Skip to main content

GENERAL COMMENTARY article

Front. Cardiovasc. Med., 09 January 2023
Sec. Lipids in Cardiovascular Disease
Volume 9 - 2022 | https://doi.org/10.3389/fcvm.2022.1086136

Commentary: Monocyte and macrophage lipid accumulation results in down-regulated type-I interferon responses

  • Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States

A Commentary on
Monocyte and macrophage lipid accumulation results in down-regulated type-I interferon responses

Willemsen, L., Chen, H. -J., van Roomen, C. P. A. A., Griffith, G. R., Siebeler, R., Neele, A. E., Kroon, J., Hoeksema, M. A., de Winther, M. P. J. (2022). Front. Cardiovasc. Med. 9:829877. doi: 10.3389/fcvm.2022.829877

Macrophages possess remarkable phenotypic and functional plasticity in response to micro-environmental stimuli (1). In atherosclerosis, macrophages sequester extracellular modified lipids, which accumulate as cytoplasmic lipid droplets, to form foamy macrophages that are crucial during all stages of the disease (2). In particular, the death of foamy macrophages contributes to the development of the lipid core and unstable plaques associated with an increased propensity to plaque rupture and acute cardiovascular events (3).

It has been thought that foamy macrophages drives chronic inflammatory responses, yet recent literature demonstrates that foamy macrophages are in fact less inflammatory than their non-foamy counterparts in the plaque (4). Mechanistically, cholesterol loading of macrophages in vitro results in the activation of liver X receptor (LXR), which upregulates genes involved in reverse cholesterol transport and exerts anti-inflammatory effects (5). Myeloid LXR deficiency induces inflammatory gene expression in foamy macrophages and accelerates atherosclerosis (6). LXRs are activated by oxysterols formed in response to increased intracellular cholesterol levels. The foamy peritoneal macrophages of atherosclerosis-prone Ldlr−/− mice fed with a high-cholesterol/high-fat diet also accumulate desmosterol, an LXR ligand with both LXR-dependent and independent effects (7). Desmosterol activates LXR target genes, inhibits sterol regulatory-element binding proteins (SREBPs) target genes, alters fatty acid metabolism, and suppresses genes involved in inflammatory responses, e.g., Il1b, Cxcl9, and Cxcl10 (7). Furthermore, peritoneal macrophages from Ldlr−/− mice fed with a high-fat diet had lower pentose phosphate pathway (PPP) metabolites than mice fed with a normal diet upon lipopolysaccharide stimulation, contributing to diminished lipopolysaccharide-induced production of pro-inflammatory genes (8). Activation of LXR does not affect PPP metabolites, supporting LXR-independent mechanisms of diminished inflammatory phenotypes of foamy macrophage (8). Despite these important progresses, there remains a major knowledge gap in the mechanistic links between lipid accumulation and immune response modulation.

In this issue of Frontiers in Cardiovascular Medicine, leveraging transcriptomic data analyses, Willemsen et al. moved one important step forward toward addressing how foamy cells are less inflammatory (9). The study analyzed four public and newly generated transcriptomic datasets, including: (1) bone marrow-derived macrophages (BMDMs) with or without loading of acetylated low-density lipoprotein (ac-LDL); (2) peritoneal macrophages derived from wildtype and Apoe−/− mice; (3) human monocytes from familial hypercholesterolemia patients and healthy controls; 4) BMDMs with or without the treatment of LXR-agonist GW3965. The results support that ac-LDL loading in murine and human macrophages specifically suppressed interferon-β (IFN-β) secretion and the expression of IFN-stimulated genes (ISGs), but not many other pro-inflammatory genes (9). The downregulation of ISGs could be rescued by exogenous IFN-β supplementation. LXR activation suppressed the expression of ISGs, resembling the effects of lipid loading (9). Monocytes of familial hypercholesterolemia patients also show a deactivated IFN signature which was restored by lipid-lowering therapy (9). The results further strengthened the notion that lipid-loaded foamy macrophages of mice and humans are less inflammatory and the specific involvement of perturbed type-I IFN responses (9).

Many questions remain unanswered. Beyond the role of LXR activation (9), if and how additional molecular mechanisms may be involved in suppressing the expression of ISGs in foamy macrophages remain undetermined. The current analyses highlighted a transcriptomic signature of suppressed IFN-β responses that is consistent in foamy macrophages in vitro and ex vivo, yet the differences among datasets may provide additional insights. Meta-analysis of RNA-seq data from multiple studies will further reveal the effects of lipoproteins with different modifications in macrophages from distinct origins and organ locations. Integrating transcriptomic data with phenotypic and biochemical characterization will inform the precise identity of foamy macrophage and the extent of lipid accumulation required for the suppression of inflammation. Further, it is unknown why the non-foamy macrophages remain not lipid-loaded in the atherosclerotic plaque; for example, if the foamy macrophages arise as a consequence of a relative increase in phagocytic activity, or if non-foamy and foamy macrophages have a distinct spatial distribution with different degrees of lipoprotein retention (10). The study (9) also provides additional mechanistic and therapeutic implications. First, it will be intriguing to determine if the molecular mechanisms of suppressed inflammation by lipid loading may be leveraged for therapeutic applications. Second, unloading excessive cholesterol to reverse the formation of foam cells represents important therapeutic strategies to decrease atherosclerotic plaque burden. The analysis supports that lipid-lowering therapy reverses IFN-β suppression (9), providing a rationale for testing the combination of lipid-lowering and anti-inflammatory therapies for atherosclerosis regression. Third, while current methods capture a snapshot of foamy macrophage profile, gaining insights into the spatial and temporal signature of foam cell development may further unleash their prognostic value.

More broadly, in addition to atherosclerosis, foamy macrophages are frequently observed in different pathological states, including infectious diseases, multiple sclerosis, and cancer (1114). Because of the important roles of type I IFNs in host defense against viruses (15), targeting lipid metabolism in monocytes using lipid-lowering treatment to promote anti-viral defense is potentially promising (9). In tuberculosis granuloma, foamy macrophages act as key participants in both sustaining persistent bacteria and contributing to tissue pathology (11). In multiple sclerosis lesions, myelin ingestion by myeloid cells induces a foamy appearance and confers anti-inflammatory function (12). Accumulation of lipids in tumor-associated macrophages (TAMs) elicits an immunosuppressive phenotype (13). Consequently, disrupting lipid droplet formation in TAMs impeded tumor growth in mice (13). Consistently, large foamy TAMs were more frequently found in colorectal liver metastasis patients with worse prognoses than patients with good prognoses (14). Lastly, other cell types, such as dendritic cells and vascular smooth muscle cells, also take up lipids and form foam cells (1618). The disease- and cell type-specific mechanisms of foam cell biogenesis and function warrant extensive research and may have broad impact.

In summary, this study by Willemsen et al. (9) further motivates the research community to fully dissect the biology of foam cells and their relationship to diseases and to explore the potential role of foam cells as prognostic and therapeutic targets at an immunometabolism level.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Funding

FL is supported by the American Heart Association Postdoctoral Fellowship 20POST35130003. HZ is supported by the NIH grants R00HL130574 and R01HL151611 and the Irving Scholar award by the National Center for Advancing Translational Sciences (NCATS) at NIH through UL1TR001873.

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.

Abbreviations

BMDMs, bone marrow-derived macrophages; IFN, interferon; ISGs, IFN-stimulated genes; LDL, low density lipoprotein; LXR, liver X receptor; PPP, pentose phosphate pathway; TAMs, tumor-associated macrophages.

References

1. Locati M, Curtale G, Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. (2020) 15:123–47. doi: 10.1146/annurev-pathmechdis-012418-012718

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Björkegren JLM, Lusis AJ. Atherosclerosis: recent developments. Cell. (2022) 185:1630–45. doi: 10.1016/j.cell.2022.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. (2013) 13:709–21. doi: 10.1038/nri3520

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Kim K, Shim D, Lee JS, Zaitsev K, Williams JW, Kim KW, et al. Transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models. Circ Res. (2018) 123:1127–42. doi: 10.1161/CIRCRESAHA.118.312804

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Im SS, Osborne TF. Liver X Receptors in Atherosclerosis and Inflammation. Circ Res. (2011) 108:996–1001. doi: 10.1161/CIRCRESAHA.110.226878

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Endo-Umeda K, Kim E, Thomas DG, Liu W, Dou H, Yalcinkaya M, et al. Myeloid Lxr (Liver X Receptor) deficiency induces inflammatory gene expression in foamy macrophages and accelerates atherosclerosis. Arterioscler Thromb Vasc Biol. (2022) 42:719–31. doi: 10.1161/ATVBAHA.122.317583

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Spann NJ, Garmire LX, McDonald JG, Myers DS, Milne SB, Shibata N, et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell. (2012) 151:138–52. doi: 10.1016/j.cell.2012.06.054

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Baardman J, Verberk SGS, Prange KHM, van Weeghel M, van der Velden S, Ryan DG, et al. A defective pentose phosphate pathway reduces inflammatory macrophage responses during hypercholesterolemia. Cell Rep. (2018) 25:2044–52.e5. doi: 10.1016/j.celrep.2018.10.092

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Willemsen L, Chen HJ, van Roomen C, Griffith GR, Siebeler R, Neele AE, et al. Monocyte and Macrophage Lipid Accumulation Results in Down-Regulated Type-I Interferon Responses. Front Cardiovasc Med. (2022) 9:829877. doi: 10.3389/fcvm.2022.829877

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Zhang H, Reilly MP. Who done it? Macrophage mayhem in atherosclerosis. Circ Res. (2018) 123:1106–8. doi: 10.1161/CIRCRESAHA.118.314006

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Russell DG, Cardona PJ, Kim MJ, Allain S, Altare F. Foamy macrophages and the progression of the human tuberculosis granuloma. Nat Immunol. (2009) 10:943–8. doi: 10.1038/ni.1781

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Boven LA, Van Meurs M, Van Zwam M, Wierenga-Wolf A, Hintzen RQ, Boot RG, et al. Myelin-laden macrophages are anti-inflammatory, consistent with foam cells in multiple sclerosis. Brain. (2006) 129:517–26. doi: 10.1093/brain/awh707

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Wu H, Han Y, Rodriguez Sillke Y, Deng H, Siddiqui S, Treese C, et al. Lipid droplet-dependent fatty acid metabolism controls the immune suppressive phenotype of tumor-associated macrophages. EMBO Mol Med. (2019) 11:e10698. doi: 10.15252/emmm.201910698

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Donadon M, Torzilli G, Cortese N, Soldani C, Di Tommaso L, Franceschini B, et al. Macrophage morphology correlates with single-cell diversity and prognosis in colorectal liver metastasis. J Exp Med. (2020) 217:47. doi: 10.1084/jem.20191847

PubMed Abstract | CrossRef Full Text | Google Scholar

15. McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol. (2015) 15:87–103. doi: 10.1038/nri3787

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Wang Y, Dubland JA, Allahverdian S, Asonye E, Sahin B, Jaw JE, et al. Smooth muscle cells contribute the majority of foam cells in apoe (Apolipoprotein E)-deficient mouse atherosclerosis. Arterioscler Thromb Vasc Biol. (2019) 39:876–87. doi: 10.1161/ATVBAHA.119.312434

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Haka AS, Singh RK, Grosheva I, Hoffner H, Capetillo-Zarate E, Chin HF, et al. Monocyte-derived dendritic cells upregulate extracellular catabolism of aggregated low-density lipoprotein on maturation, leading to foam cell formation. Arterioscler Thromb Vasc Biol. (2015) 35:2092–103. doi: 10.1161/ATVBAHA.115.305843

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Pan H, Xue C, Auerbach BJ, Fan J, Bashore AC, Cui J, et al. Single-cell genomics reveals a novel cell state during smooth muscle cell phenotypic switching and potential therapeutic targets for atherosclerosis in mouse and human. Circulation. (2020) 142:2060–75. doi: 10.1161/CIRCULATIONAHA.120.048378

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: atherosclerosis, lipids, macrophages, inflammation, immunometabolism, foam cells

Citation: Li F and Zhang H (2023) Commentary: Monocyte and macrophage lipid accumulation results in down-regulated type-I interferon responses. Front. Cardiovasc. Med. 9:1086136. doi: 10.3389/fcvm.2022.1086136

Received: 01 November 2022; Accepted: 16 December 2022;
Published: 09 January 2023.

Edited by:

Robert Kiss, McGill University, Canada

Reviewed by:

Adil Rasheed, University of Ottawa Heart Institute, Canada

Copyright © 2023 Li and Zhang. 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: Hanrui Zhang, yes hz2418@cumc.columbia.edu

Download