Thromb Haemost
DOI: 10.1055/a-2348-5697
Review Article

Monocyte Subsets in Cardiovascular Disease: A Biomarker Perspective

Michael Hristov
1   Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten (IPEK), Ludwig-Maximilians-Universität (LMU), München, Germany
,
Christian Weber
1   Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten (IPEK), Ludwig-Maximilians-Universität (LMU), München, Germany
2   Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), München, Germany
3   Munich Cluster for Systems Neurology (SyNergy), München, Germany
4   Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, The Netherlands
› Author Affiliations
Funding The authors are supported by the Deutsche Forschungsgemeinschaft (SFB 1123) and the August-Lenz-Stiftung.
 


Abstract

Endothelial dysfunctions together with a dysregulated immune response and lipid accumulation are important confounding factors in the onset and chronic development of atherosclerosis. Recently, a large body of data has emerged on the sequential involvement of different immune cell types, including monocytes, in the pathology of this disease. In this condensed review, we aim to highlight some of the recent basic research and clinical findings on monocyte subsets published since our joint European Society of Cardiology consensus document, and re-evaluate their potential relevance as surrogate biomarkers in coronary artery disease.


#

Monocyte Biology

Monocytes are the largest blood leukocytes in adults and have a rapid turnover with relatively short lifespan of only a few days in the bloodstream.[1] [2] As cells of innate immunity, they are released from the bone marrow and serve as precursors to tissue macrophages and dendritic cells. Monocytes can secrete inflammatory cytokines and have distinct surface receptors such as integrins, G-protein-coupled and toll-like receptors (TLRs) that allow adhesion, crawling, and migration, as well as direct immune response and phagocytosis.[3] [4] [5] [6] In addition to resolving inflammation, monocytes are involved in angiogenesis, tissue remodeling after injury or scavenging apoptotic cells, and toxic debris at homeostatic conditions.[3] [4] [5] [6]


#

Monocyte Heterogeneity: Phenotype and Function

According to the expression intensity of the lipopolysaccharide (LPS) receptor CD14 and the FcγIII receptor CD16, human monocytes have typically been divided into three major subsets: classical CD142+CD16, intermediate CD142+CD16+, and nonclassical CD14+CD162+.[7] These subsets differ significantly in phenotype and function.

The CD142+CD16 subset (∼85% of total monocytes) highly expresses Ccr2, Cxcr4, FcγRI, L-selectin, and scavenger receptor class A.[5] [8] [9] [10] In contrast, CD14+CD162+ monocytes (∼10%) are smaller and less dense showing higher levels of SLAN, CD31, CD11c, and Cx3cr1 but much lower levels of Ccr2 compared with the classical subset.[5] [8] [9] [10] The CD142+CD16+ subset (∼5%) can be clearly distinguished from the nonclassical by the expression of Ccr2.[10] This lowest subset is strong pro-inflammatory with some unique features that distinguish it from the other two. So far, CD142+CD16+ monocytes are enriched in bone marrow and have the highest expression of angiogenic markers, CD163, MHC class II, HLA-DR, and miR-6087of any subset.[5] [8] [9] [10] [11] Recent comprehensive data obtained by modern technology like mass cytometry with clustering algorithm and RNA sequencing suggest even higher heterogeneity among each subset with three newly identified nonclassical and four classical monocytes.[12]

Functionally, classical monocytes are professional phagocytes that generate reactive oxygen species (ROS) and secrete cytokines upon LPS stimulation.[5] In contrast, nonclassical monocytes do not generate ROS and are weak phagocytes that preferentially take up oxidized LDL (ox-LDL), but secrete substantial amounts of inflammatory cytokines after TLR-dependent activation by viruses and nucleic acids.[5] [6] Thus, nonclassical monocytes may serve as patrolling immune cells that selectively remove virally infected or injured cells and detoxify ox-LDL. The lower phagocytic activity of CD14+CD162+ compared with classical monocytes has been efficiently exploited to enumerate the number and ratio of both subsets by their differential uptake of magnetic nanoparticles.[13] Finally, the intermediate subset does not produce ROS, but shows the highest secretion of tumor necrosis factor-α and interleukin (IL)-1β in response to LPS.[6]

Monocyte subsets undergo linear maturation kinetics in the periphery that is accompanied by a gradual increase in the expression of specific miRNAs.[11] Isotope labeling has revealed that they enter the bloodstream as classical, leave it as intermediate, and re-enter as nonclassical.[14] In contrast to the circulation, we found that the intermediate subset was highly enriched in the bone marrow.[15] This medullar monocyte pool seems to require Ccr2, Cxcr4, and possibly CD44 for mobilization and may give rise to more specialized peripheral Ccr2+ classical monocytes with implication during inflammation and infection, whereas nonclassical monocytes are less abundant in the bone marrow.


#

Monocyte Subsets in CAD: Risk Prediction and Treatment

Epidemiological data 20 years ago have described leukocytosis as an independent risk factor and predictor of future cardiovascular events.[16] Monocyte counts were also positively correlated with in-stent restenosis.[17] Sequential analysis of monocytes by flow cytometry over the last decade has further refined this knowledge and allowed a more detailed assessment of the prognostic value of each subset ([Table 1]).

Table 1

Correlation of different monocyte subsets to CAD and outcomes

Investigators

Subjects (n)

Condition

Target measure

Main outcome

Shantsila et al

245

STEMI

IMD

↑MACE

Abo-Aly et al

100

STEMI

IMD

↑MACE

Boidin et al

245

STEMI

IMD

↑MACE, ↓LVEF

Foster et al

18

STEMI

CD11c+ IMD

↑Troponin, ↑CK, ↑plaque necrosis

Yamamoto et al

50

Stable CAD

IMD

↓Fibrous cap thickness

Arnold et al

40

Stable CAD

IMD, NCM

↑CAD severity

Merinopoulos et al

30

Elective PCI

IMD

↑Chronic post-PCI

Rogacev et al

951

Elective PCI

IMD

↑MACE

Lo et al

588

Asymptomatic

IMD

↑Plaque score, ↑coronary stenosis

Markofski et al

45

Older adults

TLR+ CM, CD16+

↑Cardiometabolic risk

Abbreviations: CAD, coronary artery disease; CK, creatine kinase; CM, classical monocytes; IMD, intermediate monocytes; LVEF, left ventricular ejection fraction; MACE, major adverse cardiovascular events; NCM, nonclassical monocytes; PCI, percutaneous coronary intervention; STEMI, ST-elevation myocardial infarction; TLR, toll-like receptor.


Recently published data have shown that intermediate and nonclassical monocytes predict major adverse cardiovascular events and poor outcome in patients with ST-elevation myocardial infarction (STEMI).[18] [19] [20] Intermediate monocyte activation, as assessed by CD11c expression, was also positively correlated with peak levels of troponin and creatin kinase in patients with myocardial infarction (MI).[21] Beyond MI, CD142+CD16+ monocyte counts were associated with coronary plaque vulnerability in a prospective study of patients with coronary artery disease (CAD).[22] The intermediate subset further predicts severe coronary stenosis in asymptomatic individuals as shown in another retrospective study.[23] Another study revealed a persistent increase in this subset after elective percutaneous coronary intervention as part of the residual inflammation.[24] Collectively, these data are consistent with the first prospective, large cohort study showing that intermediate monocytes are independent predictors of cardiovascular events in risk patients undergoing elective coronary angiography.[25] Furthermore, intermediate and nonclassical monocytes were positively correlated to CAD severity and older participants without manifested chronic disease but with high cardiometabolic risk had elevated percentages of only nonclassical monocytes.[26] [27]

In patients with stable CAD on statin treatment, high levels of PCSK9 associated with increased classical but decreased nonclassical monocytes while the intermediate subset did not differ.[28] The effects of statins on monocyte subsets are elusive and a prospective randomized trial is still missing. In diseases other than CAD, treatment with prednisolone in patients with immune thrombocytopenia reduced the number of intermediate monocytes and attenuated their pro-inflammatory phenotype.[29] Beta blockers also blunted the mobilization of nonclassical monocytes after acute exercise in healthy cyclist.[30] Further data in healthy donors have shown that classical monocytes generate more angiotensin and express higher levels of both angiotensin-converting enzyme type 1 (ACE1) and ACE2 than the other subsets.[31]

Finally, in healthy human volunteers under LPS-induced low-grade inflammation, intermediate and nonclassical monocytes showed the highest inflammatory response by their expression of IL-6 and IL-8.[32] Given the role of IL-6 and IL-8 in the pathogenesis of cardiovascular disease, this may have implications for risk prediction and potential new treatment options ([Table 2]).

Table 2

The magnitude of the effects of different treatments on monocyte subsets

Investigators

Subjects (n)

Condition

Treatment

Main outcome

Krychtiuk et al

69

Stable CAD

Statin

↑PCSK9, ↑CM, ↓NCM

Williams et al

11

Immune thrombocytopenia

Prednisolone

↓IMD

Graff et al

14

Exercise

Nadolol

↓NCM

Rutkowska-Zapała et al

4–12

Healthy donors

No

↑Ang II, ↑ACE1/2 in CM

Thaler et al

12

Low-grade inflammation

LPS

↑IL-6/8 in IMD and NCM

Abbreviations: ACE, angiotensin-converting enzyme; Ang, angiotensin; CAD, coronary artery disease; CM, classical monocytes; IL, interleukin; IMD, intermediate monocytes; LPS, lipopolysaccharide; NCM, nonclassical monocytes.



#

Concluding Remarks

Differential monitoring, especially of intermediate monocyte counts, appears to predict cardiovascular events and may also be of interest in identifying individuals with subclinical atherosclerosis. In addition, subset-tailored drug targeting could lead to the development of novel and more specific therapeutics. The effects of approved drugs such as glucocorticoids, statins, PCSK9- or ACE-inhibitors, and β blockers could also be re-evaluated in relation to monocyte subsets.

It is still essential to define reference values for monocyte subsets in the general population by means of a standardized flow cytometry protocol. Meanwhile, the common markers such as CD14, CD16, HLA-DR, and Ccr2 are available as dried, pre-aliquoted multicolor antibody cocktail and could be used routinely on simple flow cytometers equipped with two lasers. There is also consensus on the gating strategy and the enumeration of absolute counts for each subset.[2] However, the above reagents are currently licensed for research use only. It is therefore important to work toward having them certified for clinical diagnostic use. While standard instrument equipment and easy-to-use protocols are more useful for clinical routine, expensive, high-performance technology with enhanced resolution, such as multiparameter flow cytometry or even mass cytometry, seems essential and more appropriate for basic research.

Taken together, a comprehensive approach could integrate monocyte subsets, alone or in multiple panels, as novel reliable predictive and diagnostic biomarkers, but also as selective therapeutic targets in cardiovascular precision medicine.


#
#

Conflict of Interest

None declared.

The review process for this paper was fully handled by Gregory Y. H. Lip, Editor in Chief.


  • References

  • 1 Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med 2011; 17 (11) 1410-1422
  • 2 Weber C, Shantsila E, Hristov M. et al. Role and analysis of monocyte subsets in cardiovascular disease. Joint consensus document of the European Society of Cardiology (ESC) Working Groups “Atherosclerosis & Vascular Biology” and “Thrombosis”. Thromb Haemost 2016; 116 (04) 626-637
  • 3 Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol 2009; 27: 669-692
  • 4 Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol 2011; 11 (11) 762-774
  • 5 Ruder AV, Wetzels SMW, Temmerman L, Biessen EAL, Goossens P. Monocyte heterogeneity in cardiovascular disease. Cardiovasc Res 2023; 119 (11) 2033-2045
  • 6 Cros J, Cagnard N, Woollard K. et al. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity 2010; 33 (03) 375-386
  • 7 Ziegler-Heitbrock L, Ancuta P, Crowe S. et al. Nomenclature of monocytes and dendritic cells in blood. Blood 2010; 116 (16) e74-e80
  • 8 Wong KL, Tai JJ, Wong WC. et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood 2011; 118 (05) e16-e31
  • 9 Zawada AM, Rogacev KS, Rotter B. et al. SuperSAGE evidence for CD14++CD16+ monocytes as a third monocyte subset. Blood 2011; 118 (12) e50-e61
  • 10 Shantsila E, Wrigley B, Tapp L. et al. Immunophenotypic characterization of human monocyte subsets: possible implications for cardiovascular disease pathophysiology. J Thromb Haemost 2011; 9 (05) 1056-1066
  • 11 Zawada AM, Zhang L, Emrich IE. et al. MicroRNA profiling of human intermediate monocytes. Immunobiology 2017; 222 (03) 587-596
  • 12 Hamers AAJ, Dinh HQ, Thomas GD. et al. Human monocyte heterogeneity as revealed by high-dimensional mass cytometry. Arterioscler Thromb Vasc Biol 2019; 39 (01) 25-36
  • 13 Wildgruber M, Lee H, Chudnovskiy A. et al. Monocyte subset dynamics in human atherosclerosis can be profiled with magnetic nano-sensors. PLoS One 2009; 4 (05) e5663
  • 14 Tak T, Drylewicz J, Conemans L. et al. Circulatory and maturation kinetics of human monocyte subsets in vivo. Blood 2017; 130 (12) 1474-1477
  • 15 Mandl M, Schmitz S, Weber C, Hristov M. Characterization of the CD14++CD16+ monocyte population in human bone marrow. PLoS One 2014; 9 (11) e112140
  • 16 Madjid M, Awan I, Willerson JT, Casscells SW. Leukocyte count and coronary heart disease: implications for risk assessment. J Am Coll Cardiol 2004; 44 (10) 1945-1956
  • 17 Fukuda D, Shimada K, Tanaka A, Kawarabayashi T, Yoshiyama M, Yoshikawa J. Circulating monocytes and in-stent neointima after coronary stent implantation. J Am Coll Cardiol 2004; 43 (01) 18-23
  • 18 Shantsila E, Ghattas A, Griffiths HR, Lip GYH. Mon2 predicts poor outcome in ST-elevation myocardial infarction. J Intern Med 2019; 285 (03) 301-316
  • 19 Abo-Aly M, Shokri E, Chelvarajan L, Tarhuni WM, Tripathi H, Abdel-Latif A. Prognostic significance of activated monocytes in patients with ST-elevation myocardial infarction. Int J Mol Sci 2023; 24 (14) 11342
  • 20 Boidin M, Lip GYH, Shantsila A, Thijssen D, Shantsila E. Dynamic changes of monocytes subsets predict major adverse cardiovascular events and left ventricular function after STEMI. Sci Rep 2023; 13 (01) 48
  • 21 Foster GA, Gower RM, Stanhope KL, Havel PJ, Simon SI, Armstrong EJ. On-chip phenotypic analysis of inflammatory monocytes in atherogenesis and myocardial infarction. Proc Natl Acad Sci U S A 2013; 110 (34) 13944-13949
  • 22 Yamamoto H, Yoshida N, Shinke T. et al. Impact of CD14++CD16+ monocytes on coronary plaque vulnerability assessed by optical coherence tomography in coronary artery disease patients. Atherosclerosis 2018; 269: 245-251
  • 23 Lo SC, Lee WJ, Chen CY, Lee BC. Intermediate CD14++CD16+ monocyte predicts severe coronary stenosis and extensive plaque involvement in asymptomatic individuals. Int J Cardiovasc Imaging 2017; 33 (08) 1223-1236
  • 24 Merinopoulos I, Bhalraam U, Holmes T. et al. Circulating intermediate monocytes CD14++CD16+ are increased after elective percutaneous coronary intervention. PLoS One 2023; 18 (12) e0294746
  • 25 Rogacev KS, Cremers B, Zawada AM. et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol 2012; 60 (16) 1512-1520
  • 26 Arnold KA, Blair JE, Paul JD, Shah AP, Nathan S, Alenghat FJ. Monocyte and macrophage subtypes as paired cell biomarkers for coronary artery disease. Exp Physiol 2019; 104 (09) 1343-1352
  • 27 Markofski MM, Flynn MG. Elevated circulating CD16+ monocytes and TLR4+ monocytes in older adults with multiple cardiometabolic disease risk factors. Exp Gerontol 2021; 154: 111530
  • 28 Krychtiuk KA, Lenz M, Hohensinner P. et al. Circulating levels of proprotein convertase subtilisin/kexin type 9 (PCSK9) are associated with monocyte subsets in patients with stable coronary artery disease. J Clin Lipidol 2021; 15 (03) 512-521
  • 29 Williams EL, Stimpson ML, Lait PJP. et al. Glucocorticoid treatment in patients with newly diagnosed immune thrombocytopenia switches CD14++ CD16+ intermediate monocytes from a pro-inflammatory to an anti-inflammatory phenotype. Br J Haematol 2021; 192 (02) 375-384
  • 30 Graff RM, Kunz HE, Agha NH. et al. β2-Adrenergic receptor signaling mediates the preferential mobilization of differentiated subsets of CD8+ T-cells, NK-cells and non-classical monocytes in response to acute exercise in humans. Brain Behav Immun 2018; 74: 143-153
  • 31 Rutkowska-Zapała M, Suski M, Szatanek R. et al. Human monocyte subsets exhibit divergent angiotensin I-converting activity. Clin Exp Immunol 2015; 181 (01) 126-132
  • 32 Thaler B, Hohensinner PJ, Krychtiuk KA. et al. Differential in vivo activation of monocyte subsets during low-grade inflammation through experimental endotoxemia in humans. Sci Rep 2016; 6: 30162

Address for correspondence

Christian Weber, MD
Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten (IPEK)
LMU München, Pettenkoferstr. 9, DE-80336 München
Germany   

Publication History

Received: 30 April 2024

Accepted: 18 June 2024

Accepted Manuscript online:
19 June 2024

Article published online:
06 July 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag KG
Stuttgart · New York

  • References

  • 1 Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med 2011; 17 (11) 1410-1422
  • 2 Weber C, Shantsila E, Hristov M. et al. Role and analysis of monocyte subsets in cardiovascular disease. Joint consensus document of the European Society of Cardiology (ESC) Working Groups “Atherosclerosis & Vascular Biology” and “Thrombosis”. Thromb Haemost 2016; 116 (04) 626-637
  • 3 Auffray C, Sieweke MH, Geissmann F. Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol 2009; 27: 669-692
  • 4 Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol 2011; 11 (11) 762-774
  • 5 Ruder AV, Wetzels SMW, Temmerman L, Biessen EAL, Goossens P. Monocyte heterogeneity in cardiovascular disease. Cardiovasc Res 2023; 119 (11) 2033-2045
  • 6 Cros J, Cagnard N, Woollard K. et al. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity 2010; 33 (03) 375-386
  • 7 Ziegler-Heitbrock L, Ancuta P, Crowe S. et al. Nomenclature of monocytes and dendritic cells in blood. Blood 2010; 116 (16) e74-e80
  • 8 Wong KL, Tai JJ, Wong WC. et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood 2011; 118 (05) e16-e31
  • 9 Zawada AM, Rogacev KS, Rotter B. et al. SuperSAGE evidence for CD14++CD16+ monocytes as a third monocyte subset. Blood 2011; 118 (12) e50-e61
  • 10 Shantsila E, Wrigley B, Tapp L. et al. Immunophenotypic characterization of human monocyte subsets: possible implications for cardiovascular disease pathophysiology. J Thromb Haemost 2011; 9 (05) 1056-1066
  • 11 Zawada AM, Zhang L, Emrich IE. et al. MicroRNA profiling of human intermediate monocytes. Immunobiology 2017; 222 (03) 587-596
  • 12 Hamers AAJ, Dinh HQ, Thomas GD. et al. Human monocyte heterogeneity as revealed by high-dimensional mass cytometry. Arterioscler Thromb Vasc Biol 2019; 39 (01) 25-36
  • 13 Wildgruber M, Lee H, Chudnovskiy A. et al. Monocyte subset dynamics in human atherosclerosis can be profiled with magnetic nano-sensors. PLoS One 2009; 4 (05) e5663
  • 14 Tak T, Drylewicz J, Conemans L. et al. Circulatory and maturation kinetics of human monocyte subsets in vivo. Blood 2017; 130 (12) 1474-1477
  • 15 Mandl M, Schmitz S, Weber C, Hristov M. Characterization of the CD14++CD16+ monocyte population in human bone marrow. PLoS One 2014; 9 (11) e112140
  • 16 Madjid M, Awan I, Willerson JT, Casscells SW. Leukocyte count and coronary heart disease: implications for risk assessment. J Am Coll Cardiol 2004; 44 (10) 1945-1956
  • 17 Fukuda D, Shimada K, Tanaka A, Kawarabayashi T, Yoshiyama M, Yoshikawa J. Circulating monocytes and in-stent neointima after coronary stent implantation. J Am Coll Cardiol 2004; 43 (01) 18-23
  • 18 Shantsila E, Ghattas A, Griffiths HR, Lip GYH. Mon2 predicts poor outcome in ST-elevation myocardial infarction. J Intern Med 2019; 285 (03) 301-316
  • 19 Abo-Aly M, Shokri E, Chelvarajan L, Tarhuni WM, Tripathi H, Abdel-Latif A. Prognostic significance of activated monocytes in patients with ST-elevation myocardial infarction. Int J Mol Sci 2023; 24 (14) 11342
  • 20 Boidin M, Lip GYH, Shantsila A, Thijssen D, Shantsila E. Dynamic changes of monocytes subsets predict major adverse cardiovascular events and left ventricular function after STEMI. Sci Rep 2023; 13 (01) 48
  • 21 Foster GA, Gower RM, Stanhope KL, Havel PJ, Simon SI, Armstrong EJ. On-chip phenotypic analysis of inflammatory monocytes in atherogenesis and myocardial infarction. Proc Natl Acad Sci U S A 2013; 110 (34) 13944-13949
  • 22 Yamamoto H, Yoshida N, Shinke T. et al. Impact of CD14++CD16+ monocytes on coronary plaque vulnerability assessed by optical coherence tomography in coronary artery disease patients. Atherosclerosis 2018; 269: 245-251
  • 23 Lo SC, Lee WJ, Chen CY, Lee BC. Intermediate CD14++CD16+ monocyte predicts severe coronary stenosis and extensive plaque involvement in asymptomatic individuals. Int J Cardiovasc Imaging 2017; 33 (08) 1223-1236
  • 24 Merinopoulos I, Bhalraam U, Holmes T. et al. Circulating intermediate monocytes CD14++CD16+ are increased after elective percutaneous coronary intervention. PLoS One 2023; 18 (12) e0294746
  • 25 Rogacev KS, Cremers B, Zawada AM. et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol 2012; 60 (16) 1512-1520
  • 26 Arnold KA, Blair JE, Paul JD, Shah AP, Nathan S, Alenghat FJ. Monocyte and macrophage subtypes as paired cell biomarkers for coronary artery disease. Exp Physiol 2019; 104 (09) 1343-1352
  • 27 Markofski MM, Flynn MG. Elevated circulating CD16+ monocytes and TLR4+ monocytes in older adults with multiple cardiometabolic disease risk factors. Exp Gerontol 2021; 154: 111530
  • 28 Krychtiuk KA, Lenz M, Hohensinner P. et al. Circulating levels of proprotein convertase subtilisin/kexin type 9 (PCSK9) are associated with monocyte subsets in patients with stable coronary artery disease. J Clin Lipidol 2021; 15 (03) 512-521
  • 29 Williams EL, Stimpson ML, Lait PJP. et al. Glucocorticoid treatment in patients with newly diagnosed immune thrombocytopenia switches CD14++ CD16+ intermediate monocytes from a pro-inflammatory to an anti-inflammatory phenotype. Br J Haematol 2021; 192 (02) 375-384
  • 30 Graff RM, Kunz HE, Agha NH. et al. β2-Adrenergic receptor signaling mediates the preferential mobilization of differentiated subsets of CD8+ T-cells, NK-cells and non-classical monocytes in response to acute exercise in humans. Brain Behav Immun 2018; 74: 143-153
  • 31 Rutkowska-Zapała M, Suski M, Szatanek R. et al. Human monocyte subsets exhibit divergent angiotensin I-converting activity. Clin Exp Immunol 2015; 181 (01) 126-132
  • 32 Thaler B, Hohensinner PJ, Krychtiuk KA. et al. Differential in vivo activation of monocyte subsets during low-grade inflammation through experimental endotoxemia in humans. Sci Rep 2016; 6: 30162