Skip to main content

EDITORIAL article

Front. Cardiovasc. Med., 31 May 2024
Sec. Cardiovascular Metabolism
This article is part of the Research Topic Exploiting Cellular Immunometabolism as a Strategy for Innovative Cardiovascular Therapies View all 5 articles

Editorial: Exploiting cellular immunometabolism as a strategy for innovative cardiovascular therapies

  • 1Department of Excellence of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università Degli Studi di Milano, Milan, Italy
  • 2Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
  • 3Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States

Editorial on the Research Topic
Exploiting cellular immunometabolism as a strategy for innovative cardiovascular therapies

In the last few years, immuno-inflammation has emerged as one of the driving risk factors for cardiovascular disease. (CVD) (1). This picture is well illustrated by atherosclerosis, a chronic inflammatory lipid-driven disease of the arteries and a major cause of CVD; in this context, the heterogenous activation of the immune response has indeed been shown to be not just bystander to lipid overload, but instead active player of disease progression (2). This is supported by the observation that CVD patients have systemic alterations in the number, proportion and function of immune cells, showing a pro-inflammatory activation that already occurs at the level of haematopoietic precursors in the bone marrow through mechanisms of functional priming, associated with innate trained immunity, and/or somatic mutations in progenitor cells leading to clonal haematopoiesis of indeterminate potential (CHIP) (36). In line with this, neutrophil counts have recently been causally associated to CVD by both observational and genetic approaches (7). This calls for the inclusion of loss of the immuno-inflammatory balance as a risk factor for CVD, and encourages the implementation of therapeutic approaches in the clinic to limit this immuno-inflammatory response (8).

Seminal studies in animal models of atherosclerosis have demonstrated the close relationship between hypercholesterolemia and inflammation in the atherosclerotic plaque. This is also mediated by the excessive release of activated monocytes from the bone marrow and the spleen, which eventually accumulate in the plaque where they differentiate into macrophages (9). The increased proliferation of haematopoietic cells under hypercholesterolemic conditions is linked to changes in sterol metabolism, due to cellular accumulation of cholesterol, and energy metabolism, as a result of a metabolic shift toward glycolysis. The crosstalk between immune cell activation and metabolic adaptations has been further demonstrated in humans by positron emission tomography/computed tomography (PET/CT) imaging using 18F-fluorodeoxyglucose (18F-FDG). In fact, 18F-FDG uptake, which reflects glucose metabolism—particularly increased in metabolically active cells—can non-invasively assess arterial inflammation, which is mainly caused by macrophage uptake in atherosclerotic plaques. Increased 18F-FDG uptake was found in the aorta, bone marrow and spleen of dyslipidaemic patients compared with normocholesterolaemic subjects and was associated with inflammatory biomarkers (10). These observations have also been extended to patients with acute myocardial infarction (11), thus suggesting an increased metabolic activity in these haematopoietic lymphoid districts associated with vascular inflammation, fostering an increased interest in the use of more specific tracers and their combination to better stratify inflammatory risk in CVD patients (12).

This scenario poses the challenge of investigating how the plasticity of cellular metabolism influences the function of cells in the atherosclerotic plaque, leading to the identification of novel molecular pathways that can be targeted to correct the immune-inflammatory response in CVD (13).

From the bench side, cutting-edge research has identified cellular metabolic “checkpoints” whose activity is coupled to functional cellular reprogramming of immune cells. This is the case of the various modulators of cholesterol metabolism, such as the apolipoprotein E, ABCA1 and ABCG1 transporters, the LDL receptor, which have been shown to modulate cellular sterol metabolism in haematopoietic precursors, macrophages, dendritic cells and lymphocytes, in addition to their effect on systemic lipidaemia, thereby controlling cellular functions (1418). Similarly, modulation of glucose and amino acid metabolism in haematopoietic cells differentially modulates the progression of atherosclerosis [reviewed in detail (19)] In parallel, the identification of autoimmunity against modified lipoproteins (20) (particularly against naïve and oxidised LDL and its protein and lipid components) has set the stage for testing atherosclerosis vaccines in clinical trials with the gaol to stimulate the production of antibodies against LDL, training antigen-specific or vasculotropic immunosuppressive T regulatory cells (21, 22). Indeed, the safety of low-dose IL-2 to promote Treg expansion has been demonstrated in patients with stable and acute CVD (23) and is now being tested for clinical benefit (24). While promising, these therapies rely on the patient's immune system, which may carry genetic or epigenetic “scars” that would potentially limit their clinical efficacy. In this setting, cellular immunotherapies based on ex vivo engineered T cells may overcome this limitation, also thanks to their rapid expansion beyond haematological cancers; indeed, recent experimental data suggest that this approach protects against experimental age-related metabolic dysfunction (25), paving the way to for the use of immune cell therapy in the context of cardiovascular and metabolic diseases.

On the other hand, clinical evidence has shown that lipid-lowering interventions have a cardiovascular benefit that goes beyond improving the plasma lipid profile and may be associated with a reduction in inflammatory burden (26). However, it remains to be proven whether the direct effect can be achieved in vivo (after liver metabolism and distribution), as shown in in vitro studies. In line with this, the recent evidence for the beneficial effects of SGLT2 inhibitors on cardiac function in patients with or without diabetes (T2M) has been linked to direct anti-inflammatory and immunomodulatory properties, beyond the improved systemic metabolic phenotype (27, 28).

So, what can we learn from these experimental and clinical evidence to improve CVD stratification and treatment? Cutting-edge technologies are improving our understanding of the function, localisation and metabolism of different cell subsets within the atherosclerotic plaque, helping to identify cell-specific immune and/or metabolic dysfunctions that drive cardiovascular inflammation. Therefore, innovative therapeutic approaches may, on the one hand, take advantage of the repositioning of “metabolic drugs” to target molecular “checkpoints” that couple the reprogramming of the cellular energy machinery with its functional modulation, and, on the other hand, adopt a tailored strategy to target specific subset/cells associated with atheroslcerosis-severity, thereby limiting the side effects of off-target modulation of the immune response.

Author contributions

FB: Writing – original draft, Writing – review & editing. DD-M: Writing – review & editing.

Funding

FB is supported by Progetti di Rilevante Interesse Nazionale (PRIN 2022 2022NBKCWP), Roche Foundation (2022), Fondazione Cariplo (1560-2019).

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. Libby P. Inflammation in atherosclerosis—no longer a theory. Clin Chem. (2021) 67:131–42. doi: 10.1093/clinchem/hvaa275

PubMed Abstract | Crossref Full Text | Google Scholar

2. Bonacina F, Di Costanzo A, Genkel V, Kong XY, Kroon J, Stimjanin E, et al. The heterogeneous cellular landscape of atherosclerosis: implications for future research and therapies. A collaborative review from the EAS young fellows. Atherosclerosis. (2023) 372:48–56. doi: 10.1016/j.atherosclerosis.2023.03.021

PubMed Abstract | Crossref Full Text | Google Scholar

3. De Winther MPJ, Bäck M, Evans P, Gomez D, Goncalves I, Jørgensen HF, et al. Translational opportunities of single-cell biology in atherosclerosis. Eur Heart J. (2023) 44(14):1216–30. doi: 10.1093/eurheartj/ehac686

PubMed Abstract | Crossref Full Text | Google Scholar

4. Bahrar H, Bekkering S, Stienstra R, Netea MG, Riksen NP. Innate immune memory in cardiometabolic disease. Cardiovasc Res. (2023) 119:2774–86. doi: 10.1093/cvr/cvad030

Crossref Full Text | Google Scholar

5. Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC, Chakraborty R, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. (2017) 355(6327):842–7. doi: 10.1126/science.aag1381

PubMed Abstract | Crossref Full Text | Google Scholar

6. Baragetti A, Bonacina F, Catapano AL, Norata GD. Effect of lipids and lipoproteins on hematopoietic cell metabolism and commitment in atherosclerosis. Immunometabolism. (2021) 3(2):e210014. doi: 10.20900/immunometab20210014

PubMed Abstract | Crossref Full Text | Google Scholar

7. Luo J, Thomassen JQ, Nordestgaard BG, Tybjærg-Hansen A, Frikke-Schmidt R. Neutrophil counts and cardiovascular disease. Eur Heart J. (2023) 44:4953–64. doi: 10.1093/eurheartj/ehad649

PubMed Abstract | Crossref Full Text | Google Scholar

8. Engelen SE, Robinson AJB, Zurke YX, Monaco C. Therapeutic strategies targeting inflammation and immunity in atherosclerosis: how to proceed? Nat Rev Cardiol. (2022) 19:522–42. doi: 10.1038/s41569-021-00668-4

PubMed Abstract | Crossref Full Text | Google Scholar

9. Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science. (2013) 339:161–6. doi: 10.1126/science.1230719

PubMed Abstract | Crossref Full Text | Google Scholar

10. Toutouzas K, Skoumas J, Koutagiar I, Benetos G, Pianou N, Georgakopoulos A, et al. Vascular inflammation and metabolic activity in hematopoietic organs and liver in familial combined hyperlipidemia and heterozygous familial hypercholesterolemia. J Clin Lipidol. (2018) 12(1):33–43. doi: 10.1016/j.jacl.2017.10.019

PubMed Abstract | Crossref Full Text | Google Scholar

11. Kim EJ, Kim S, Kang DO, Seo HS. Metabolic activity of the spleen and bone marrow in patients with acute myocardial infarction evaluated by 18F-fluorodeoxyglucose positron emission tomograpic imaging. Circ Cardiovasc Imaging. (2014) 7:454–60. doi: 10.1161/CIRCIMAGING.113.001093

PubMed Abstract | Crossref Full Text | Google Scholar

12. Maier A, Teunissen AJP, Nauta SA, Lutgens E, Fayad ZA, van Leent MMT. Uncovering atherosclerotic cardiovascular disease by PET imaging. Nat Rev Cardiol. (2024). doi: 10.1038/s41569-024-01009-x. [Epub ahead of print].38575752

PubMed Abstract | Crossref Full Text | Google Scholar

13. Bonacina F, Da Dalt L, Catapano AL, Norata GD. Metabolic adaptations of cells at the vascular-immune interface during atherosclerosis. Mol Aspects Med. (2021) 77:100918. doi: 10.1016/j.mam.2020.100918

PubMed Abstract | Crossref Full Text | Google Scholar

14. Bonacina F, Coe D, Wang G, Longhi MP, Baragetti A, Moregola A, et al. Myeloid apolipoprotein E controls dendritic cell antigen presentation and T cell activation. Nat Commun. (2018) 9(1):3083. doi: 10.1038/s41467-018-05322-1

PubMed Abstract | Crossref Full Text | Google Scholar

15. Westerterp M, Gautier EL, Ganda A, Molusky MM, Wang W, Fotakis P, et al. Cholesterol accumulation in dendritic cells links the inflammasome to acquired immunity. Cell Metab. (2017) 25(6):1294–1304.e6. doi: 10.1016/j.cmet.2017.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

16. Zhao Y, Zhang L, Liu L, Zhou X, Ding F, Yang Y, et al. Specific loss of ABCA1 (ATP-binding cassette transporter A1) suppresses TCR (T-cell receptor) signaling and provides protection against atherosclerosis. Arterioscler Thromb Vasc Biol. (2022) 42(12):e311–26. doi: 10.1161/ATVBAHA.122.318226

PubMed Abstract | Crossref Full Text | Google Scholar

17. Yvan-Charvet L, Pagler T, Gautier EL, Avagyan S, Siry RL, Han S, et al. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science. (2010) 328(5986):1689–93. doi: 10.1126/science.1189731

PubMed Abstract | Crossref Full Text | Google Scholar

18. Bonacina F, Moregola A, Svecla M, Coe D, Uboldi P, Fraire S, et al. The low-density lipoprotein receptor–mTORC1 axis coordinates CD8+ T cell activation. J Cell Biol. (2022) 221(11):e202202011. doi: 10.1083/jcb.202202011

PubMed Abstract | Crossref Full Text | Google Scholar

19. Sarrazy V, Viaud M, Westerterp M, Ivanov S, Giorgetti-Peraldi S, Guinamard R, et al. Disruption of Glut1 in hematopoietic stem cells prevents myelopoiesis and enhanced glucose flux in atheromatous plaques of ApoE -/- mice. Circ Res. (2016) 118(7):1062–77. doi: 10.1161/CIRCRESAHA.115.307599

PubMed Abstract | Crossref Full Text | Google Scholar

20. Wolf D, Gerhardt T, Winkels H, Michel NA, Pramod AB, Ghosheh Y, et al. Pathogenic autoimmunity in atherosclerosis evolves From initially protective apolipoprotein B(100)-reactive CD4(+) T-regulatory cells. Circulation. (2020) 142(13):1279–93. doi: 10.1161/CIRCULATIONAHA.119.042863

PubMed Abstract | Crossref Full Text | Google Scholar

21. Hansson GK, Nilsson J. Developing a vaccine against atherosclerosis. Nat Rev Cardiol. (2020) 17:451–2. doi: 10.1038/s41569-020-0407-7

PubMed Abstract | Crossref Full Text | Google Scholar

22. Bonacina F, Martini E, Svecla M, Nour J, Cremonesi M, Beretta G, et al. Adoptive transfer of CX3CR1 transduced-T regulatory cells improves homing to the atherosclerotic plaques and dampens atherosclerosis progression. Cardiovasc Res. (2021) 117(9):2069–82. doi: 10.1093/cvr/cvaa264

PubMed Abstract | Crossref Full Text | Google Scholar

23. Zhao TX, Sriranjan RS, Tuong ZK, Lu Y, Sage AP, Nus M, et al. Regulatory T-cell response to low-dose interleukin-2 in ischemic heart disease. NEJM Evid. (2022) 1(1):EVIDoa2100009. doi: 10.1056/EVIDoa2100009

PubMed Abstract | Crossref Full Text | Google Scholar

24. Sriranjan R, Zhao TX, Tarkin J, Hubsch A, Helmy J, Vamvaka E, et al. Low-dose interleukin 2 for the reduction of vascular inflammation in acute coronary syndromes (IVORY): protocol and study rationale for a randomised, double-blind, placebo-controlled, phase II clinical trial. BMJ Open. (2022) 12(10):e062602. doi: 10.1136/bmjopen-2022-062602

PubMed Abstract | Crossref Full Text | Google Scholar

25. Amor C, Fernández-Maestre I, Chowdhury S, Ho YJ, Nadella S, Graham C, et al. Prophylactic and long-lasting efficacy of senolytic CAR T cells against age-related metabolic dysfunction. Nat Aging. (2024) 4(3):336–49. doi: 10.1038/s43587-023-00560-5

PubMed Abstract | Crossref Full Text | Google Scholar

26. Xie S, Galimberti F, Olmastroni E, Luscher TF, Carugo S, Catapano AL, et al. Effect of lipid-lowering therapies on C-reactive protein levels: a comprehensive meta-analysis of randomized controlled trials. Cardiovasc Res. (2024) 120(4):333–44. doi: 10.1093/cvr/cvae034

PubMed Abstract | Crossref Full Text | Google Scholar

27. Markousis-Mavrogenis G, Baumhove L, Al-Mubarak AA, Aboumsallem JP, Bomer N, Voors AA, et al. Immunomodulation and immunopharmacology in heart failure. Nat Rev Cardiol. (2024) 21(2):119–49. doi: 10.1038/s41569-023-00919-6

PubMed Abstract | Crossref Full Text | Google Scholar

28. Osto E, Bonacina F, Pirillo A, Norata GD. Neutral effect of SGLT2 inhibitors on lipoprotein metabolism: from clinical evidence to molecular mechanisms. Pharmacol Res. (2023) 188:106667. doi: 10.1016/j.phrs.2023.106667

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: cardiovascular disease (CVD), atherosclerosis, immune response, immunometabolism, immunomodulation

Citation: Bonacina F and Della-Morte D (2024) Editorial: Exploiting cellular immunometabolism as a strategy for innovative cardiovascular therapies. Front. Cardiovasc. Med. 11:1435850. doi: 10.3389/fcvm.2024.1435850

Received: 21 May 2024; Accepted: 22 May 2024;
Published: 31 May 2024.

Edited and Reviewed by: Ichiro Manabe, Chiba University, Japan

© 2024 Bonacina and Della-Morte. 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: F. Bonacina, fabrizia.bonacina@unimi.it

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.