English Polski
Vol 30, No 1 (2024)
Review paper
Published online: 2024-06-04

open access

Page views 118
Article views/downloads 41
Get Citation

Connect on Social Media

Connect on Social Media

Immune crossroads in atherosclerosis: role of CD8+ T Cells, CD4+ T cells and toll-like receptors in lower extremity arterial disease

Michał Terpiłowski1, Piotr Terlecki1, Stanisław Przywara1, Tomasz Zubilewicz1

Abstract

This review delves into the intricate roles of CD8+ T cells, Th17 cells, regulatory T cells (Tregs), and Toll-like
receptors (TLRs) in the pathogenesis of atherosclerosis, with a particular focus on Lower Extremity Arterial Disease
(LEAD). CD8+ T cells are highlighted for their dual role in atherosclerosis, acting as both exacerbators and
potential protectors within the atherosclerotic environment. Their cytotoxic activity towards cells within plaques
can promote necrotic core formation and plaque instability, while certain subsets, particularly regulatory CD8+
T cells, may exert atheroprotective effects through immunosuppressive functions. Th17 cells, known for their
production of pro-inflammatory cytokines, are implicated in promoting inflammation and disease progression,
suggesting that targeting Th17 cells could be a viable therapeutic strategy. Conversely, Tregs are identified for
their potential to maintain immune balance and prevent excessive inflammatory responses, thereby stabilizing
atherosclerotic lesions. The article also explores the role of TLRs in recognizing pathogen-associated and
damage-associated molecular patterns, triggering inflammatory responses that contribute to atherosclerosis
development and progression. By understanding the complex interplay between these immune components, the
article suggests that modulating the activity of CD8+ T cells, balancing Th17 and Treg responses, and targeting
TLR-mediated signalling pathways could offer new avenues for therapeutic intervention in atherosclerosis and
LEAD. This comprehensive review underscores the need for targeted therapies that can modulate immune
responses, highlighting the potential of immune system targeting in managing atherosclerosis and preventing
LEAD complications.

Article available in PDF format

View PDF Download PDF file

References

  1. Lin J, Chen Y, Jiang N, et al. Burden of peripheral artery disease and its attributable risk factors in 204 countries and territories from 1990 to 2019. Front Cardiovasc Med. 2022; 9: 868370.
  2. Abramson BL, Al-Omran M, Anand SS, et al. Primary Panel, Secondary Panel:. Canadian Cardiovascular Society 2022 Guidelines for Peripheral Arterial Disease. Can J Cardiol. 2022; 38(5): 560–587.
  3. Hirsch AT, Criqui MH, Treat-Jacobson D, et al. Peripheral arterial disease detection, awareness, and treatment in primary care. JAMA. 2001; 286(11): 1317–1324.
  4. Annex BH, Cooke JP. New directions in therapeutic angiogenesis and arteriogenesis in peripheral arterial disease. Circ Res. 2021; 128(12): 1944–1957.
  5. Cecchini AL, Biscetti F, Manzato M, et al. Current medical therapy and revascularization in peripheral artery disease of the lower limbs: impacts on subclinical chronic inflammation. Int J Mol Sci. 2023; 24(22).
  6. Saenz-Pipaon G, Martinez-Aguilar E, Orbe J, et al. The role of circulating biomarkers in peripheral arterial disease. Int J Mol Sci. 2021; 22(7).
  7. Yang WX, Wang FF, Li FF, et al. Immunological analysis of peripheral artery disease. .
  8. Hinkley H, Counts DA, VonCanon E, et al. T cells in atherosclerosis: key players in the pathogenesis of vascular disease. Cells. 2023; 12(17).
  9. Jin M, Fang J, Wang JJ, et al. Regulation of toll-like receptor (TLR) signaling pathways in atherosclerosis: from mechanisms to targeted therapeutics. Acta Pharmacol Sin. 2023; 44(12): 2358–2375.
  10. Mayer A, Zhang Y, Perelson AS, et al. Regulation of T cell expansion by antigen presentation dynamics. Proc Natl Acad Sci U S A. 2019; 116(13): 5914–5919.
  11. Zhou J, Dimayuga PC, Zhao X, et al. CD8(+)CD25(+) T cells reduce atherosclerosis in apoE(-/-) mice. Biochem Biophys Res Commun. 2014; 443(3): 864–870.
  12. Feil S, Fehrenbacher B, Lukowski R, et al. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ Res. 2014; 115(7): 662–667.
  13. 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(9): 1028–1039.
  14. Blanco P, Pitard V, Viallard JF, et al. Increase in activated CD8+ T lymphocytes expressing perforin and granzyme B correlates with disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 2005; 52(1): 201–211.
  15. Schürch 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(4): 460–472.
  16. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011; 473(7347): 317–325.
  17. Cochain C, Zernecke A. Protective and pathogenic roles of CD8 T cells in atherosclerosis. Basic Res Cardiol. 2016; 111(6): 71.
  18. Laera N, Malerba P, Vacanti G, et al. Impact of immunity on coronary artery disease: an updated pathogenic interplay and potential therapeutic strategies. Life (Basel). 2023; 13(11).
  19. Cochain C, Koch M, Chaudhari SM, et al. CD8+ T cells regulate monopoiesis and circulating Ly6C-high monocyte levels in atherosclerosis in mice. Circ Res. 2015; 117(3): 244–253.
  20. Zhou J, Dimayuga PC, Zhao X, et al. CD8(+)CD25(+) T cells reduce atherosclerosis in apoE(-/-) mice. Biochem Biophys Res Commun. 2014; 443(3): 864–870.
  21. Depuydt MAC, Schaftenaar FH, Prange KHM, et al. Single-cell T cell receptor sequencing of paired human atherosclerotic plaques and blood reveals autoimmune-like features of expanded effector T cells. Nat Cardiovasc Res. 2023; 2(2): 112–125.
  22. Maga P, Mikolajczyk TP, Partyka L, et al. Involvement of CD8+ T cell subsets in early response to vascular injury in patients with peripheral artery disease in vivo. Clin Immunol. 2018; 194: 26–33.
  23. Tedgui A, Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. 2006; 86(2): 515–581.
  24. Takatori H, Kanno Y, Watford WT, et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 2009; 206(1): 35–41.
  25. Wang Y, Li W, Zhao T, et al. Interleukin-17-producing CD4+ T cells promote inflammatory response and foster disease progression in hyperlipidemic patients and atherosclerotic mice. Front Cardiovasc Med. 2021; 8: 667768.
  26. Tiemessen MM, Jagger AL, Evans HG, et al. CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proc Natl Acad Sci U S A. 2007; 104(49): 19446–19451.
  27. Wang J, Gao Y, Yuan Y, et al. Th17 Cells and IL-17A in ischemic stroke. Mol Neurobiol. 2024; 61(4): 2411–2429.
  28. Taleb S, Tedgui A, Mallat Z. IL-17 and Th17 cells in atherosclerosis: subtle and contextual roles. Arterioscler Thromb Vasc Biol. 2015; 35(2): 258–264.
  29. Moaaz M, Lotfy H. Changes and significance of T helper-9 cells and interleukin-9 in patients with atherosclerotic chronic lower limb ischemia: Effect on IL-17 release. Vascular. 2020; 28(4): 378–389.
  30. Hata T, Takahashi M, Hida S, et al. Critical role of Th17 cells in inflammation and neovascularization after ischaemia. Cardiovasc Res. 2011; 90(2): 364–372.
  31. Yamaguchi T, Wing JB, Sakaguchi S. Two modes of immune suppression by Foxp3(+) regulatory T cells under inflammatory or non-inflammatory conditions. Semin Immunol. 2011; 23(6): 424–430.
  32. Yamaguchi T, Kishi A, Osaki M, et al. Construction of self-recognizing regulatory T cells from conventional T cells by controlling CTLA-4 and IL-2 expression. Proc Natl Acad Sci U S A. 2013; 110(23): E2116–E2125.
  33. Joly AL, Andersson J. Alternative splicing, FOXP3 and cardiovascular disease. Aging (Albany NY). 2019; 11(7): 1905–1906.
  34. Joly AL, Seitz C, Liu S, et al. Alternative splicing of controls regulatory T cell effector functions and is associated with human atherosclerotic plaque stability. Circ Res. 2018; 122(10): 1385–1394.
  35. Meng X, Yang J, Dong M, et al. Regulatory T cells in cardiovascular diseases. Nat Rev Cardiol. 2016; 13(3): 167–179.
  36. Hu W, Wei R, Wang L, et al. Correlations of MMP-1, MMP-3, and MMP-12 with the degree of atherosclerosis, plaque stability and cardiovascular and cerebrovascular events. Exp Ther Med. 2018; 15(2): 1994–1998.
  37. Ait-Oufella H, Salomon BL, Potteaux S, et al. Natural regulatory T cells control the development of atherosclerosis in mice. Nat Med. 2006; 12(2): 178–180.
  38. Ren J, Li B. The functional stability of FOXP3 and RORγt in Treg and Th17 and their therapeutic applications. Adv Protein Chem Struct Biol. 2017; 107: 155–189.
  39. Xie Jj, Wang J, Tang Tt, et al. The Th17/Treg functional imbalance during atherogenesis in ApoE(-/-) mice. Cytokine. 2010; 49(2): 185–193.
  40. Chai Y, Yin Z, Fan Q, et al. Protective effects of angong niuhuang pill on early atherosclerosis in apoe mice by reducing the inflammatory response. Evid Based Complement Alternat Med. 2019; 2019: 9747212.
  41. Cheng X, Yu X, Ding YJ, et al. The Th17/Treg imbalance in patients with acute coronary syndrome. Clin Immunol. 2008; 127(1): 89–97.
  42. Vijay K. Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int Immunopharmacol. 2018; 59: 391–412.
  43. Curtiss LK, Black AS, Bonnet DJ, et al. Atherosclerosis induced by endogenous and exogenous toll-like receptor (TLR)1 or TLR6 agonists. J Lipid Res. 2012; 53(10): 2126–2132.
  44. Mullick AE, Tobias PS, Curtiss LK. Modulation of atherosclerosis in mice by Toll-like receptor 2. J Clin Invest. 2005; 115(11): 3149–3156.
  45. Fukuda D, Nishimoto S, Aini K, et al. Toll-Like Receptor 9 Plays a Pivotal Role in Angiotensin II-Induced Atherosclerosis. J Am Heart Assoc. 2019; 8(7): e010860.
  46. Sager HB, Dutta P, Dahlman JE, et al. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction. Sci Transl Med. 2016; 8(342): 342ra80.
  47. Gimbrone MA, García-Cardeña G. Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res. 2016; 118(4): 620–636.
  48. Ishibashi M, Sayers S, D'Armiento JM, et al. TLR3 deficiency protects against collagen degradation and medial destruction in murine atherosclerotic plaques. Atherosclerosis. 2013; 229(1): 52–61.
  49. Bhaskar S, Sudhakaran PR, Helen A. Quercetin attenuates atherosclerotic inflammation and adhesion molecule expression by modulating TLR-NF-κB signaling pathway. Cell Immunol. 2016; 310: 131–140.
  50. Patel H, Shaw SG, Shi-Wen Xu, et al. Toll-like receptors in ischaemia and its potential role in the pathophysiology of muscle damage in critical limb ischaemia. Cardiol Res Pract. 2012; 2012: 121237.
  51. Grelier A, Cras A, Balitrand N, et al. Toll-like receptor 3 regulates cord blood-derived endothelial cell function in vitro and in vivo. Angiogenesis. 2013; 16(4): 821–836.
  52. Cen X, Wang B, Liang Y, et al. Small molecule SMU-CX24 targeting toll-like receptor 3 counteracts inflammation: A novel approach to atherosclerosis therapy. Acta Pharm Sin B. 2022; 12(9): 3667–3681.
  53. Li Y, Wang Y, Chen Y, et al. Corilagin ameliorates atherosclerosis in peripheral artery disease via the toll-like receptor-4 signaling pathway and in vivo. Front Immunol. 2020; 11: 1611.