Skip to main content

The causal relationship between thoracic aortic aneurysm and immune cells: a mendelian randomization study

Abstract

One of the pathogenic causes of thoracic aortic aneurysm (TAA), a dangerous vascular condition that can cause aortic rupture, is autoimmune disorders. Currently, immune cell clustering is becoming more and more refined, and the specific immune cell phenotypes involved are yet unknown. Here, we want to clarify the causal link between TAA risk and 731 immune cell traits. There was a Mendelian randomization analysis (MR). We discovered that the presence of TAA led to an increase in CD45 on CD33 HLA-DR myeloid cells, an increase in CD45 on natural killer cells, and a decrease in FSC-A on granulocytes after applying FDR correction. Our research also revealed a strong correlation between the incidence of TAA and an increase in immune cells with CD3 on CD39+ CD4+, and CD25 on IgD CD27 phenotypes. Through genetic techniques, our research has shown the intimate relationship between immune cells and TAA, offering direction for future clinical investigations.

Peer Review reports

Introduction

A thoracic aortic aneurysm (TAA) is characterized by abnormal enlargement, distortion, and a tumor-like protrusion in a segment of the thoracic aorta. The most severe manifestation of TAA is aortic dissection or rupture, yet a significant number of patients remain asymptomatic [1]. The recognized causes include syphilis infection, autoimmune diseases (such as arthritis), connective tissue disorders (such as Marfan syndrome, Loeys Dietz syndrome, and Ehlers-Danlos syndrome), vascular degeneration (linked to factors like smoking and hypertension), and the presence of a bicuspid aortic valve [2]. At present, antihypertensive medications stand as the sole effective palliative treatment for TAA, while surgical replacement of the dilated aorta remains the only proven treatment strategy [3].

Evidence indicates an overexpression of immune response genes in the aortic media of dilated TAA samples, suggesting that inflammation plays a more significant role in the development of TAA in these patients [4, 5]. The role of immune cells in inflammation is substantial; thus, there is a close relationship between immune cells and the onset and progression of TAA [6]. In recent years, there has been a growing identification of new immune cell types with unique immunological roles. A comprehensive investigation into the relationship between TAA and immune cells is imperative. Determining the precise causative association between immune cells and TAA in traditional observational research is challenging due to limitations in sample size, potential reverse causal bias, and the presence of confounding variables [7].

To assess the causal relationship between immune cells and TAA, large-scale whole-genome association studies (GWAS) and Mendelian randomization (MR) techniques can be employed. These approaches help reverse causal correlations in causal inference and mitigate the impact of confounding variables [8]. GWAS serves as a powerful and reliable tool for MR research. Therefore, we gathered GWAS data encompassing 731 distinct types of immune cells and TAA. Subsequently, we utilized MR analysis to delve into the causal relationship between immune cells and TAA.

Method

Study design

Through a two-sample MR analysis, we systematically examined the causal relationship between 731 distinct types of immune cells and TAA. In this statistical method, three key hypotheses were employed to infer the causal connection between exposure factors and outcomes: (1) instrumental variables are strongly associated with exposure factors, (2) instrumental variables are not associated with confounding factors, and (3) instrumental variables are only associated with exposure and outcomes.

Data sources for TAA and 731 immune cells

The Genome-Wide Association Study (GWAS) Catalog offers summary statistics for TAA (GCST90027266) and 731 immune cell traits (accession numbers GCST0002121 to GCST0001391) from publicly accessible GWAS datasets. The TAA data, based on European ethnicity, comprises 1,351 cases and 18,295 controls. Following quality control and attribution, an examination of more than 23 million single nucleotide polymorphisms (SNPs) was conducted [9]. As reported by Valeria Orrù et al., approximately 22 million SNPs impacted 731 immune cell traits across seven groups in a study involving 3,757 Sardinian individuals. Notably, for 459 cell traits at 70 loci, the researchers identified 122 significant (P < 1.28 × 10− 11) independent association signals, with 53 being novel. Among the 731 immunophenotypes, categories included relative cell counts (n = 192), morphological parameters [MP] (n = 32), surface antigen-reflecting median fluorescence intensity (MFI) (n = 389), and absolute cell counts (n = 118) (Supplementary Table 3) [10].

Selection of instrumental variables (IVs)

By current study guidelines, the significance level for each immunological trait’s SNPs was determined at 1 × 10− 5 [10, 11]. Screening only a small subset of SNPs associated with TAA occurs when the P-value is set to 5 × 10-8. To explore more plausible causal relationships and delve into the interaction between TAA and immune cells in greater detail, we adjusted the P-value to 5 × 10-6. Additionally, to mitigate the effects of strong linkage disequilibrium (LD), we employed PLINK software (version v1.90) to implement the clump method, setting the LD r2 threshold to < 0.1 within a distance of 1000 kb [12]. Subsequently, we calculated the F-statistic for each SNP, eliminating those with weak associations (F < 10). Finally, we utilized the screened SNPs for the two-sample MR analysis.

Statistical analysis

For all research, R 4.3.1 (http://www.Rproject.org) was utilized.

To assess the causal association between TAA and immune cells, we employed five different methods: inverse variance weighting (IVW) [13], MR-Egger [14], weighted median [15], weighted model [16], and simple model [17]. Next, the horizontal pleiotropy was explained and ruled out using the MR-PRESSO [18] and MR-Egger approaches. Simultaneously, we assessed the differences between various instrumental variables using Cochrane’s Q statistics. Finally, a leave-one-out analysis was conducted to evaluate the impact of each SNP on MR. The results of the calculations are presented visually through a forest plot, scatter plot, and funnel plot.

Results

The causal effect of TAA on immunophenotypes

Initially, we conducted a two-sample MR analysis to examine the impact of TAA on immune traits. The False Discovery Rate (FDR) approach was employed for P-value adjustment, with the Inverse Variance Weighting (IVW) method serving as the primary analytical technique. A PFDR of 0.05 was set, resulting in the observation of one immune trait. Subsequently, by increasing the PFDR to 0.1, we identified three immune cell traits. Notably, the presence of TAA was associated with increases in CD45 on CD33 HLA DR (β = 0.102, 95% CI = 1.036–1.183, P = 0.003, PFDR = 0.054, Fig. 1, Supplementary Table 1) and CD45 on Natural Killer (β = 0.089, 95% CI = 1.040–1.150, P < 0.001, PFDR = 0.012, Fig. 1, Supplementary Table 1), while FSC − A on granulocyte exhibited a decrease (β = -0.076, 95% CI = 0.880–0.976, P = 0.004, PFDR = 0.083, Fig. 1, Supplementary Table 1). These associations were corroborated by four other MR techniques: MR Egger, Weighted Median, Simple Mode, and Weighted Mode (Fig. 1, Supplementary Fig. 2). The robustness of these causal associations was further assessed through Leave-One-Out analyses, evaluations for horizontal pleiotropy, and heterogeneity tests (Supplementary Fig. 1). Scatter plots and funnel plots were employed as visualizations to depict the results (Supplementary Figs. 1 & 2).

Fig. 1
figure 1

Causal Relationship between TAA and immune cell traits were showed by forest plots. TAA: thoracic aortic aneurysm

The causal effect of immunophenotypes on TAA

We applied the same approach to investigate the impact of immune traits on TAA. Despite being unable to identify any immunophenotype substantially correlated with TAA after FDR adjustment (PFDR < 0.05), two intriguing immunological characteristics emerged when we relaxed the significance threshold to 0.30. Specifically, associations were observed on CD3 on CD39+ CD4+ (β = 0.121, 95% CI = 1.037–1.229, P = 0.005, PFDR = 0.123, Fig. 2, Supplementary Table 2) and CD25 on IgD CD27 (β = 0.209, 95% CI = 1.052–1.445, P = 0.010, PFDR = 0.231). These connections were validated by four additional MR techniques: MR Egger, Weighted Median, Simple Mode, and Weighted Mode (Fig. 2, Supplementary Fig. 4). The strength of these causal linkages was further verified using heterogeneity tests, horizontal pleiotropy evaluations, and Leave-One-Out analysis (Supplementary Fig. 3). Visual representations, such as scatter plots and funnel plots, can be used to illustrate the results (Supplementary Figs. 3 & 4).

Fig. 2
figure 2

Causal Relationship between immune cell traits and TAA were showed by forest plots. TAA: thoracic aortic aneurysm

Discussion

There has been substantial discourse regarding the connection between immunity and the pathophysiology of TAA. In our investigation, we explored the causal relationship between 731 immune cell traits and TAA using publicly available genetic datasets. Notably, three immunophenotypes were identified as being causally affected by TAA (PFDR < 0.10), with an additional two immunophenotypes showing significant causal impact by TAA at a relaxed significance threshold (PFDR < 0.30).

We observed that TAA is associated with an increase in the CD45 expression on the CD33 HLA-DR subgroup of myeloid cells. However, limited information about this specific cell type is available in prior research. Our study underscores the significance of this cell type, highlighting the need for further investigation to delineate its types and functions. Additionally, we found an increase in natural killer (NK) cells following TAA. NK cells, which develop from common lymphoid progenitor cells, are large granular lymphocytes and play a crucial role in connecting adaptive immunity with innate immunity [19]. According to a study utilizing single-cell sequencing data to explore the immunogenicity of ascending thoracic aortic aneurysm, it was found that NK cells communicate with endothelial cells through the CXCL12-CXCR4 and CCL5-ACKR1 axes [20]. This intercellular communication may contribute to the development of TAA. Moreover, it is noteworthy that TAA was associated with a decrease in FSC-A on granulocytes. Within the innate immune system, granulocytes, encompassing neutrophils, eosinophils, and basophils, represent the most prevalent circulating cells. Upon activation, these myeloid polymorphonuclear leukocytes can release characteristic cytoplasmic particles into the surrounding environment [21]. Studies have revealed a rise in neutrophil infiltration within TAA [22]. Neutrophils contribute to the formation of aortic aneurysms by secreting neutrophil extracellular traps and the inflammatory molecule IL-6 [23]. While the association between TAA and eosinophils and basophils has not been thoroughly studied, we hypothesize that a decline in these two cell types may have contributed to an overall decrease in granulocyte counts.

Subsequently, we identified a significant association between the occurrence of TAA and CD3 on CD39+ CD4+ Treg and CD25 on IgD CD27 B cells. Regulatory T cells (Treg), characterized by the expression of Foxp3, CD25, and CD4, exhibit notable immunosuppressive effects [24]. The reduction of the Treg cell population and impaired function in the aorta of patients with abdominal aortic aneurysms is associated with chronic inflammation [23]. Initially identified as a biomarker of activated B cells, CD39 is expressed on activated T cell subpopulations, monocytes, dendritic cells (DCs), and natural killer cells. Recent findings indicate that FOXP3+ Treg cells also express CD39, and ATP hydrolysis is considered a novel mechanism of Treg cell suppression [25]. Furthermore, research has revealed that Treg cells play a protective role against Aortic Aneurysms and Dissections (AAD). Depletion of Treg cells in a mouse model of AAD has been shown to lead to an increased incidence of AAD [26]. Research indicates that the predominant B cell population in experimental abdominal aortic aneurysms (AAA) is comprised of B2 cells. Moreover, in the absence of other B cell subpopulations, B2 cells have been found to suppress the development of AAA and enhance the population of regulatory T cells in the spleen [27]. These findings align with the results of our research, although further evidence will be necessary in the future.

TAA can arise from various immunological disorders, including Behcet’s disease, giant cell arteritis, big arteritis, and others. Therefore, monitoring the local and systemic inflammatory condition of the patients is beneficial for multiple purposes, such as follow-up care, perioperative management, patient stratification, TAA prevention, and surgical timing decisions. To improve patient stratification, numerous inflammatory markers have been investigated in recent decades, but their clinical translation is still pending. To establish a comprehensive monitoring system, it is crucial to explore the variations in immune cell subtypes at different stages of TAA, in conjunction with clinical features such as imaging, blood pressure, heart rate, blood lipids, and inflammation markers. This integrated approach aims to prevent aneurysm enlargement, reduce the risk of rupture, and intervene prior to the development of dissection or rupture. In addition to the common complications of rupture and bleeding, TAA can also lead to uncommon intestinal fistulas, where aortic inflammation and direct friction collaborate to spontaneously form primary fistulas [28, 29]. The infiltration of immune cells could potentially explain the structural changes in blood vessel walls. Unfortunately, the use of Mendelian randomization techniques to investigate the relationship between immune cells and aortic fistulas is not feasible due to the lack of available GWAS data for these patients.

Our investigation is subject to certain limitations. Firstly, the use of GWAS data limited to the European population resulted in incomplete data sources. To facilitate comparison with diverse ethnic origins, additional data covering a broader range of racial/ethnic groups is necessary. Secondly, the potential impact on our results from not exploring interactions between immune cells should be acknowledged. Thirdly, a larger GWAS dataset related to TAA, with an increased number of cases, is essential to ensure the screening of an adequate number of SNPs.

Conclusion

Through our comprehensive MR investigation, we successfully mitigated the impact of potential confounding variables, reverse causal linkages, and other factors. As a result, we established a causal relationship between numerous immunological phenotypes and TAA. This discovery opens up promising avenues for further research into the roles of immune cell subtypes in multiple sclerosis, potentially providing novel insights into early intervention strategies and TAA treatments.

Data availability

The datasets used and/or analysed during the current study available from the corresponding author on reasonable request.

Abbreviations

TAA:

Thoracic aortic aneurysm

AAD:

Aortic aneurysms and dissections

AAA:

Abdominal aortic aneurysms

GWAS:

Genome-wide association study

IVs:

Instrumental variables

IVW:

Inverse variance weighting

MR:

Mendelian Randomization

OR:

Odds ratio

LD:

Linkage disequilibrium

SNPs:

Single Nucleotide Polymorphisms

FDR:

False Discovery Rate

NK:

Natural killer

Treg:

Regulatory T cells

DCs:

Dendritic cells

References

  1. Salameh MJ, Black JH 3rd, Ratchford EV. Thoracic aortic aneurysm. Vasc Med. 2018;23(6):573–8.

  2. Rega S, Farina F, Bouhuis S, de Donato S, Chiesa M, Poggio P, Cavallotti L, Bonalumi G, Giambuzzi I, Pompilio G, et al. Multi-omics in thoracic aortic aneurysm: the complex road to the simplification. Cell Biosci. 2023;13(1):131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Papakonstantinou NA, Rorris FP. Elective replacement of the ascending aorta: is the 5.5-cm threshold appropriate? The insidious, small aorta. Eur J Cardiothorac Surg. 2021;59(3):554–61.

    Article  PubMed  Google Scholar 

  4. Folkersen L, Wagsater D, Paloschi V, Jackson V, Petrini J, Kurtovic S, Maleki S, Eriksson MJ, Caidahl K, Hamsten A, et al. Unraveling divergent gene expression profiles in bicuspid and tricuspid aortic valve patients with thoracic aortic dilatation: the ASAP study. Mol Med. 2011;17(11–12):1365–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Perrucci GL, Rurali E, Gowran A, Pini A, Antona C, Chiesa R, Pompilio G, Nigro P. Vascular smooth muscle cells in Marfan syndrome aneurysm: the broken bricks in the aortic wall. Cell Mol Life Sci. 2017;74(2):267–77.

    Article  CAS  PubMed  Google Scholar 

  6. Li Y, Ren P, Dawson A, Vasquez HG, Ageedi W, Zhang C, Luo W, Chen R, Li Y, Kim S, et al. Single-cell transcriptome analysis reveals dynamic cell populations and Differential Gene expression patterns in control and Aneurysmal Human aortic tissue. Circulation. 2020;142(14):1374–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Fan X, Peng J, Lei L, He J, Huang J, Zheng D, Xu W, Cai S, Chen J. Integrated analysis of immunocyte infiltration and differential gene expression in tricuspid aortic valve-associated thoracic aortic aneurysms. Ann Transl Med. 2020;8(6):285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lawlor DA, Harbord RM, Sterne JA, Timpson N, Davey Smith G. Mendelian randomization: using genes as instruments for making causal inferences in epidemiology. Stat Med. 2008;27(8):1133–63.

    Article  PubMed  Google Scholar 

  9. Roychowdhury T, Lu H, Hornsby WE, Crone B, Wang GT, Guo DC, Sendamarai AK, Devineni P, Lin M, Zhou W, et al. Regulatory variants in TCF7L2 are associated with thoracic aortic aneurysm. Am J Hum Genet. 2021;108(9):1578–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Orru V, Steri M, Sidore C, Marongiu M, Serra V, Olla S, Sole G, Lai S, Dei M, Mulas A, et al. Complex genetic signatures in immune cells underlie autoimmunity and inform therapy. Nat Genet. 2020;52(10):1036–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yu XH, Yang YQ, Cao RR, Bo L, Lei SF. The causal role of gut microbiota in development of osteoarthritis. Osteoarthritis Cartilage. 2021;29(12):1741–50.

    Article  PubMed  Google Scholar 

  12. Genomes Project C, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA, et al. A global reference for human genetic variation. Nature. 2015;526(7571):68–74.

    Article  Google Scholar 

  13. Burgess S, Small DS, Thompson SG. A review of instrumental variable estimators for mendelian randomization. Stat Methods Med Res. 2017;26(5):2333–55.

    Article  PubMed  Google Scholar 

  14. Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–25.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in mendelian randomization with some Invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40(4):304–14.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hartwig FP, Davey Smith G, Bowden J. Robust inference in summary data mendelian randomization via the zero modal pleiotropy assumption. Int J Epidemiol. 2017;46(6):1985–98.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Zhu G, Zhou S, Xu Y, Gao R, Li H, Zhai B, Liu X, He Y, Wang X, Han G, et al. Mendelian randomization study on the causal effects of omega-3 fatty acids on rheumatoid arthritis. Clin Rheumatol. 2022;41(5):1305–12.

    Article  PubMed  Google Scholar 

  18. Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from mendelian randomization between complex traits and diseases. Nat Genet. 2018;50(5):693–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kucuksezer UC, Aktas Cetin E, Esen F, Tahrali I, Akdeniz N, Gelmez MY, Deniz G. The role of natural killer cells in Autoimmune diseases. Front Immunol. 2021;12:622306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tian Z, Zhang P, Li X, Jiang D. Analysis of immunogenic cell death in ascending thoracic aortic aneurysms based on single-cell sequencing data. Front Immunol. 2023;14:1087978.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Vorobjeva NV, Chelombitko MA, Sud’ina GF, Zinovkin RA, Chernyak BV. Role of Mitochondria in the Regulation of Effector Functions of Granulocytes. Cells 2023, 12(18).

  22. Wu D, Choi JC, Sameri A, Minard CG, Coselli JS, Shen YH, LeMaire SA. Inflammatory cell infiltrates in Acute and chronic thoracic aortic dissection. Aorta (Stamford). 2013;1(6):259–67.

    Article  PubMed  Google Scholar 

  23. Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg. 2017;54(3):95–155.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Ohkura N, Sakaguchi S. Transcriptional and epigenetic basis of Treg cell development and function: its genetic anomalies or variations in autoimmune diseases. Cell Res. 2020;30(6):465–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Herrath J, Chemin K, Albrecht I, Catrina AI, Malmstrom V. Surface expression of CD39 identifies an enriched Treg-cell subset in the rheumatic joint, which does not suppress IL-17A secretion. Eur J Immunol. 2014;44(10):2979–89.

    Article  CAS  PubMed  Google Scholar 

  26. Ait-Oufella H, Wang Y, Herbin O, Bourcier S, Potteaux S, Joffre J, Loyer X, Ponnuswamy P, Esposito B, Dalloz M, et al. Natural regulatory T cells limit angiotensin II-induced aneurysm formation and rupture in mice. Arterioscler Thromb Vasc Biol. 2013;33(10):2374–9.

    Article  CAS  PubMed  Google Scholar 

  27. Meher AK, Johnston WF, Lu G, Pope NH, Bhamidipati CM, Harmon DB, Su G, Zhao Y, McNamara CA, Upchurch GR Jr, et al. B2 cells suppress experimental abdominal aortic aneurysms. Am J Pathol. 2014;184(11):3130–41.

  28. Kehagias D, Mulita F, Marlafeka I, Verras GI, Panagiotopoulos I, Kehagias I. Primary aortoenteric fistula: a rare complication of an eroding duodenal stent. Kardiochir Torakochirurgia Pol. 2022;19(3):161–3.

    PubMed  PubMed Central  Google Scholar 

  29. Kehagias D, Mulita F, Panagiotopoulos I, Lampropoulos C, Markopoulos G, Verras GI, Kehagias I. Primary aortoenteric fistula: is endovascular repair the prime option? A review of the literature. Kardiochir Torakochirurgia Pol. 2022;19(4):220–5.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Thank you to all authors for their contributions.

Funding

No Applicable Funding.

Author information

Authors and Affiliations

Authors

Contributions

AW, SP, and GL designed the study. AW, HX, ML, and GL collected and analyzed the data. SP, HX, and AW wrote the manuscript. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Hongli Xia or Ansen Wu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Conflict of interest

The authors declare no competing interests.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, G., Pan, S., Xia, H. et al. The causal relationship between thoracic aortic aneurysm and immune cells: a mendelian randomization study. BMC Cardiovasc Disord 24, 212 (2024). https://doi.org/10.1186/s12872-024-03876-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12872-024-03876-1

Keywords