |
Other | 5 (8.8%) | 8 (18.6%) |
None | 31 (54.4%) | 22 (51.2%) |
Unknown | 11 (19.3%) | 9 (20.9%) |
Characterization of the Lesions and Histological Definitions
Aortic samples not showing any signs of intimal thickening and intimal inflammation where classified as normal. The other samples were classified based on the most advanced lesion type present in the section. Lesions were defined as adaptive intimal thickening, intimal xanthoma, pathological intimal thickening, early fibroatheroma, late fibroatheroma, thin‐cap fibroatheroma, acute plaque rupture, healed plaque rupture, and fibrotic calcified plaque. Detailed descriptions concerning a complete overview on plaque processing, morphological analysis, and additional information concerning the studied population are provided in van Dijk et al12
Immunohistochemistry
All specimens used for immunohistochemistry (IHC) were washed in PBS, formalin fixed, and decalcified (Kristensens solution), and paraffin embedded using standard procedures. The details of the antibodies used for IHC are listed in Table 2. Conjugated biotinylated horse anti‐mouse (1:400 dilution; Vector laboratories, Amsterdam, the Netherlands) or Envision+ System–horseradish peroxidase–labeled polymer anti‐mouse/anti‐rabbit (prediluted; Dako, Heverlee, Belgium) functioned as secondary antibodies. Sections were developed with Nova Red or DAB and counterstained with Mayer's hematoxylin or methyl green. Positive and negative controls were always included and performed by adding or omitting the primary antibody on 3‐aminopropyltriethoxysilane slides containing human tonsils.
Table 2.
Antibody, Clone | Host Isotype; Subclass | Specificity | Pretreatment | Dilution | Reference/Source | |
---|---|---|---|---|---|---|
CD3, polyclonal | Rabbit | Pan T cells | 10x Tris/EDTA | pH 9.2 | 1:200 | DAKO |
CD4, 4B12 | Mouse, IgG1‐κ | T‐helper cells | 10x Tris/EDTA | pH 9.2 | 1:200 | DAKO |
CD8, C8/144B | Mouse, IgG1‐κ | Cytotoxic T cells | 10x Tris/EDTA | pH 9.2 | 1:200 | DAKO |
CD45RA, HI100 | Mouse, IgG2b‐κ | Naïve T cells | Citrate | pH 6.0 | 1:2000 | BioLegend |
CD45RO, UCHL1 | Mouse, IgG2a‐κ | Memory T cells | Citrate | pH 6.0 | 1:1000 | BioLegend |
CD20, L26 | Mouse | Pan B cells | Citrate | pH 6.0 | 1:1000 | DAKO |
CD21, IF8(4) | Mouse, IgG1‐κ | Follicular dendritic Cells, mature B cells | Citrate | pH 6.0 | 1:400 | DAKO |
CD138, MI15 | Mouse | Plasma cells | 10x Tris/EDTA | pH 9.2 | 1:1000 | DAKO |
CCR7, ab191575 | Rabbit, polyclonal | Activated T‐lymphocytes | Citrate | pH 9.2 | 1:200 | Abcam |
Tbet, polyclonal | Rabbit, sc‐21003 | T‐helper1 cells | 10x Tris/EDTA | pH 9.2 | 1:800 | Santa Cruz |
IL‐17, polyclonal | Goat | Th‐17 cells | – | pH 9.0 | 1:50 | R&D |
FoxP3, clone 236A/E7 | Mouse | Regulatory T cells | – | pH 9.0 | 1:50 | Abcam |
CXCL13, cat no AF801 | Goat | Lymphorganogenic chemokine | Citrate | pH 6.0 | 1:100 | R&D |
Anti‐AID, EK2 5G9 | Rat | Activation‐induced cytidine deaminase | Citrate | pH 6.0 | 1:16.000 | Cell Signalling |
AID indicates activation‐induced cytidine deaminase; Ig, immunoglobulin; IL, interleukin.
The Foxp3/CD3 and interleukin‐17/CD3 double staining was performed to identify regulatory T cells and Th17 cells, respectively (Table 1). Conjugated polymer anti‐mouse/anti‐rabbit (Immunologic) functioned as secondary antibodies. After the goat‐derived anti‐interleukin‐17 incubation, a rabbit anti‐goat secondary antibody (Southern Biotech, Birmingham, AL) served as a bridge reagent for the next step with alkaline phosphatase anti‐rabbit polymer (Immunologic, Duiven, the Netherlands). All immunohistochemical stainings were performed as previously described.14 Despite many attempts and various techniques, we did not succeed in using GATA‐3 as the T‐helper 2 marker. We therefore had to turn to double staining for CD4/Tbet to identify the T‐helper lineage (Lineage: Th1=CD4+/Tbet+ and non‐Th1=CD4+/Tbet−) having regard to the provision that non‐Th1 may also include Th0, Th17, and Th22 populations.
Assessment of Immunolocalization of T Cells (and T‐Cell Subset), B Cells, and Plasma Cells
The tissue block showing the most advanced plaque was used for further studies. When the aortic sample contained more than 1 lesion, the most advanced lesion on the slide was used. The lesion was defined as the area between the endothelium and the first elastic lamina over a distance of 1 mm. In the presence of a necrotic core, the distance was expanded with another 500 μm on both sides of the necrotic core to include the adjacent shoulder region.12 The regions of interest were the intima (and in the presence of an early or late necrotic core, the intima was divided into a cap and both shoulder regions), the media, and the adventitia (Figure 1). Within the regions of interest (ie, intima, media, and adventitia), 3 representative adjacent images were made at a ×200 magnification. A total of 9 images per atherosclerotic lesion were separately analyzed from the lesion. Immunostaining intensity was quantitatively assessed with an image‐processing program (Image J; plug‐in Cellcounter). Representative examples of the immunological stainings are provided in Figure 2.
Statistical Analysis
Data in figures are presented as mean±SEM. Spearman's correlation was used to demonstrate the relationship between the amount of positive cells in the intima, media, and adventitia within 1 phase of the atherosclerotic disease and compared with the previous and/or next phase in atherosclerosis (SPSS 20.0; Chicago, IL). The study population consisted of 114 individuals. However, during the staining process some tissue samples were lost. Therefore, the total number of samples for each staining may be less than 114. The Wilcoxon–Mann–Whitney test was used to analyze the non‐normally distributed data of the mean total number of cells within each atherosclerotic phase, and the Kruskal–Wallis test was used to control for a type 1 error. A value of P<0.05 was considered statistically significant.
Results
Studied Population
Characteristics of the studied population are provided in Table 1. The male/female ratio was evenly distributed (57% male), as was the mean age for each sex (≈49 years). There was a strong correlation between donor age and atherosclerosis progression12 (Table 3). The mean donor age for the aortic wall samples classified as normal was 23 years, whereas the mean age for aortas with fibrotic calcified plaques, representing the final stage of the disease, was 62 years. Nearly 40% of patients had a known history of smoking, and 2 patients received statin therapy. Twenty‐three patients had a known history of hypertension; 17 of these patients received antihypertensive medication.
Table 3.
Morphological Description | Abbreviation | Male | Female | Total | ||
---|---|---|---|---|---|---|
N | Mean Age (SD) | N | Mean Age (SD) | N | ||
Normal aorta | N | 7 | 23.8 (18.9) | 5 | 10.0 (3.7) | 12 |
Nonprogressive intimal lesions | ||||||
Adaptive intimal thickening | AIT | 5 | 40.5 (10.0) | 5 | 30.4 (9.0) | 10 |
Intimal xanthoma | IX | 7 | 37.3 (10.0) | 5 | 34.2 (14.8) | 12 |
Progressive atherosclerotic lesions | ||||||
Pathological intimal thickening | PIT | 5 | 46.0 (8.9) | 7 | 55.4 (11.5) | 12 |
Early fibroatheroma | EFA | 7 | 50.9 (5.9) | 5 | 46.0 (3.5) | 12 |
Late fibroatheroma | LFA | 5 | 55.0 (3.6) | 7 | 51.1 (11.6) | 12 |
Vulnerable atherosclerotic lesions | ||||||
Thin‐cap fibroatheroma | TCFA | 7 | 57.2 (5.2) | 4 | 60.3 (5.7) | 11 |
Plaque rupture | PR | 7 | 56.9 (2.6) | 3 | 45.0 (13.1) | 10 |
Stabilizing lesions | ||||||
Healing rupture | HR | 8 | 58.1 (11.1) | 4 | 57.0 (0) | 12 |
Fibrotic calcified plaque | FCP | 6 | 62.7 (6.8) | 5 | 53.0 (11.4) | 11 |
64 | 49.4 (14.3) | 50 | 43.8 (17.3) | 114 |
AIT indicates adaptive intimal thickening; EFA, early fibroatheroma; FCP, fibrotic calcified plaque; HR, healed rupture; IX, Intimal xanthoma; LFA, late fibroatheroma; N, normal; PIT, pathological intimal thickening; PR, plaque rupture; SD, standard deviation; TCFA, thin cap fibroatheroma.
Normal Aorta and Nonprogressive Atherosclerotic Lesions
The intima and media layers of the normal (nonatherosclerotic) aortic wall are devoid of CD3+ T cells (Figures (Figures33 through 10). Scattered T cells found at the medial–adventitial border are mainly CD45RO+ (memory) T cells. The CD4+ (Thelper) to CD8+ (cytotoxic) T‐cell ratio is approximately 1:2 (Figure 3A). Th1 cells) and non‐Th1 cells were also expressed in a 1:2 ratio (Figure 3B).
Naïve T cells (CD45RA+), regulatory T cells (CD3+/FoxP3+), Th17 cells (CD3+/IL17A+), B cells (CD20+), and plasma cells (CD138+) are all absent in aortas classified as normal.
Isolated T cells in the thickened intima, and progressive numbers of T cells in the medial/adventitial border characterize so‐called nonprogressive lesions (ie, adaptive intimal thickening and intimal xanthomas). Memory T cells remain the dominant phenotype, and the subset ratios remains unchanged (CD4+/8+; Th1/Th2). Regulatory T cells, Th17 cells, naïve T cells, B cells, and plasma cells are absent in these early lesions (Figure 4).
Progressive Atherosclerotic Lesions
A significant increase is seen in intimal CD3+ T cells (P<0.0001) in progressive lesions (early fibroatheroma and late fibroatheroma) with beginning infiltration of the medial wall as well. The predominance of cytotoxic T cells persists (CD4+: CD8+ T‐cell ratio 1:5) in the early fibroatheroma, and T‐helper cells are dominated by the Th1 lineage at a 1:3 ratio. Transition from an early to late fibroatheroma stage is accompanied by the accumulation of T cells in the intima shoulder regions of the lesion, and the CD4+/CD8+ T‐cell ratio shifts from 1:5 in early fibroat‐heroma to ≈1.5:1 in late fibroatheroma (Figures (Figures33 through through99).
Memory T cells remain the dominant phenotype in advanced lesions, yet scattered naïve T cells (CD45RA+) start to appear within the medial–adventitial border in lesions classified as early fibroatheroma. A further increase in naïve T cells and T memory cells is seen in late fibroatheroma with a notable accumulation of T cells alongside infiltrating vasa vasorum in the outer layers of the media underlying the necrotic core. In general, B cells and plasma cells are absent, although present in a minority of individuals (Figure 9). Regulatory T cells and Th17 cells are not identified in the progressive lesions.
The Vulnerable Lesions
Thin‐cap fibroatheromas and ruptures are hallmarked by a peak in T‐cell infiltration in the adventitia underneath the culprit lesion (Figures (Figures33 through through9).9). The thin cap remains devoid of T cells, and the number of T‐helper cells decreases, resulting in an approximate 1:1 CD4/CD8 cell ratio. Non‐Th1 cells remain the dominant T‐helper lineage in the vulnerable phase, due to doubling of the number of non‐Th1 cells (Figure 6). Dispersed memory T cells and naïve T cells are abundantly present in the media and adventitia near the vasa vasorum. Absence of CD45RO/CCR7 double‐positive cells indicates that all CD45RO+ T cells within the lesion and infiltrates are effector memory T cell and the only few central memory T cells are identified in the aortic wall located within the vasa vasorum of the adventitia (Figure 10).
A unique finding in the vulnerable lesions is the appearance of B cells and occasional plasma cells in tertiary folliclelike structures. Staining for CXCL13, a homeostatic, lymphorganogenic chemokine that is critical for organization of folliclelike structures, shows CXCL13 expression exclusively in vulnerable lesions (viz, no CXCL13 staining was found in the other stages of the disease) and reveals a particular staining pattern of a dendritic network with extension from the follicles (Figure 9). Additional staining for mature B cells (CD2) and activation‐induced cytidine deaminase was mainly negative, indicating the lack of B‐cell maturation. Scattered FoxP3+ T cells are found within the intima and underlying adventitia in 2 of 9 aortic sections of thin‐cap fibroatheroma (Figure 4). On the other hand, Th17 cells are absent in vulnerable lesions.
Stabilized Lesions
Healing plaque ruptures exhibit a dramatic decrease in T cells (P<0.001), naïve T cells, and disappearance of B cells and plasma cells (Figures (Figures22 through through9).9). The decline in T cells is greater for the CD4+ cells than for the CD8+ cells for all vascular layers. Within the thin‐cap fibroatheroma, non‐Th1 cells dominate over Th1 with a 5‐fold‐increase, and in stable lesions the Th1/non‐Th1 cell ratio returns to 1:2 as seen in normal aortas (Figure 3B). The decline in memory T cells predominantly reflects a reduction localized to the intima (P<0.01). The overall number of memory T cells in the media and adventitia remains stable. Native T‐cell numbers are highly variable. The folliclelike structures vanish and CXCL13 disappeared from the arterial wall. No regulatory T cells or Th17 cells are detected in the stabilized lesions.
Discussion
A wealth of preclinical evidence supports a critical role of the adaptive immunity in the initiation and progression of atherosclerosis.17–18 This study, based on histological observations in human aorta sections, confirms extensive and progressive involvement of cellular components of the adaptive immune response in atherosclerosis.19 Findings point to profound changes in the nature of the response in the lead‐up to plaque destabilization and show extinguishing of inflammatory processes upon plaque stabilization. This study reveals fundamental differences between the human atherosclerotic disease and mouse models of the disease, particularly with respect to a very limited presence of regulatory T cells, absence of Th17 cells throughout the atherosclerotic process, and lack of B cells in the early‐, intermediate‐, and final stages of the process.
Insight into the atherosclerotic process greatly depends on observations from murine models of the disease.20 Indeed, genetically modified mouse models have been critical for understanding the atherosclerotic process. Yet, by virtue of the metabolic adaptations in the lipoprotein metabolism, necessary to induce atherosclerotic lesion formation, the process in these animals is essentially lipid driven (a situation that may not fully mimic the human situation).21–22 Translation of rodent findings is further obscured by critical dependence on genetic backgrounds with Th1‐dominated immune responses in order for atherosclerosis to develop; by the fundamental and intrinsic differences in inflammatory and immune responses between mice and humans; and by failure of the experimental lesions to progress to culprit lesions (vulnerable plaque) formation.23,7,24 Consequently, information provided by these models may be biased, and is incomplete at least with respect to vulnerable lesions. As a result, the preclinical observations may not directly translate to the human situation.8–9
Data on human atherosclerosis are also limited, a situation largely reflecting the fact that most observations are made on material obtained during surgical procedures (eg, endarterectomy). This material typically represents the final stage(s) of the disease and, in the case of an endarterectomy material, will not provide information on the outer media and the adventitia, both major interphases in vessel wall inflammation. With this in mind, we set up a biobank of aortic wall samples from organ grafts designated for transplantation. Material from this bank almost covers the full life span (5 to 80 years) and shows a nearly equal sex distribution. The relatively healthy premortal status of the donors is reflected by minimal use of statins and antihypertensive drugs. Classification was done for all individual tissue sections in the bank (viz, each individual tissue block was Movat and hematoxylin stained, and histologically staged using an established adapted version of the AHA classification system).12–13 Modifications in the adapted classification system highlight specific critical morphological events in the final stages of the disease process. This allows for a more precise interpretation of processes occurring during plaque destabilization and subsequent healing. An earlier systematic evaluation of material in the biobank showed that the bank covers the full spectrum of atherosclerotic disease.12 Exact morphologic descriptions and examples of the different lesions have been published and discussed previously.12
Immunohistochemical staining for CD3, CD4, and CD8 shows progressive T‐cell accumulation during the atherosclerotic process. The earlier phases are dominated by diffuse cytotoxic T‐cell infiltration, but progressive quantities of T‐helper cells are found during progression of the disease, resulting in an increase in the CD4+/CD8+ T‐cell ratio during disease progression. These observations are in line with an earlier report on renal artery atherosclerosis, and with observations from other progressive inflammatory disorders.25–26 Due to inherent limitations of paraffin‐embedded tissue, we were unable to test whether these shifts reflect an (auto)immune phenomenon, as has been proposed in the context of advanced atherosclerotic disease.27–28
CD4/T‐Bet double staining was used to identify Th1 cells. T‐bet is the lineage‐defining transcription factor for Th1 cells.29 In the absence of Th17 and with minimal regulatory T cells throughout the atherosclerotic spectrum, we defined the CD4+/T‐bet‐negative cells as non‐Th1 cells. Quantification of the Th1/non‐Th1 ratio on the basis of the CD4/T‐Bet double staining did not confirm Th1 dominance in the human atherosclerotic process. This may indicate that the presumed Th1 dominance in atherosclerosis reflects an artificial phenomenon related to the requirement of Th1‐dominated backgrounds in mouse models in order for atherosclerosis to develop. Yet, alternative explanations are that identification based on T‐Bet positivity underestimates the number of Th1 cells or, vice versa, that identification based on cytokine profiles overestimates the number of Th1 cells.30
Findings for the CD45RO (T‐memory cells) and CD45RA (naïve T cells) largely follow observations for other solid organs (predominance of T‐memory cells) and extend those made in rodent models of atherosclerotic disease showing increasing T‐cell infiltration during disease progression, with a clear maximum during plaque destabilization followed by a rapid decline during plaque stabilization (plaque healing).31 A remarkable and novel observation is the change in the inflammatory footprint that accompanies plaque destabilization, and that fully resolves during plaque stabilization. This change appears to precede plaque destabilization, and is confined to the area adjacent to the culprit lesion. On the cellular level, this change is characterized by a sharp increase in the number of T‐helper cells, emergence of naïve T‐cells, and the appearance of B cells and occasional plasma cells in tertiary folliclelike structures. B cells in the follicles are largely activation‐induced cytidine deaminase and CD21 negative, indicating gross absence of B‐cell maturation.32–33 These observations suggest that signals promoting B‐cell homing and follicle assembly accompany (and may even precede) plaque destabilization, but that essential signals critical for maturation of B cells are missing.
The chemokine CXCL13 is a pivotal homeostatic signal for follicle formation.34–35 As for the appearance of folliclelike structures, we performed immunostaining for CXCL‐13 and observed CXCL‐13 expression exclusively in association with the folliclelike structures in the vulnerable lesions (viz, CXCL‐13 was not detected in the other stages of the disease). CXCL13 distribution shows a remarkable radiating pattern of tubelike structures that may reflect expression in dendritic cells.36
In light of the remarkable association between CXCL13 and plaque instability, we tested an association between plasma CXCL13 levels and acute cardiovascular events (acute myocardial infarction) in plasma samples of the Mission study.36 CXCL13 was largely undetectable, and no association was found between CXCL‐13 and cardiovascular events (results not shown). Absence of such an observation may reflect the highly localized character of CXCL13 expression in the vulnerable lesion.
B cells are considered critical players in the atherosclerotic process, albeit their role (protective, detrimental) is still under debate.37–38 Our findings of a very restricted presence of B cells in human atherosclerosis follow observations from Frostegård et al39 Hence, these findings and gross absence of signs of B‐cell maturation in the infiltrating B cells exclude an autocrine or paracrine role of B cells in the human atherosclerotic process. Yet, humoral factors released by B cells in paravascular lymph nodes or more distant locations may well be involved in the disease process.
Regulatory T cells were incidentally found in a subset of the vulnerable lesions, whereas Th17 cells were fully absent in the aortic sections. This latter observation follows results of a comprehensive analysis of cytokines in atherosclerotic wall samples that failed to detect interleukin‐17 in all samples tested (detection threshold of the assay <1 pg/L).40 Although these observations do not exclude a distant role for these cell populations, they seem to contradict observations from murine models implying these cells as local key‐players in the atherosclerotic process.
Limitations
This study was performed on aortic sections of deceased individuals, as such the continuous data in this study are composed of incidental findings from a large series of patients and therefore may not necessarily reflect longitudinal data. Another limitation of the study is the fact the all findings are based on IHC using paraffin‐embedded tissue sections. Although IHC has the advantage of showing the spatial relationships, multiple stainings are elaborative, and only allow for very limited marker sets. Consequently, findings in this study should be considered at a global level. That is, the apparent mismatch between the total number of CD3 counts and the sum of CD4 and CD8 may reflect abundance of other CD3‐expressing populations such as NK and NK‐T cells but also reflect technical limitations of IHC with different epitope availabilities and antibody efficacies. Yet, it is likely that the latter phenomena would equally apply to all tissue sections, and it is thus unlikely that this would influence the conclusions of the study.
In conclusion, this study shows clear changes in the cellular components of the adaptive immune system in anticipation of and during plaque destabilization. Observations suggest that changes in the inflammatory footprint precede and accompany plaque instability. It is tempting to speculate that delineation of this chain of events may provide clues for early culprit lesion detection and plaque stabilization.
References
- Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005; 352:1685-1695. [Abstract] [Google Scholar]
- Hansson GK, Libby P. The immune response in atherosclerosis: a double‐edged sword. Nat Rev Immunol. 2006; 6:508-519. [Abstract] [Google Scholar]
- Mallat Z, Taleb S, Ait‐Oufella H, Tedgui A. The role of adaptive T cell immunity in atherosclerosis. J Lipid Res. 2009; 50:S364-S369. [Europe PMC free article] [Abstract] [Google Scholar]
- Libby P, Ridker PM, Hansson GK. Inflammation in atherosclerosis: from pathophysiology to practice. J Am Coll Cardiol. 2009; 54:2129-2138. [Europe PMC free article] [Abstract] [Google Scholar]
- Seok J, Warren HS, Cuenca AG, Mindrinos MN, Baker HV, Xu W, Richards DR, McDonald‐Smith GP, Gao H, Hennessy L, Finnerty CC, López CM, Honari S, Moore EE, Minei JP, Cuschieri J, Bankey PE, Johnson JL, Sperry J, Nathens AB, Billiar TR, West MA, Jeschke MG, Klein MB, Gamelli RL, Gibran NS, Brownstein BH, Miller‐Graziano C, Calvano SE, Mason PH, Cobb JP, Rahme LG, Lowry SF, Maier RV, Moldawer LL, Herndon DN, Davis RW, Xiao W, Tompkins RG. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci. 2013; 110:3507-3512. [Europe PMC free article] [Abstract] [Google Scholar]
- Weber C, Zernecke A, Libby P. The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models. Nat Rev Immunol. 2008; 8:802-815. [Abstract] [Google Scholar]
- Mestas J, Hughes CCW. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004; 172:2731-2738. [Abstract] [Google Scholar]
- Schulte S, Sukhova GK, Libby P. Genetically programmed biases in Th1 and Th2 immune responses modulate atherogenesis. Am J Pathol. 2008; 172:1500-1508. [Europe PMC free article] [Abstract] [Google Scholar]
- Dansky HM, Charlton SA, Sikes JL, Heath SC, Simantov R, Levin LF, Shu P, Moore KJ, Breslow JL, Smith JD. Genetic background determines the extent of atherosclerosis in ApoE‐deficient mice. Arterioscler Thromb Vasc Biol. 1999; 19:1960-1968. [Abstract] [Google Scholar]
- De Boer OJ, Van Der Meer JJ, Teeling P, Van Der Loos CM, Van Der Wal AC. Low numbers of FOXP3 positive regulatory T cells are present in all developmental stages of human atherosclerotic lesions. PLoS One. 2007; 2:e779. [Europe PMC free article] [Abstract] [Google Scholar]
- Tavora F, Kutys R, Li L, Ripple M, Fowler D, Burke A. Adventitial lymphocytic inflammation in human coronary arteries with intimal atherosclerosis. Cardiovas Pathol. 2009; 19:61-68. [Abstract] [Google Scholar]
- van Dijk RA, Virmani R, von der Thüsen JH, Schaapherder AF, Lindeman JHN. The natural history of aortic atherosclerosis: a systematic histopathological evaluation of the peri‐renal region. Atherosclerosis. 2010; 210:100-106. [Abstract] [Google Scholar]
- Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2000; 20:1262-1275. [Abstract] [Google Scholar]
- de Boer OJ, Li X, Teeling P, Mackaay C, Ploegmakers HJ, van der Loos CM, Daemen MJ, de Winter RJ, van der Wal AC. Neutrophils, neutrophil extracellular traps and interleukin‐17 associate with the organisation of thrombi in acute myocardial infarction. Thromb Haemost. 2013; 109:290-297. [Abstract] [Google Scholar]
- Chen L, Bea F, Wangler S, Celik S, Wang Y, Böckler D, Dengler TJ. Inhibition of IL‐17A attenuates atherosclerotic lesion development in apoE‐deficient mice. J Immunol. 2009; 183:8167-8175. [Abstract] [Google Scholar]
- Mackay CR. Dual personality of memory T cells. Nature. 1999; 401:659-660. [Abstract] [Google Scholar]
- Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science. 2013; 339:161-166. [Europe PMC free article] [Abstract] [Google Scholar]
- Wigren M, Nilsson J, Kolbus D. Lymphocytes in atherosclerosis. Clin Chim Acta. 2012; 413:1562-1568. [Abstract] [Google Scholar]
- Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011; 473:317-325. [Abstract] [Google Scholar]
- Kleemann R, Zadelaar S, Kooistra T. Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res. 2008; 79:360-376. [Europe PMC free article] [Abstract] [Google Scholar]
- Emerging Risk Factors Collaboration. ;Di Angelantonio E, Sarwar N, Perry P, Kaptoge S, Ray KK, Thompson A, Wood AM, Lewington S, Sattar N, Packard CJ, Collins R, Thompson SG, Danesh J. Major lipids, apolipoproteins, and risk of vascular disease. J Am Med Assoc. 2009; 302:1993-2000. [Europe PMC free article] [Abstract] [Google Scholar]
- Lichtman AH. Adaptive immunity and atherosclerosis: mouse tales in the AJP. Am J Pathol. 2013; 182:5-9. [Europe PMC free article] [Abstract] [Google Scholar]
- Huber SA, Sakkinen P, David C, Newell MK, Tracy RP. T helper‐cell phenotype regulates atherosclerosis in mice under conditions of mild hypercholesterolemia. Circulation. 2001; 103:2610-2616. [Abstract] [Google Scholar]
- Ni M, Chen WQ, Zhang Y. Animal models and potential mechanisms of plaque destabilisation and disruption. Heart. 2009; 95:1393-1398. [Abstract] [Google Scholar]
- Kotliar C, Juncos L, Inserra F, de Cavanagh EM, Chuluyan E, Aquino JB, Hita A, Navari C, Sánchez R. Local and systemic cellular immunity in early renal artery atherosclerosis. Clin J Am Soc Nephrol. 2012; 7:224-230. [Abstract] [Google Scholar]
- Aoshiba K, Koinuma M, Yokohori N, Nagai ARespiratory Failure Research Group in Japan. Differences in the distribution of CD4+ and CD8+ T cells in emphysematous lungs. Respiration. 2004; 71:184-190. [Abstract] [Google Scholar]
- Artemiadis AK, Anagnostouli MC. Apoptosis of oligodendrocytes and post‐translational modifications of myelin basic protein in multiple sclerosis: possible role for the early stages of multiple sclerosis. Eur Neurol. 2010; 63:65-72. [Abstract] [Google Scholar]
- Hussein MR, Hassan HI, Hofny ER, Elkholy M, Fatehy NA, Abd Elmoniem AE, Ezz El‐Din AM, Afifi OA, Rashed HG. Alterations of mononuclear inflammatory cells, CD4/CD8+ T cells, interleukin 1beta, and tumour necrosis factor alpha in the bronchoalveolar lavage fluid, peripheral blood, and skin of patients with systemic sclerosis. J Clin Pathol. 2005; 58:178-184. [Europe PMC free article] [Abstract] [Google Scholar]
- Oestreich KJ, Weinmann AS. T‐bet employs diverse regulatory mechanisms to repress transcription. Trends Immunol. 2012; 33:78-83. [Europe PMC free article] [Abstract] [Google Scholar]
- de Boer OJ, van der Wal AC, Verhagen CE, Becker AE. Cytokine secretion profiles of cloned T cells from human aortic atherosclerotic plaques. J Pathol. 1999; 188:174-179. [Abstract] [Google Scholar]
- Alberts‐Grill N, Rezvan A, Son DJ, Qiu H, Kim CW, Kemp ML, Weyand CM, Jo H. Dynamic immune cell accumulation during flow‐induced atherogenesis in mouse carotid artery: an expanded flow cytometry method. Arterioscler Thromb Vasc Biol. 2012; 32:623-632. [Europe PMC free article] [Abstract] [Google Scholar]
- Houtkamp MA, de Boer OJ, van der Loos CM, van der Wal AC, Becker AE. Adventitial infiltrates associated with advanced atherosclerotic plaques: structural organization suggests generation of local humoral immune responses. J Pathol. 2001; 193:263-269. [Abstract] [Google Scholar]
- Hu Y, Ericsson I, Doseth B, Liabakk NB, Krokan HE, Kavli B. Activation‐induced cytidine deaminase (AID) is localized to subnuclear domains enriched in splicing factors. Exp Cell Res. 2014; 322:178-192. [Abstract] [Google Scholar]
- Smedbakken LM, Halvorsen B, Daissormont I, Ranheim T, Michelsen AE, Skjelland M, Sagen EL, Folkersen L, Krohg‐Sørensen K, Russell D, Holm S, Ueland T, Fevang B, Hedin U, Yndestad A, Gullestad L, Hansson GK, Biessen EA, Aukrust P. Increased levels of the homeostatic chemokine CXCL13 in human atherosclerosis—potential role in plaque stabilization. Atherosclerosis. 2012; 224:266-273. [Abstract] [Google Scholar]
- Daissormont I, Lipp M, Biessen EAL. The CXCL13‐CXCR5 axis contributes to atherosclerosis development in LDLr−/− mice by regulating lymphocyte migration to perivascular lymph organs. Cardiovasc Res. 2010; 87:S43-S44. [Abstract] [Google Scholar]
- Liem SS, van der Hoeven BL, Oemrawsingh PV, Bax JJ, van der Bom JG, Bosch J, Viergever EP, van Rees C, Padmos I, Sedney MI, van Exel HJ, Verwey HF, Atsma DE, van der Velde ET, Jukema JW, van der Wall EE, Schalij MJ. MISSION!: optimization of acute and chronic care for patients with acute myocardial infarction. Am Heart J. 2007; 153:e1-e11. [Abstract] [Google Scholar]
- Ait‐Oufella H, Herbin O, Bouaziz JD, Binder CJ, Uyttenhove C, Laurans L, Taleb S, Van Vré E, Esposito B, Vilar J, Sirvent J, Van Snick J, Tedgui A, Tedder TF, Mallat Z. B cell depletion reduces the development of atherosclerosis in mice. J Exp Med. 2010; 207:1579-1587. [Europe PMC free article] [Abstract] [Google Scholar]
- Caligiuri G, Nicoletti A, Poirier B, Hansson GK. Protective immunity against atherosclerosis carried by B cells of hypercholesterolemic mice. J Clin Invest. 2002; 109:745-753. [Europe PMC free article] [Abstract] [Google Scholar]
- Frostegård J, Ulfgren A‐K, 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. [Abstract] [Google Scholar]
- Lindeman JH, Abdul‐Hussien H, Schaapherder AF, Van Bockel JH, Von der Thüsen JH, Roelen DL, Kleemann R. Enhanced expression and activation of pro‐inflammatory transcription factors distinguish aneurysmal from atherosclerotic aorta: IL‐6 and IL‐8 dominated inflammatory responses prevail in the human aneurysm. Clin Sci. 2008; 114:687-697. [Abstract] [Google Scholar]
Articles from Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease are provided here courtesy of Wiley
Full text links
Read article at publisher's site: https://doi.org/10.1161/jaha.114.001403
Read article for free, from open access legal sources, via Unpaywall: https://www.ahajournals.org/doi/pdf/10.1161/JAHA.114.001403
Citations & impact
Impact metrics
Article citations
Enrichment of type 1 innate lymphoid cells in the course of human atherosclerotic plaque development suggests contribution to atherogenesis.
Front Immunol, 15:1354617, 04 Apr 2024
Cited by: 1 article | PMID: 38638438 | PMCID: PMC11024276
Cytokine Profiling of Plasma and Atherosclerotic Plaques in Patients Undergoing Carotid Endarterectomy.
Int J Mol Sci, 25(2):1030, 14 Jan 2024
Cited by: 0 articles | PMID: 38256102 | PMCID: PMC10816498
Cellular and Molecular Mechanisms of Mast Cells in Atherosclerotic Plaque Progression and Destabilization.
Clin Rev Allergy Immunol, 66(1):30-49, 30 Jan 2024
Cited by: 1 article | PMID: 38289515 | PMCID: PMC10973084
Review Free full text in Europe PMC
Bone marrow mesenchymal stem cellsderived exosomes stabilize atherosclerosis through inhibiting pyroptosis.
BMC Cardiovasc Disord, 23(1):441, 07 Sep 2023
Cited by: 2 articles | PMID: 37679676 | PMCID: PMC10486039
Meta-Analysis of Single-Cell RNA-Seq Data Reveals the Mechanism of Formation and Heterogeneity of Tertiary Lymphoid Organ in Vascular Disease.
Arterioscler Thromb Vasc Biol, 43(10):1867-1886, 17 Aug 2023
Cited by: 2 articles | PMID: 37589134 | PMCID: PMC10521807
Go to all (55) article citations
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
Cytotoxic and proinflammatory CD8+ T lymphocytes promote development of vulnerable atherosclerotic plaques in apoE-deficient mice.
Circulation, 127(9):1028-1039, 08 Feb 2013
Cited by: 167 articles | PMID: 23395974
Infectious agents, inflammation, and growth factors: how do they interact in the progression or stabilization of mild human atherosclerotic lesions?
Ann Vasc Surg, 20(5):638-645, 17 Sep 2006
Cited by: 8 articles | PMID: 16983590
Artery tertiary lymphoid organs contribute to innate and adaptive immune responses in advanced mouse atherosclerosis.
Circ Res, 114(11):1772-1787, 01 May 2014
Cited by: 80 articles | PMID: 24855201
Review
[Immunological Analysis of Human Atherosclerotic Plaques in ex vivo Culture System].
Kardiologiia, 56(11):78-85, 01 Dec 2016
Cited by: 2 articles | PMID: 28290822