Skip to main content

EDITORIAL article

Front. Immunol., 09 August 2023
Sec. Immunological Tolerance and Regulation
This article is part of the Research Topic Heme Proteins: Key Players in the Regulation of Immune Responses View all 5 articles

Editorial: Heme proteins: key players in the regulation of immune responses

Claudia Volpi*Claudia Volpi1*Benoît J Van den Eynde,,,Benoît J Van den Eynde2,3,4,5Ciriana OrabonaCiriana Orabona1
  • 1Department of Medicine and Surgery, University of Perugia, Perugia, Italy
  • 2Ludwig Institute for Cancer Research, Brussels, Belgium
  • 3de Duve Institute, Université catholique de Louvain, Brussels, Belgium
  • 4Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
  • 5Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom

Heme is the iron-containing prosthetic group of heme proteins, with varied functions ranging from gas carriers (i.e., hemoglobin) to metabolic enzymes (i.e., cytochromes, indoleamine dioxygenases). Free heme is scavenged by hemopexin (Hx), and degraded by the activity of the heme oxygenases HO-1 and HO-2. In response to heme-mediated inflammation, cells upregulate the expression of HO-1, an inducible enzyme that degrades heme into biliverdin, carbon monoxide (CO), and iron; biliverdin is subsequently converted to bilirubin. Thus, HO-1 induction functions as a negative feedback mechanism to mitigate the pro-inflammatory effects of heme by reducing its availability (1). Similarly, biliverdin, CO, and bilirubin are also endowed with anti-inflammatory and cytoprotective properties (2). Since free heme is generated by mechanisms of red blood cells damage and tissue injuries, it is considered a danger-associated molecular pattern (DAMP) capable of activating Toll-like receptor 4 (TLR4). Heme-triggered TLR4 can induce the production of pro-inflammatory cytokines, chemokines, and reactive oxygen species (ROS) in various immune cells, such as macrophages, neutrophils, and endothelial cells, promoting the recruitment and activation of further immune cells and causing tissue damage and inflammation (3).

Heme is a well-established pro-inflammatory signaling molecule involved in different pathological settings. Seika et al. elegantly demonstrate that the accumulation of free heme in the colon amplifies the DNA damage, the abnormal proliferation of epithelial cells and can sustain a state of chronic inflammation. This mechanism provides a novel explanation for gastrointestinal syndrome (GIS) and bleeding, two severe gastrointestinal side effects observed during anticancer therapy. Based on this evidence, the Authors propose the depletion of free heme by increasing HO-1 activity and Hx sequestration as a potential therapeutic approach to ameliorate the side effects of common antitumoral therapies (Seika et al.). In this scenario, although the effects on cancer prevention of many antioxidant compounds and food supplements are debated (4, 5), novel therapeutic approaches could be exploited targeting antioxidant heme proteins to ameliorate the side effects of anti-cancer therapies.

Free heme may also exacerbate the inflammatory response during bacterial or viral infections with simultaneous intravascular hemolysis. Thus, the antioxidant effects of bilirubin can be identified as a potential therapeutic tool in a context of high oxidative stress resulting from endotoxemia. Dorresteijn et al. propose that hyperbilirubinemia induced by atazanavir can potentiate the antioxidant capacity and restrain the vascular effects, causing a significant decrease in arterial pressure and preventing vascular hyporeactivity in human systemic inflammation elicited by experimental endotoxemia. These results confirm that a physiologic oxidative state can mitigate a detrimental inflammatory response and this could be beneficial both in cardiovascular diseases, by preventing the progression of vascular dysfunctions, and also in septic patients. As suggested by the Dorresteijn et al., inflammatory and oxidative mechanisms could provide optimal therapeutic targets for sepsis resolution, as the eradication of invading pathogens.

In addition to the proinflammatory effect of free heme, heme-containing enzymes may also play a pivotal role in modulating inflammation and oxidative stress, representing potential therapeutic targets for the treatment of various inflammatory/immune-related diseases. In a murine model of low-dose endotoxemia, Mannarino et al. describe a significant reduction of circulating endothelial progenitor cells (EPCs) caused by the chronic inflammatory condition. Interestingly, the administration of l-kynurenine (l-kyn), the main metabolite of the heme-containing enzyme Indoleamine 2,3-dioxygenase 1 (IDO1), reverted EPC decrease. Accordingly, in patients affected by low-grade inflammation, high level of systemic l-kyn directly correlated to the presence of protective EPCs, suggesting a relationship between the immunomodulatory properties of l-kyn and the number of circulating EPCs. This study also highlights the relevant function of the heme-containing enzyme IDO1 in the control of the pro-inflammatory response in several chronic inflammatory conditions (68). Intriguingly, many anti-inflammatory effects exerted by IDO1 in various pathological settings rely on the agonistic activity of its major metabolic product l-kyn on the Aryl Hydrocarbon Receptor (AhR) (911). Similarly, both bilirubin and biliverdin can bind the same intracellular receptor and trigger modulatory pathways in different immune cells during innate and adaptive immune responses (1215). Overall, metabolites generated by different heme-containing enzymes, such as IDO1 or HO-1, could converge into a common mechanism that controls the inflammatory response, thus pinpointing novel potential therapeutic targets for a translational perspective. Furthermore, the heme protein IDO1 represents an appealing target by virtue of its dual nature responsible for its enzymatic and signaling activity. In many tumors, IDO1 represents a key mechanism of acquired immune tolerance in the tumor microenvironment (TME). The IDO1 catalytic activity, by depleting tryptophan and generating immunoregulatory kynurenines, hinders an effective anti-tumor immune response (1618). Therefore, the inhibition of the catalytic activity of IDO1 represented so far a promising and rational therapeutic strategy for the development of effective antitumoral therapies. Several IDO1 inhibitors reached the clinical trials and epacadostat advanced to the large phase 3 trial ECHO-301/KEYNOTE-252 where its association with pembrolizumab (anti-programmed cell death-1/PD-1 antibody) was evaluated. Unfortunately, while a large piece of pre-clinical evidence supported the use of IDO1 inhibition in the context of immunotherapy in solid tumors (19, 20), the treatment of melanoma patients with the association pembrolizumab/epacadostat failed to improve progression-free survival compared to the monotherapy with pembrolizumab (21). The reasons for the trial failure are various (22) and require a better understanding of IDO1 biology, since IDO1 pathway remains a relevant target in cancer immunotherapy. In the current topic, Panfili et al. describe a further on-target activity of epacadostat demonstrating that, besides the catalytic inhibition of IDO1, it could promote the interaction of IDO1 with molecular partners that mediate the signaling function of IDO1, conferring an immunosuppressive phenotype on plasmacytoid dendritic cells. The role of the signaling activity of IDO1 protein has been recently demonstrated in a murine model of melanoma where it incites the progression of the tumor independently of the catalytic function of IDO1 (23). The dual function of IDO1 relies on different conformations of the protein (i.e., the apo- and holo-IDO1) that are dependent on the intracellular heme availability. Thus, heme could affect the dynamic balance between apo- and holo-IDO1 and thus promote the shift towards the catalytic activity of IDO1.

Overall, the intricate interplay between heme proteins, inflammation, and immune regulation highlights the multifaceted roles of these molecules beyond their traditional functions. This Topic underlines how free heme and heme-containing proteins have emerged as critical modulators of inflammatory responses and immune cell functions. Understanding their mechanisms of action and exploring their therapeutic potential may open up new avenues for developing targeted treatments for inflammatory diseases and immune-related disorders.

Author contributions

CV: Writing – original draft, Writing – review & editing. BE: Writing – review & editing. CO: Writing – review & editing.

Funding

This work was supported by the Italian Ministry of Education, University, and Research (PRIN 20173EAZ2Z; to C.V.) and performed within the activities of the MMMAINSTREAM and INFLANOTCH projects, granted by the University of Perugia (Fondo Ricerca di Ateneo, edizione 2021).

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.

The authors declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

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. Ryter SW. Heme oxygenase-1: an anti-inflammatory effector in cardiovascular, lung, and related metabolic disorders. Antioxidants (Basel). (2022) 11(3):555. doi: 10.3390/antiox11030555

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Nitti M, Furfaro AL, Mann GE. Heme oxygenase dependent bilirubin generation in vascular cells: A role in preventing endothelial dysfunction in local tissue microenvironment? Front Physiol (2020) 11:23. doi: 10.3389/fphys.2020.00023

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Janciauskiene S, Vijayan V, Immenschuh S. TLR4 signaling by heme and the role of heme-binding blood proteins. Front Immunol (2020) 11:1964. doi: 10.3389/fimmu.2020.01964

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Fortmann SP, Burda BU, Senger CA, Lin JS, Whitlock EP. Vitamin and mineral supplements in the primary prevention of cardiovascular disease and cancer: an updated systematic evidence review for the U.S. Preventive Services Task Force. Ann Internal Med (2013) 159(12):824–34. doi: 10.7326/0003-4819-159-12-201312170-00729

CrossRef Full Text | Google Scholar

5. Sunjic SB, Zarkovic N. Editorial on anticancer antioxidants. Antioxidants (2021) 10(11):1782. doi: 10.3390/antiox10111782

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Jung ID, Lee MG, Chang JH, Lee JS, Jeong YI, Lee CM, et al. Blockade of indoleamine 2,3-dioxygenase protects mice against lipopolysaccharide-induced endotoxin shock. J Immunol (2009) 182(5):3146–54. doi: 10.4049/jimmunol.0803104

PubMed Abstract | CrossRef Full Text | Google Scholar

7. ROmani L, Fallarino F, De Luca A, Montagnoli C, D’Angelo C, Zelante T, et al. Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature (2008) 451(7175):211–5. doi: 10.1038/nature06471

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Baumgartner R, Berg M, Matic L, Polyzos KP, Forteza MJ, Hjorth SA, et al. Evidence that a deviation in the kynurenine pathway aggravates atherosclerotic disease in humans. J Intern Med (2021) 289(1):53–68. doi: 10.1111/joim.13142

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Mondanelli G, Coletti A, Greco FA, Pallotta MT, Orabona C, Iacono A, et al. Positive allosteric modulation of indoleamine 2,3-dioxygenase 1 restrains neuroinflammation. Proc Natl Acad Sci U S A. (2020) 117(7):3848–57. doi: 10.1073/pnas.1918215117

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Matino D, Afraz S, Zhao G, Tieu P, Gargaro M, Fallarino F, et al. Tolerance to FVIII: role of the immune metabolic enzymes indoleamine 2,3 Dyoxigenase-1 and heme Oxygenase-1. Front Immunol (2020) 11:620. doi: 10.3389/fimmu.2020.00620

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature (2014) 511(7508):184–90. doi: 10.1038/nature13323

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Longhi MS, Vuerich M, Kalbasi A, Kenison JE, Yeste A, Csizmadia E, et al. Bilirubin suppresses Th17 immunity in colitis by upregulating CD39. JCI Insight (2017) 2(9):e92791. doi: 10.1172/jci.insight.92791

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Gazzin S, Vitek L, Watchko J, Shapiro SM, Tiribelli C. A novel perspective on the biology of bilirubin in health and disease. Trends Mol Med (2016) 22:758–68. doi: 10.1016/j.molmed.2016.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Vítek L. Bilirubin as a signaling molecule. Med Res Rev (2020) 40(4):1335–51. doi: 10.1002/med.21660

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Solvay M, Holfelder P, Klaessens S, Pilotte L, Stroobant V, Lamy J, et al. Tryptophan depletion sensitizes the AHR pathway by increasing AHR expression and GCN2/LAT1-mediated kynurenine uptake, and potentiates induction of regulatory T lymphocytes. J Immunother Cancer. (2023) 11(6):e006728. doi: 10.1136/jitc-2023-006728

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Uyttenhove C, Pilotte L, Théate I, Stroobant V, Colau D, Parmentier N, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med (2003) 9(10):1269–74. doi: 10.1038/nm934

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Meireson A, Devos M, Brochez L. IDO expression in cancer: different compartment, different functionality? Front Immunol (2020) 11:531491. doi: 10.3389/fimmu.2020.531491

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med (1999) 189(9):1363–72. doi: 10.1084/jem.189.9.1363

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, Tobias A, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res (2014) 20(20):5290–301. doi: 10.1158/1078-0432.CCR-14-0514

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Spranger S, Koblish HK, Horton B, Scherle PA, Newton R, Gajewski TF. Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment. J Immunother Cancer. (2014) 2:3. doi: 10.1186/2051-1426-2-3

PubMed Abstract | CrossRef Full Text | Google Scholar

21. G.v. Long R, Hamid O, Gajewski TF, Caglevic C, Dalle S, Arance A, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol (2019) 20:30274–8. doi: 10.1016/S1470-2045(19)30274-8

CrossRef Full Text | Google Scholar

22. Van den Eynde B, Baren N, Baurain J-F. Is there a clinical future for IDO1 inhibitors after the failure of epacadostat in melanoma? Ann Rev Cancer Biol (2020) 4(1):241–56. doi: 10.1146/annurev-cancerbio-030419-033635

CrossRef Full Text | Google Scholar

23. Orecchini E, Belladonna ML, Pallotta MT, Volpi C, Zizi L, Panfili E, et al. The signaling function of IDO1 incites the Malignant progression of mouse B16 melanoma. Oncoimmunology (2023) 12(1):2170095. doi: 10.1080/2162402X.2023.2170095

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: heme proteins, indoleamine 2,3-dioxygenase, heme oxygenases, immune response, heme

Citation: Volpi C, Van den Eynde BJ and Orabona C (2023) Editorial: Heme proteins: key players in the regulation of immune responses. Front. Immunol. 14:1263384. doi: 10.3389/fimmu.2023.1263384

Received: 19 July 2023; Accepted: 21 July 2023;
Published: 09 August 2023.

Edited and Reviewed by:

Stephen Robert Daley, Queensland University of Technology, Australia

Copyright © 2023 Volpi, Van den Eynde and Orabona. 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: Claudia Volpi, claudia.volpi@unipg.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.