Skip to main content

EDITORIAL article

Front. Immunol., 04 October 2022
Sec. Molecular Innate Immunity
This article is part of the Research Topic Endothelial Cells as Innate Immune Cells View all 7 articles

Editorial: Endothelial cells as innate immune cells

  • 1Centers of Cardiovascular Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
  • 2Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
  • 3Department of Medical Genetics and Molecular Biochemistry, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
  • 4Center for Translational Medicine, Department of Medicine, Simmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States

Introduction

With the great effort and support from the authors and editorial team, our Research Topic entitled “Endothelial cells as innate immune cells” in Frontiers in Immunology, Molecular Innate Immunity Section has achieved a great success and have attracted so far 6,775 views and numerous submissions. Endothelial cells are the innermost cell type lined along vessels in all the organs and tissues through hosts, indicating their anatomic and physiological roles in regulating vascular tone, preventing blood components from leaking and maintaining vascular functions (1). The traditional concept emphasized that innate immune cells are non-T cells and non-B cells, which migrate from blood circulation to inflammatory/injury/tumor sites. Our new concept states that immune responses and inflammation and tumorigenesis are highly coordinated processes. Regardless of migration and structural cell types, any cell types can be defined as innate immune cells, which are capable in autonomously sensing danger/pathogen associated molecular patterns (DAMPs/PAMPs) and providing signaling supports to these processes via cell membrane proteins (for example, clusters of differentiation, CDs) and secretomes/cytokines/chemokines/growth factors (2, 3). The pathological roles of endothelial cells in participating in various inflammations and immune responses were reported (3). However, immunological characters of endothelial cells have been poorly characterized due to the fact that endothelial cells have not been recognized as innate immune cells in the field. To fill in this significant knowledge gap, ten years ago based on more than ten major innate immune functional aspects that are shared by the prototypic innate immune cell type - macrophages and endothelial cells, we proposed a new concept that endothelial cells are innate immune cells (2, 4), which was further supported by our experimental data (513) analyses (Shao et al., 15). The Molecular Innate Immunity field is continuously evolving, and we greatly appreciate the Molecular Innate Immunity section editors gave us this opportunity to organize this Research Topic and work with other investigators to explore this important topic further. Here, we are excited to see nearly five publications collected in our topic since 2021 (Table 1).

TABLE 1
www.frontiersin.org

Table 1 Summary for 5 highlighted studies in Frontiers in Immunology: 2022.

Trained immunity is a novel mechanism for persistent hyperactivation and synergies between DAMPs/PAMPs

One of the significant progresses in the innate immunity field is the identification of innate immune memory or trained immunity (1518). Ever since investigators found memory adaptive immune cells such as memory T cells and memory B cells, we have always been puzzled by two questions whether innate immune cells have memory functional status for challenged stimuli and whether innate immune cells have memory cell subtypes similar to that of adaptive immune cells. In response to danger/pathogen associated molecular patterns (DAMPs/PAMPs) and conditional DAMPs (19) derived from injury, lipid peroxidation (15), chronic kidney disease-related uremic toxins stimulation (2022), hyperlipidemia, hyperglycemia (23), hyperhomocysteinemia (2426), metabolic syndrome, hypertension, cigarette smoke, bacterial infections, and virus infections, inflammations take place in vasculature (27). However, two significant questions remain whether vascular innate immune cell types including endothelial cells and vascular smooth muscle cells (VSMCs) have any innate immune memory function to remember risk factor challenges; and whether innate immune cells response differently to the second stimulation after exposed to the first stimulation. Benefit from immunological, metabolic and epigenetic research progresses, new innate immune memory function or trained immunity has been identified. The inflammatory microenvironment and DAMPs/PAMPs can keep aortic endothelial cells, VSMCs, monocytes, macrophages and other innate immune cells persistent hyperactivation and will develop exacerbated immunologic response to the second stimulation (trained immunity) (16, 28, Zhong et al.). For current understanding, trained immunity (15, 16, Zhong et al.) is formed via metabolic reprogramming (29) and epigenetic memory (10, 30, 31). All the cell types in aorta participate in this process including but not limited to endothelial cells, VSMCs (21, 32), monocytes, macrophages, neutrophils, T cells, CD4+ regulatory T cells (Treg) (3337) (Ni et al.; Shao et al.; Xu et al., Zhang et al.) and B cells. In this editorial we will discuss the most recent research and novel insight in endothelial cells (Shao et al.), macrophages (Barhoumi et al.; Dominguez et al.; Lai et al.; Li et al.; Zhang et al.) and neutrophil (Domer et al.; Perez-Figueroa et al.; Rydzynska et al.), which may contribute to the formation of trained immunity in aorta.

It has been reported that endothelial cell is an innate immune cell and can upregulated three major metabolic pathways including glycolysis pathway, mevalonate pathway and acetyl-Co-A synthesis to build immunologic memory by stimulations of various DAMPs/PAMPs such as lysophosphatidylcholine (LPC) (17). Shao et al. paper further reported that the expressions of 1311 innate immune regulators are modulated in 21 human endothelial cell transcriptomic datasets by various DAMPs/PAMPs including Middle East Respiratory Syndrome Coronavirus (COVID-19 homologous virus), lipopolysaccharide (LPS), LPC, shear stress, hyperlipidemia and oxidized low-density lipoprotein (oxLDL) (Shao et al.). In addition, another major cell type in the aortic wall, VSMCs can also undergo metabolic reprogramming to build this immunologic memory by oxidized low-density lipoprotein (oxLDL) stimulation (Schnack et al.). Last but not least, it was a well-documented concept that monocytes and macrophages as prototypic innate immune cell types can establish trained immunity (38). Recent studies further confirmed that the macrophages can be polarized into pro-inflammatory macrophages (M1), anti-inflammatory macrophages (M2), tumor-associated macrophages, adipose tissue macrophages and many other macrophage subsets (Lai et al.) in different conditions (Zhang et al.). The LPS/interferon-γ or SARS-CoV-2 Coronavirus spike protein treatment can push pro-inflammatory macrophage (M1) formation and upregulate ATP-citrate lyase (ACLY), which is the key enzymes catalyzing metabolic reprogramming. The M1 macrophages then switch their metabolism from oxidative phosphorylation (OXPHOS) to glycolysis (Barhoumi et al.; Dominguez et al.). Finally, in a recent paper found after transcriptomic and epigenetic rewiring of granulopoiesis, the trained granulopoiesis promotes an anti-tumor immune phenotype in neutrophils (39). In the inflammatory conditions, the secretion of neutrophil extracellular traps can further stimulate neutrophils to amplify inflammatory response (Domer et al.; Perez-Figueroa et al.). All the evidences indicate that aorta can build the immunologic memory and amplify the immunologic responses in pathological conditions.

A new concept: Aorta may serve as an immune organ in pathologies

Because of the ability to provide a niche for immune cell maturation, differentiation, and activation, lymph nodes and spleen were defined as a peripheral immune organ (40). Current understanding in this field indicated that the aorta may have similar abilities to provide a microenvironment/niche for endothelial cell phenotype switch to mesenchymal cells (4, 16, 41), vascular smooth muscle cells (VSMCs) phenotype switch to macrophages (21, 42) and other five different plastic cell types (43), monocyte differentiation (26, 44, 45), macrophage differentiation (Zhang et al.; 46, 47), neutrophil activation (Domer et al.) as well as T cell and B cell differentiation (44, 48, 49). Based on the principle the same as immunologists previously defined lymph nodes and spleen as immune organs, in terms of function, the aorta can serve as an immune organ in pathological conditions. However, the key knowledge gaps are a) how immune cell differentiation, trans-differentiation, activation take place in aorta and what are the molecular mechanisms underlying the processes. A recent paper found the secretomes (18, 50) (Ni et al.) in aorta play a significant role in providing the microenvironment for immune cell maturation, differentiation and activation. In this paper, author found that approximately 53.7% out of 21,306 human protein-encoding genes are classified into six secretomes including canonical secretome (secretory proteins with signal peptide), caspase 1-GSDMD secretome, caspase 4-GSDMD secretome, exosome secretomes, Weibel–Palade bodies, and autophagy secretome. Those six types of secretomes were significantly modulated in the aorta of pathological condition, including aorta in atherosclerosis, chronic kidney disease, abdominal aortic aneurysm or endothelial cell treated with Middle East Respiratory Syndrome Coronavirus, vascular smooth muscle cell treated with Angiotensin II (Lu et al.). This finding not only filled the knowledge gap by providing us a novel insight that aorta may serve as an immune organ and regulate the immunological response via six types of secretomes in pathological conditions, but also raise the potential targets for future therapeutic interventions for inflammation, cardiovascular disease and autoimmune disease.

In this editorial, we summarized five significant papers (Ellen et al.; Lu et al.; Stolarz et al.; Xu et al.) published on our Research Topic in Frontiers in Immunology, Molecular Innate Immunity Section to illustrate the most recent understanding on molecular innate immunity.

Author contributions

YL and YuS carried out literature collections, research analyses, and drafted the manuscript, KX, YiS, FS, NS, LY, JY, SW, WH, JS, and HW provided editing input. XY supervised and edited the manuscript. All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Funding

This work was supported by the National Institutes of Health Grants to XY (HL132399-01A1; HL138749-01; and HL147565-01), HW (DK104116; DK113775; and HL131460)

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Shao Y, Cheng Z, Li X, Chernaya V, Wang H, Yang XF, et al. Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction-a novel mechanism for maintaining vascular function. J Hematol Oncol (2014) 7(1):80. doi: 10.1186/s13045-014-0080-6

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Shao Y, Saredy J, Yang WY, Sun Y, Lu Y, Saaoud F, et al. Vascular endothelial cells and innate immunity. Arterioscler Thromb Vasc Biol (2020) 40(6):e138–52. doi: 10.1161/ATVBAHA.120.314330

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Yang XF, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for pamps and suppressed by regulatory t cells. Drug Discovery Today Ther Strateg (2008) 5(2):125–42. doi: 10.1016/j.ddstr.2008.11.003

CrossRef Full Text | Google Scholar

4. Mai J, Virtue A, Shen J, Wang H, Yang XF. An evolving new paradigm: Endothelial cells–conditional innate immune cells. J Hematol Oncol (2013) 6:61. doi: 10.1186/1756-8722-6-61

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Yin Y, Li X, Sha X, Xi H, Li YF, Shao Y, et al. Early hyperlipidemia promotes endothelial activation via a caspase-1-sirtuin 1 pathway. Arteriosc Thromb Vasc Biol (2015) 35(4):804–16. doi: 10.1161/ATVBAHA.115.305282

CrossRef Full Text | Google Scholar

6. Lopez-Pastrana J, Ferrer LM, Li YF, Xiong X, Xi H, Cueto R, et al. Inhibition of caspase-1 activation in endothelial cells improves angiogenesis -a novel therapeutic potential for ischemia. J Biol Chem (2015) 290(28):17485–94. doi: 10.1074/jbc.M115.641191

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Li Y-F, Huang X, Li X, Gong R, Yin Y, Nelson J, et al. Caspase-1 mediates hyperlipidemia-weakened progenitor cell vessel repair. Front Biosci (Landmark Edition) (2015) 20(in press):1–1. doi: 10.2741/4383

CrossRef Full Text | Google Scholar

8. Lu Y, Nanayakkara G, Sun Y, Liu L, Xu K, Drummer C, et al. Procaspase-1 patrolled to the nucleus of proatherogenic lipid LPC-activated human aortic endothelial cells induces ROS promoter CYP1B1 and strong inflammation. Redox Biol (2021) 47:102142. doi: 10.1016/j.redox.2021.102142

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Li X, Fang P, Li Y, Kuo Y-M, Andrews AJ, Nanayakkara G, et al. Mitochondrial reactive oxygen species mediate lysophosphatidylcholine-induced endothelial cell activation. Arterioscler Thromb Vasc Biol (2016) 36(6):1090–100. doi: 10.1161/ATVBAHA.115.306964

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Li X, Shao Y, Sha X, Fang P, Kuo Y-M, Andrews AJ, et al. IL-35 (Interleukin-35) suppresses endothelial cell activation by inhibiting mitochondrial reactive oxygen species-mediated site-specific acetylation of H3K14 (Histone 3 lysine 14). Arterioscler Thromb Vasc Biol (2018) 38(3):599–609. doi: 10.1161/ATVBAHA.117.310626

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Li X, Fang P, Sun Y, Shao Y, Yang WY, Jiang X, et al. Anti-inflammatory cytokines IL-35 and IL-10 block atherogenic lysophosphatidylcholine-induced, mitochondrial ROS-mediated innate immune activation, but spare innate immune memory signature in endothelial cells. Redox Biol (2020) 28:101373. doi: 10.1016/j.redox.2019.101373

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Li X, Wang L, Fang P, Sun Y, Jiang X, Wang H, et al. Lysophospholipids induce innate immune transdifferentiation of endothelial cells, resulting in prolonged endothelial activation. J Biol Chem (2018) 293(28):11033–45. doi: 10.1074/jbc.RA118.002752

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Liu M, Wu N, Xu K, Saaoud F, Vasilopoulos E, Shao Y, et al. Organelle crosstalk regulators are regulated in diseases, tumors, and regulatory T cells: Novel classification of organelle crosstalk regulators. Front Cardiovasc Med (2021) 8:713170. doi: 10.3389/fcvm.2021.713170

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Shen H, Wu N, Nanayakkara G, Fu H, Yang Q, Yang WY, et al. Co-Signaling receptors regulate T-cell plasticity and immune tolerance. Front Biosci (Landmark Ed) (2019) 24:96–132. doi: 10.2741/4710

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Drummer C 4th, Saaoud F, Sun Y, Atar D, Xu K, Lu Y, et al. Hyperlipidemia may synergize with hypomethylation in establishing trained immunity and promoting inflammation in NASH and NAFLD. J Immunol Res (2021) 2021:3928323. doi: 10.1155/2021/3928323

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Drummer CT, Saaoud F, Shao Y, Sun Y, Xu K, Lu Y, et al. Trained immunity and reactivity of macrophages and endothelial cells. Arterioscler Thromb Vasc Biol (2021) 41(3):1032–46. doi: 10.1161/ATVBAHA.120.315452

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Lu Y, Sun Y, Drummer C, Nanayakkara GK, Shao Y, Saaoud F, et al. Increased acetylation of H3K14 in the genomic regions that encode trained immunity enzymes in lysophosphatidylcholine-activated human aortic endothelial cells - novel qualification markers for chronic disease risk factors and conditional DAMPs. Redox Biol (2019) 24:101221. doi: 10.1016/j.redox.2019.101221

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Fagenson AM, Xu K, Saaoud F, Nanayakkara G, Jhala NC, Liu L, et al. Liver ischemia reperfusion injury, enhanced by trained immunity, is attenuated in caspase 1/Caspase 11 double gene knockout mice. Pathogens (2020) 9(11):879. doi: 10.3390/pathogens9110879

CrossRef Full Text | Google Scholar

19. Wang X, Li YF, Nanayakkara G, Shao Y, Liang B, Cole L, et al. Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation-novel paradigm and therapeutic potential. J Cardiovasc Transl Res (2016) 9(4):343–59. doi: 10.1007/s12265-016-9700-6

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Yang J, Xu K, Saaoud F, Nanayakkara G, Jhala NC, Liu L, et al. Chronic kidney disease induces inflammatory CD40+ monocyte differentiation via homocysteine elevation and DNA hypomethylation. Circ Res (2016) 119(11):1226–41. doi: 10.1161/CIRCRESAHA.116.308750

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Monroy MA, Fang J, Li S, Ferrer L, Birkenbach MP, Lee IJ, et al. Chronic kidney disease alters vascular smooth muscle cell phenotype. Front Biosci (Landmark Ed) (2015) 20:784–95. doi: 10.2741/4337

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Sun Y, Johnson C, Zhou J, Wang L, Li Y-F, Lu Y, et al. Uremic toxins are conditional danger- or homeostasis-associated molecular patterns. Front Biosci (Landmark Ed) (2018) 23:348–87. doi: 10.2741/4595

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Fang P, Zhang D, Cheng Z, Yan C, Jiang X, Kruger WD, et al. Hyperhomocysteinemia potentiates hyperglycemia-induced inflammatory monocyte differentiation and atherosclerosis. Diabetes (2014) 63(12):4275–90. doi: 10.2337/db14-0809

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Wang H, Jiang XH, Yang F, Gaubatz JW, Ma L, Magera MJ, et al. Hyperhomocysteinemia accelerates atherosclerosis in cystathionine beta-synthase and apolipoprotein e double knock-out mice with and without dietary perturbation. Blood (2003) 101(10):3901–7. doi: 10.1182/blood-2002-08-2606

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Zhang D, Fang P, Jiang X, Nelson J, Moore JK, Kruger WD, et al. Severe hyperhomocysteinemia promotes bone marrow-derived and resident inflammatory monocyte differentiation and atherosclerosis in LDLr/CBS-deficient mice. Circ Res (2012) 111(1):37–49. doi: 10.1161/CIRCRESAHA.112.269472

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Zhang D, Jiang X, Fang P, Yan Y, Song J, Gupta S, et al. Hyperhomocysteinemia promotes inflammatory monocyte generation and accelerates atherosclerosis in transgenic cystathionine beta-synthase-deficient mice. Circulation (2009) 120(19):1893–902. doi: 10.1161/CIRCULATIONAHA.109.866889

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Willerson JT, Ridker PM. Inflammation as a cardiovascular risk factor. Circulation (2004) 109(21 Suppl 1):II2–10. doi: 10.1161/01.CIR.0000129535.04194.38

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Flores-Gomez D, Bekkering S, Netea MG, Riksen NP. Trained immunity in atherosclerotic cardiovascular disease. Arterioscler Thromb Vasc Biol (2021) 41(1):62–9. doi: 10.1161/ATVBAHA.120.314216

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Sun Y, Lu Y, Saredy J, Wang X, Drummer C IV, Shao Y, et al. ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol (2020) 37:101696. doi: 10.1016/j.redox.2020.101696

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Shao Y, Chernaya V, Johnson C, Yang WY, Cueto R, Sha X, et al. Metabolic diseases downregulate the majority of histone modification enzymes, making a few upregulated enzymes novel therapeutic targets-”Sand out and gold stays”. J Cardiovasc Transl Res (2016) 9(1):49–66. doi: 10.1007/s12265-015-9664-y

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Shao Y, Chernaya V, Johnson C, Yang WY, Cueto R, Sha X, et al. Metabolic diseases downregulate the majority of histone modification enzymes, making a few upregulated enzymes novel therapeutic targets–”Sand out and gold stays”. J Cardiovasc Transl Res (2016) 9(1):49–66. doi: 10.1007/s12265-015-9664-y

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Ferrer LM, Monroy AM, Lopez-Pastrana J, Nanayakkara G, Cueto R, Li Y-F, et al. Caspase-1 plays a critical role in accelerating chronic kidney disease-promoted neointimal hyperplasia in the carotid artery. J Cardiovasc Transl Res (2016) 9(2):135–44. doi: 10.1007/s12265-016-9683-3

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Xiong Z, Song J, Yan Y, Huang Y, Cowan A, Wang H, et al. Higher expression of bax in regulatory T cells increases vascular inflammation. Front Biosci (2008) 13:7143–55. doi: 10.2741/3217

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Shao Y, Yang WY, Saaoud F, Drummer C 4th, Sun Y, Xu K, et al. IL-35 promotes CD4+Foxp3+ tregs and inhibits atherosclerosis via maintaining CCR5-amplified treg-suppressive mechanisms. JCI Insight (2021) 6(19):e152511. doi: 10.1172/jci.insight.152511

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Ke X, Wang J, Li L, Chen IH, Wang H, Yang X-F, et al. Roles of CD4+CD25(high) FOXP3+ tregs in lymphomas and tumors are complex. Front Biosci (2008) 13:3986–4001. doi: 10.2741/2986.

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Yang Q, Nanayakkara GK, Drummer C, Sun Y, Johnson C, Cueto R, et al. Low-intensity ultrasound-induced anti-inflammatory effects are mediated by several new mechanisms including gene induction, immunosuppressor cell promotion, and enhancement of exosome biogenesis and docking. Front Physiol (2017) 8:818. doi: 10.3389/fphys.2017.00818

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Lopez-Pastrana J, Shao Y, Chernaya V, Wang H, Yang X-F. Epigenetic enzymes are the therapeutic targets for CD4(+)CD25(+/high)Foxp3(+) regulatory T cells. Transl Res (2015) 165(1):221–40. doi: 10.1016/j.trsl.2014.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol (2020) 20(6):375–88. doi: 10.1038/s41577-020-0285-6

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Kalafati L, Kourtzelis I, Schulte-Schrepping J, Li X, Hatzioannou A, Grinenko T, et al. Innate immune training of granulopoiesis promotes anti-tumor activity. Cell (2020) 183(3):771–785 e12. doi: 10.1016/j.cell.2020.09.058

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Grasso C, Pierie C, Mebius RE, Baarsen van LGM. Lymph node stromal cells: subsets and functions in health and disease. Trends Immunol (2021) 42(10):920–36. doi: 10.1016/j.it.2021.08.009

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Evrard SM, Lecce L, Michelis KC, Nomura-Kitabayashi A, Pandey G, Purushothaman K-R, et al. Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat Commun (2016) 7:11853. doi: 10.1038/ncomms11853

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Shankman LS, Gomez D, Cherepanova OA, Salmon M, Alencar GF, Haskins RM, et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat Med (2015) 21(6):628–37. doi: 10.1038/nm.3866

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Yap C, Mieremet A, Vries CJM, Micha D, Waard V, et al. Six shades of vascular smooth muscle cells illuminated by KLF4 (Kruppel-like factor 4). Arterioscler Thromb Vasc Biol (2021) 41(11):2693–707. doi: 10.1161/ATVBAHA.121.316600

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Winkels H, Ehinger E, Vassallo M, Buscher K, Dinh HQ, Kobiyama K, et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell RNA-sequencing and mass cytometry. Circ Res (2018) 122(12):1675–88. doi: 10.1161/CIRCRESAHA.117.312513

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Fang P, Li X, Dai J, Cole L, Camacho JA, Zhang Y, et al. Immune cell subset differentiation and tissue inflammation. J Hematol Oncol (2018) 11(1):97. doi: 10.1186/s13045-018-0637-x

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Goldstein JL, Ho YK, Basu SK, Brown MS. Binding site on macrophages that mediates uptake and degradation of acetylated low density lipoprotein, producing massive cholesterol deposition. Proc Natl Acad Sci U.S.A. (1979) 76(1):333–7. doi: 10.1073/pnas.76.1.333

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Gistera A, Hansson GK. The immunology of atherosclerosis. Nat Rev Nephrol (2017) 13(6):368–80. doi: 10.1038/nrneph.2017.51

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Butcher MJ, Filipowicz AR, Waseem TC, McGary CM, Crow KJ, Magilnick N, et al. Atherosclerosis-driven treg plasticity results in formation of a dysfunctional subset of plastic IFNgamma+ Th1/Tregs. Circ Res (2016) 119(11):1190–203. doi: 10.1161/CIRCRESAHA.116.309764

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Stemme S, Faber B, Holm J, Wiklund O, Witztum JL, Hansson GK, et al. T Lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc Natl Acad Sci U.S.A. (1995) 92(9):3893–7. doi: 10.1073/pnas.92.9.3893

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Zhang R, Saredy J, Shao Y, Yao T, Liu L, Saaoud F, et al. End-stage renal disease is different from chronic kidney disease in upregulating ROS-modulated proinflammatory secretome in PBMCs - a novel multiple-hit model for disease progression. Redox Biol (2020) 34:101460. doi: 10.1016/j.redox.2020.101460

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: endothelial cells as immune cells, aorta as immune organ, secretome, vascular inflammation, trained immunity

Citation: Lu Y, Sun Y, Xu K, Shao Y, Saaoud F, Snyder NW, Yang L, Yu J, Wu S, Hu W, Sun J, Wang H and Yang X (2022) Editorial: Endothelial cells as innate immune cells. Front. Immunol. 13:1035497. doi: 10.3389/fimmu.2022.1035497

Received: 02 September 2022; Accepted: 20 September 2022;
Published: 04 October 2022.

Edited and Reviewed by:

Ya-Xiong Tao, Auburn University, United States

Copyright © 2022 Lu, Sun, Xu, Shao, Saaoud, Snyder, Yang, Yu, Wu, Hu, Sun, Wang and Yang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Xiaofeng Yang, xfyang@temple.edu

†These authors share first authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.