Abstract
Macrophage efferocytosis prevents apoptotic cell (AC) accumulation and triggers inflammation-resolution pathways. The mechanisms linking efferocytosis to resolution often involve changes in macrophage metabolism, but many gaps remain in our understanding of these processes. We now report that efferocytosis triggers an indoleamine 2,3-dioxygenase-1 (IDO1)-dependent tryptophan (Trp) metabolism pathway that promotes several key resolution processes, including the induction of pro-resolving proteins, such interleukin-10, and further enhancement of efferocytosis. The process begins with upregulation of Trp transport and metabolism, and it involves subsequent activation of the aryl hydrocarbon receptor (AhR) by the Trp metabolite kynurenine (Kyn). Through these mechanisms, macrophage IDO1 and AhR contribute to a proper resolution response in several different mouse models of efferocytosis-dependent tissue repair, notably during atherosclerosis regression induced by plasma low-density lipoprotein (LDL) lowering. These findings reveal an integrated metabolism programme in macrophages that links efferocytosis to resolution, with possible therapeutic implications for non-resolving chronic inflammatory diseases, notably atherosclerosis.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Similar content being viewed by others
Data availability
All data supporting the study are available in the manuscript and supplementary information. Source data are provided with this paper.
Change history
16 September 2024
A Correction to this paper has been published: https://doi.org/10.1038/s42255-024-01142-4
References
Panigrahy, D., Gilligan, M. M., Serhan, C. N. & Kashfi, K. Resolution of inflammation: an organizing principle in biology and medicine. Pharmacol. Ther. 227, 107879 (2021).
Nathan, C. & Ding, A. Nonresolving inflammation. Cell 140, 871–882 (2010).
Hochreiter-Hufford, A. & Ravichandran, K. S. Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb. Perspect. Biol. 5, a008748 (2013).
Morioka, S., Maueroder, C. & Ravichandran, K. S. Living on the edge: efferocytosis at the interface of homeostasis and pathology. Immunity 50, 1149–1162 (2019).
Doran, A. C., Yurdagul, A. Jr. & Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 20, 254–267 (2020).
Henson, P. M. Cell removal: efferocytosis. Annu. Rev. Cell Dev. Biol. 33, 127–144 (2017).
Linton, M. F. et al. Macrophage apoptosis and efferocytosis in the pathogenesis of atherosclerosis. Circ. J. 80, 2259–2268 (2016).
Kojima, Y., Weissman, I. L. & Leeper, N. J. The role of efferocytosis in atherosclerosis. Circulation 135, 476–489 (2017).
A-Gonzalez, N. et al. Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR. Immunity 31, 245–258 (2009).
Yurdagul, A. Jr. et al. Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab. 31, 518–533 (2020).
Yurdagul, A. Jr. et al. ODC (ornithine decarboxylase)-dependent putrescine synthesis maintains MerTK (MER tyrosine-protein kinase) expression to drive resolution. Arterioscler. Thromb. Vasc. Biol. 41, e144–e159 (2021).
Gerlach, B. D. et al. Efferocytosis induces macrophage proliferation to help resolve tissue injury. Cell Metab. 33, 2445–2463 (2021).
Ampomah, P. B. et al. Macrophages use apoptotic cell-derived methionine and DNMT3A during efferocytosis to promote tissue resolution. Nat. Metab. 4, 444–457 (2022).
Zhang, S. et al. Efferocytosis fuels requirements of fatty acid oxidation and the electron transport chain to polarize macrophages for tissue repair. Cell Metab. 29, 443–456 (2018).
Moffett, J. R. & Namboodiri, M. A. Tryptophan and the immune response. Immunol. Cell Biol. 81, 247–265 (2003).
Pillai, S. M. & Meredith, D. SLC36A4 (hPAT4) is a high affinity amino acid transporter when expressed in Xenopus laevis oocytes. J. Biol. Chem. 286, 2455–2460 (2011).
Shang, P. et al. The amino acid transporter SLC36A4 regulates the amino acid pool in retinal pigmented epithelial cells and mediates the mechanistic target of rapamycin, complex 1 signaling. Aging Cell 16, 349–359 (2017).
Park, S. Y. et al. Rapid cell corpse clearance by stabilin-2, a membrane phosphatidylserine receptor. Cell Death Differ. 15, 192–201 (2008).
Flannagan, R. S., Canton, J., Furuya, W., Glogauer, M. & Grinstein, S. The phosphatidylserine receptor TIM4 utilizes integrins as coreceptors to effect phagocytosis. Mol. Biol. Cell 25, 1511–1522 (2014).
Williamson, A. P. & Vale, R. D. Spatial control of Draper receptor signaling initiates apoptotic cell engulfment. J. Cell Biol. 217, 3977–3992 (2018).
Moon, H. et al. Crbn modulates calcium influx by regulating Orai1 during efferocytosis. Nat. Commun. 11, 5489 (2020).
Pallotta, M. T. et al. Indoleamine 2,3-dioxygenase 1 (IDO1): an up-to-date overview of an eclectic immunoregulatory enzyme. FEBS J. 289, 6099–6118 (2022).
Yoshimori, T., Yamamoto, A., Moriyama, Y., Futai, M. & Tashiro, Y. Bafilomycin A1, a specific inhibitor of vacuolar-type H+-ATPase, inhibits acidification and protein degradation in lysosomes of cultured cells. J. Biol. Chem. 266, 17707–17712 (1991).
Park, D. et al. Continued clearance of apoptotic cells critically depends on the phagocyte Ucp2 protein. Nature 477, 220–224 (2011).
Koblish, H. K. et al. Hydroxyamidine inhibitors of indoleamine-2,3-dioxygenase potently suppress systemic tryptophan catabolism and the growth of IDO-expressing tumors. Mol. Cancer Ther. 9, 489–498 (2010).
Liu, X. et al. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood 115, 3520–3530 (2010).
Ravichandran, K. S. & Lorenz, U. Engulfment of apoptotic cells: signals for a good meal. Nat. Rev. Immunol. 7, 964–974 (2007).
Proto, J. D. et al. Regulatory T cells promote macrophage efferocytosis during inflammation resolution. Immunity 49, 666–677.e666 (2018).
Vallés, A. M., Beuvin, M. & Boyer, B. Activation of Rac1 by paxillin–Crk–DOCK180 signaling complex is antagonized by Rap1 in migrating NBT-II cells. J. Biol. Chem. 279, 44490–44496 (2004).
Fadok, V. A. et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-β, PGE2, and PAF. J. Clin. Invest. 101, 890–898 (1998).
Cervenka, I., Agudelo, L. Z. & Ruas, J. L. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science 357, eaaf9794 (2017).
Navarro-Xavier, R. A. et al. A new strategy for the identification of novel molecules with targeted proresolution of inflammation properties. J. Immunol. 184, 1516–1525 (2010).
Hezaveh, K. et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 55, 324–340 (2022).
Campesato, L. F. et al. Blockade of the AHR restricts a Treg–macrophage suppressive axis induced by l-kynurenine. Nat. Commun. 11, 4011 (2020).
Nguyen, N. T. et al. Aryl hydrocarbon receptor and kynurenine: recent advances in autoimmune disease research. Front Immunol. 5, 551 (2014).
Mezrich, J. D. et al. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 185, 3190–3198 (2010).
Opitz, C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011).
Tsuji, N. et al. The activation mechanism of the aryl hydrocarbon receptor (AhR) by molecular chaperone HSP90. FEBS Open Bio. 4, 796–803 (2014).
Kim, S. H. et al. Novel compound 2-methyl-2H-pyrazole-3-carboxylic acid (2-methyl-4-o-tolylazo-phenyl)-amide (CH-223191) prevents 2,3,7,8-TCDD-induced toxicity by antagonizing the aryl hydrocarbon receptor. Mol. Pharmacol. 69, 1871–1878 (2006).
Vogel, C. F., Goth, S. R., Dong, B., Pessah, I. N. & Matsumura, F. Aryl hydrocarbon receptor signaling mediates expression of indoleamine 2,3-dioxygenase. Biochem. Biophys. Res. Commun. 375, 331–335 (2008).
Hoffman, E. C. et al. Cloning of a factor required for activity of the Ah (dioxin) receptor. Science 252, 954–958 (1991).
Park, S. Y. et al. Stabilin-1 mediates phosphatidylserine-dependent clearance of cell corpses in alternatively activated macrophages. J. Cell Sci. 122, 3365–3373 (2009).
Favata, M. F. et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J. Biol. Chem. 273, 18623–18632 (1998).
Cox, M. B. & Miller, C. A. III Cooperation of heat shock protein 90 and p23 in aryl hydrocarbon receptor signaling. Cell Stress Chaperones 9, 4–20 (2004).
Pappas, B. et al. p23 protects the human aryl hydrocarbon receptor from degradation via a heat shock protein 90-independent mechanism. Biochem. Pharmacol. 152, 34–44 (2018).
Raber, L. et al. Effect of alirocumab added to high-intensity statin therapy on coronary atherosclerosis in patients with acute myocardial infarction: the PACMAN-AMI Randomized Clinical Trial. JAMA 327, 1771–1781 (2022).
Nicholls, S. J. et al. Effect of evolocumab on coronary plaque phenotype and burden in statin-treated patients following myocardial infarction. JACC Cardiovasc. Imaging 15, 1308–1321 (2022).
Sharma, M. et al. Regulatory T cells license macrophage pro-resolving functions during atherosclerosis regression. Circ. Res. 127, 335–353 (2020).
Shinde, R. et al. Apoptotic cell-induced AhR activity is required for immunological tolerance and suppression of systemic lupus erythematosus in mice and humans. Nat. Immunol. 19, 571–582 (2018).
Koenis, D. S., de Matteis, R., Rajeeve, V., Cutillas, P. & Dalli, J. Efferocyte-derived MCTRs metabolically prime macrophages for continual efferocytosis via Rac1-mediated activation of glycolysis. Adv. Sci. 11, e2304690 (2023).
Mellor, A. L. et al. Cutting edge: CpG oligonucleotides induce splenic CD19+ dendritic cells to acquire potent indoleamine 2,3-dioxygenase-dependent T cell regulatory functions via IFN type 1 signaling. J. Immunol. 175, 5601–5605 (2005).
Ravishankar, B. et al. Tolerance to apoptotic cells is regulated by indoleamine 2,3-dioxygenase. Proc. Natl Acad. Sci. USA 109, 3909–3914 (2012).
Ravishankar, B. et al. The amino acid sensor GCN2 inhibits inflammatory responses to apoptotic cells promoting tolerance and suppressing systemic autoimmunity. Proc. Natl Acad. Sci. USA 112, 10774–10779 (2015).
Liu, Y. et al. Tumor-repopulating cells induce PD-1 expression in CD8+T cells by transferring kynurenine and AhR activation. Cancer Cell 33, 480–494 (2018).
Tan, Z., Huang, M., Puga, A. & Xia, Y. A critical role for MAP kinases in the control of Ah receptor complex activity. Toxicol. Sci. 82, 80–87 (2004).
Metghalchi, S. et al. Indoleamine 2,3-dioxygenase fine-tunes immune homeostasis in atherosclerosis and colitis through repression of interleukin-10 production. Cell Metab. 22, 460–471 (2015).
Cole, J. E. et al. Indoleamine 2,3-dioxygenase-1 is protective in atherosclerosis and its metabolites provide new opportunities for drug development. Proc. Natl Acad. Sci. USA 112, 13033–13038 (2015).
Baumgartner, R. et al. Evidence that a deviation in the kynurenine pathway aggravates atherosclerotic disease in humans. J. Intern Med 289, 53–68 (2021).
Yang, L., Zheng, C., Xia, Y. F., Dai, Y. & Wei, Z. F. 3,3′-diindolylmethane enhances macrophage efferocytosis and subsequently relieves visceral pain via the AhR/Nrf2/Arg-1-mediated arginine metabolism pathway. Phytomedicine 116, 154874 (2023).
Tian, L. X. et al. Cytochrome P450 1A1 enhances arginase-1 expression, which reduces LPS-induced mouse peritonitis by targeting JAK1/STAT6. Cell Immunol. 349, 104047 (2020).
Zhu, K. et al. Aryl hydrocarbon receptor pathway: role, regulation and intervention in atherosclerosis therapy. Mol. Med. Rep. 20, 4763–4773 (2019).
Wu, D. et al. Activation of aryl hydrocarbon receptor induces vascular inflammation and promotes atherosclerosis in apolipoprotein E−/− mice. Arterioscler. Thromb. Vasc. Biol. 31, 1260–1267 (2011).
Mehrotra, P. & Ravichandran, K. S. Drugging the efferocytosis process: concepts and opportunities. Nat. Rev. Drug Discov. 21, 601–620 (2022).
Morioka, S. et al. Chimeric efferocytic receptors improve apoptotic cell clearance and alleviate inflammation. Cell 185, 4887–4903 (2022).
Kojima, Y. et al. CD47-blocking antibodies restore phagocytosis and prevent atherosclerosis. Nature 536, 86–90 (2016).
Flores, A. M. et al. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. Nat. Nanotechnol. 15, 154–161 (2020).
Schrodt, M. V. et al. Efferocytosis of viable versus heat-inactivated MSC induces human monocytes to distinct immunosuppressive phenotypes. Stem Cell Res. Ther. 14, 206 (2023).
Li, Q. et al. Associations between plasma tryptophan and indole-3-propionic acid levels and mortality in patients with coronary artery disease. Am. J. Clin. Nutr. 116, 1070–1077 (2022).
Yu, E. et al. Increases in plasma tryptophan are inversely associated with incident cardiovascular disease in the Prevención con Dieta Mediterránea (PREDIMED) study. J. Nutr. 147, 314–322 (2017).
Murr, C. et al. Low serum tryptophan predicts higher mortality in cardiovascular disease. Eur. J. Clin. Invest 45, 247–254 (2015).
Qiu, H. et al. Efferocytosis: an accomplice of cancer immune escape. Biomed. Pharmacother. 167, 115540 (2023).
Werfel, T. A. et al. Treatment-induced tumor cell apoptosis and secondary necrosis drive tumor progression in the residual tumor microenvironment through MerTK and IDO1. Cancer Res. 79, 171–182 (2019).
Wetzel, L. A., Hurtado, M., MacDowell Kaswan, Z. A., McCusker, R. H. & Steelman, A. J. Deletion of indoleamine 2,3 dioxygenase (Ido)1 but not Ido2 exacerbates disease symptoms of MOG(35-55)-induced experimental autoimmune encephalomyelitis. Brain Behav. Immun. Health 7, 100116 (2020).
Wang, Y. et al. Mitochondrial fission promotes the continued clearance of apoptotic cells by macrophages. Cell 171, 331–345 (2017).
Zhu, W. et al. Quantitative profiling of tryptophan metabolites in serum, urine, and cell culture supernatants by liquid chromatography-tandem mass spectrometry. Anal. Bioanal. Chem. 401, 3249–3261 (2011).
Schilperoort, M., Ngai, D., Katerelos, M., Power, D. A. & Tabas, I. PFKFB2-mediated glycolysis promotes lactate-driven continual efferocytosis by macrophages. Nat. Metab. 5, 431–444 (2023).
Ngai, D., Schilperoort, M. & Tabas, I. Efferocytosis-induced lactate enables the proliferation of pro-resolving macrophages to mediate tissue repair. Nat. Metab. 5, 2206–2219 (2023).
Acknowledgements
We thank R. Ramakrishnan (Columbia University) for his guidance on statistical analysis of the data in this study. We thank B. Gerlach for his invaluable initial discussions, which greatly contributed to the development of this research. This work was supported by NIH/NHLBI grant nos. R35-HL145228 and P01-HL087123 (to I.T.) and R01-HL159012 (to J.S. and I.T.). D.N. was supported by American Heart Association postdoctoral award no. 24POST1192241. Immunofluorescence imaging experiments were conducted in the Columbia Center for Translational Immunology Core Facility, funded by NIH grant nos. P30CA013696, S10RR027050 and S10OD020056. Flow cytometry experiments were conducted using the Herbert Irving Comprehensive Cancer Center Flow Cytometry Shared Resources, funded in part through NIH grant no. P30CA013696. Samples for histological analysis were prepared in the Molecular Pathology Shared Resource of the Herbert Irving Comprehensive Cancer Center at Columbia University, supported by NIH grant no. P30CA013696. The confocal microscopy work in this study was conducted in the Confocal and Specialized Microscopy Shared Resource of the Herbert Irving Comprehensive Cancer Center at Columbia University, supported by NIH grants nos. P30CA013696 and S10RR025686. This work was supported in part by the Proteomics & Metabolomics Core at Moffitt Cancer Center and funded as part of an NCI-designated Comprehensive Cancer Center (P30 CA076292).
Author information
Authors and Affiliations
Contributions
S.R.S. and I.T. conceived the project. P.B.A., D.N., X.W., J.S. and R.H.M. provided additional intellectual input in the development of the project. L.N.F.D. conducted the LC–MS analyses under the guidance of J.M.K. G.K. and Y.X. helped with the mouse atherosclerosis experiments. S.R.S. and I.T. wrote the manuscript and the other co-authors provided comments and revisions.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Metabolism thanks Derek W. Gilroy, Laurent Yvan-Charvet and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Alfredo Giménez-Cassina in collaboration with the Nature Metabolism team.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Related to Fig. 1: Macrophage efferocytosis drives Trp metabolism.
a, Tryptophan and kynurenine content expressed as pmol/μg cell protein in BMDMs incubated ± ACs from the experiment in Fig. 1a (n = 6 biological replicates/group). b,c, Additional metabolite values from BMDMs incubated ± ACs from the experiment in Fig. 1a (n = 6 biological replicates/group). Data are mean ± SEM, and significance was determined by two-tailed Student’s t-test.
Extended Data Fig. 2 Related to Fig. 1: Examples of extracted ion chromatograms.
a, Reversed phase separation and mass spectrometry detection of 12 neat tryptophan metabolite standards (10 ng) individually analyzed. Each trace shows ion signal at a given mass-to-charge ratio (m/z) as the compounds elute from the Atlantis T3 reversed phase column. b, Reversed phase separation on an Atlantis T3 reversed phase column and mass spectrometry detection of tryptophan metabolites from an AC- wild type sample. Each trace shows ion signal at a given mass-to-charge ratio (m/z) with a mass tolerance of 5 ppm. For both sets of chromatograms, data are shown for: (i) 2−picolinic acid), (ii) nicotinic acid, (iii) nicotinamide, (iv) quinolinic acid, (v) nicotinamide adenine dinucleotide (NAD)+, (vi) serotonin, (vii) DL-kynurenine, (viii) N-formylkynurenine, (ix) L-tryptophan, (x) tryptamine, (xi) kynurenic acid, and (xii) anthranilic acid. The normalization level (NL) indicates the intensity of the base peak for each spectrum. In b, although more background was observed in the extracted ion chromatogram for anthranilic acid, the anthranilic acid signal is easily differentiated from the background.
Extended Data Fig. 3 Related to Fig. 1: Absolute quantitation of kynurenine and tryptophan.
a-d, Quantitation using a known amount of isotope-labeled standards. a,b, Examples of extracted ion chromatograms showing reversed phase separation and mass spectrometry detection for kynurenine (10 ng) and D4-kynurenine (50 ng); and tryptophan (10 ng) and 13C11-tryptophan (50 ng). Each trace shows the ion signal at a given mass-to-charge ratio (m/z) as the compounds elute from an Atlantis T3 reversed phase column. c,d, Positive ion mass spectra of kynurenine and D4-kynurenine; and tryptophan and 13C11-tryptophan. e-h, Quantitation of kynurenine and tryptophan in an AC− wild type sample. e,f, Examples of extracted ion chromatograms showing reversed phase separation and mass spectrometry detection for endogenous kynurenine and spiked D4-kynurenine (50 ng); and endogenous tryptophan and spiked 13C11-tryptophan (50 ng). Each trace shows the ion signal at a given mass-to-charge ratio (m/z) as the compounds elute from an Atlantis T3 reversed phase column. g,h, Positive ion mass spectra of endogenous kynurenine and D4-kynurenine; and endogenous tryptophan and 13C11-tryptophan. The normalization level (NL) indicates the intensity of the base peak for each spectrum.
Extended Data Fig. 4 Related to Fig. 1: The role of SLC36A4 in Trp metabolism in efferocytosing macrophages.
a, Immunofluorescence microscopy of SLC36A4 (green) and LAMP-1 (red) in Scr- or siSlc36a4-transfected BMDMs incubated 45 mins with PKH26-labelled ACs (pseudocolored white); DAPI (blue) nuclear stain. Scale bar, 50 μm. Another set of cells was assayed for Slc36a4 mRNA and immunoblotted for SLC36A4 (n = 3 biological replicates/group). b, Immunofluorescence microscopy of SLC36A4 (green) in BMDMs incubated with PKH26-labelled ACs (red) for 45 min. Scale bar, 50 μm. White arrows, engulfed ACs; blue arrows, unengulfed ACs. c, Representative image of Scr-transfected macrophages not incubated with ACs and then stained for SLC36A4 (green) and DAPI (blue); note low expression of SLC36A4 compared with AC+ macrophages in Fig. 1b. Image is representative of 3 biological triplicates. Scale bar, 50 μm. d, BMDMs were incubated ± apoptotic Jurkat cells (apJCs) or apoptotic macrophages (apMϕs), chased for 3 h, and assayed for Slc36a4 (n = 6 biological replicates/group). e, BMDMs were incubated ± ACs or PS-beads for 1 h, chased for 3 h, and immunoblotted for SLC36A4. f, BMDMs pre-treated with ± 20 µM MG132 were incubated ± ACs for 1 h, chased for 3 h, and immunoblotted for SLC36A4. g, Scr- or siSlc36a4-transfected BMDMs were incubated with PKH26-labelled ACs (red) and quantified for the percentage of PKH26+ macrophages (arrows) of total macrophages. Scale bar, 50 μm (n = 5 biological replicates/group). h, Tryptophan and kynurenine in Scr- or siSlc36a4-transfected BMDMs incubated with ACs (see Fig. 1d; n = 6 biological replicates/group). i, BMDMs were incubated ± apoptotic macrophages for 1 h and assayed for Ido1 (n = 3 biological replicates/group). j, Scr- or siSLC36A4-transfected HMDMs were for SLC36A4 (n = 3 biological replicates/group). All mRNA data are expressed relative to the indicated control groups. Data are mean ± SEM, and significance was determined by two-tailed Student’s t-test or one-way ANOVA with Fisher’s LSD post-hoc analysis.
Extended Data Fig. 5 Related to Figs. 2 and 3: Additional data on the roles of SLC36A4 and IDO1 in resolution.
a, Control or IDO1-KO BMDMs incubated with PKH26-labelled ACs were quantified for percent PKH26+ macrophages (arrows). Scale bar, 50 μm (n = 6 biological replicates/group). b, Scr- or siIdo1-transfected BMDMs incubated with pHrodo-Red-labelled ACs were quantified for percent pHrodo-Red+ macrophages of total macrophages by flow cytometry (n = 3 biological replicates/group); immunoblotted for IDO1 (n = 3 samples/group); and assayed for Ido1 (n = 6 biological replicates/group). c, Flow cytometry contour plots for the experiment in Fig. 2b. d, BMDMs treated with ACs ± epacadostat were assayed for Arg1 and Mcf2 after a 6-h or 2-h chase, respectively (n = 3 biological replicates/group). e,f, Contour plots for the experiments in Fig. 2g, h. g, Scr- or siIDO1-transfected HMDMs were assayed for IDO1 (n = 3 biological replicates/group). h, Scr- or siSlc36a4-transfected BMDMs were incubated ± apoptotic macrophages, chased for 6 h, and assayed for Tgfb1 and Il10 (n = 3 biological replicates/group). i, Scr- or siSlc36a4-transfected BMDMs were pre-treated ± kynurenine and then incubated ± ACs for 1 h, chased for 6 h in Trp-deficient medium, and assayed for Tgfb1 and Il10 mRNA. Right, normal and Trp-depleted media were assayed for Trp by LC−MS/MS (right) (n = 3 biological replicates/group). j, BMDMs pre-treated for 1 h with vehicle or 50 µM Trp were incubated ± ACs, chased for 6 h, and assayed for Tgfb1 and Il10 (n = 3 biological replicates/group). k, Scr- or siSlc36a4-transfected BMDMs were pre-treated ± 50 μM Trp for 1 h, incubated ± ACs. and assayed for Tgfb1 and Il10 (n = 3 biological replicates/group). All mRNA data are expressed relative to the indicated control groups. Data are mean ± SEM, and significance was determined by two-tailed Student’s t-test or one-way ANOVA with Fisher’s LSD post-hoc analysis.
Extended Data Fig. 6 Related to Figs. 4 and 5: Additional in-vivo and in-vitro data on the IDO1-Kyn-AhR pathway.
a-c, The thymi of the mice from Fig. 4g–e were immunostained for IDO1 (red) and Mac2 (green) and quantified for IDO1 MFI in Mac2+ areas (arrows). Also shown are thymus weight, thymus cellularity, and F4/80+ macrophages/thymus (n = 8 mice/group). d, Control and Ido1−/− BMDMs (top 2 graphs), or Scr- and siSlc364-transfected BMDMs (bottom graph), were pre-treated for 1 h ± Kyn and incubated ± ACs. After a 6-h chase, the cells were assayed for Cyp1a1 and Cyp1b1 (n = 3 biological replicates/group). e, BMDMs were incubated ± apoptotic macrophages and assayed for Cyp1a1. f, Scr- or siSLC36A4-transfected HMDMs were incubated ± ACs, chased for 3 h, and assayed for CYP1A1 and CYP1B1 (n = 3 biological replicates/group). g, Scr- or siAhr-transfected BMDMs were incubated with DiD-labelled ACs and quantified for percent DiD+ macrophages (arrows). Scale bar, 50 μm (n = 4 biological replicates/group). h, BMDMs pre-treated ± CH223191 were assayed for continuing efferocytosis as in Fig. 2a. Arrows, PKH26+PKH67+ macrophages. Scale bar, 50 μm (n = 3 biological replicates/group). i, As in panel h, but one of the cohorts was also treated with cytochalasin D before the second round of efferocytosis. Arrows, PKH26+PKH67+ macrophages. Scale bar, 50 μm (n = 3 biological replicates/group). j, Scr- or siAhr-transfected BMDMs were assayed for Ahr (n = 3 biological replicates/group) and immunoblotted for AhR protein (n = 3 samples/group). k, Scr- or siAHR-transfected HMDMs were assayed for AHR (n = 3 biological replicates/group). l, Scr- or siArnt-transfected BMDMs were assayed for Arnt (n = 3 biological replicates/group). All mRNA data are expressed relative to the indicated control groups. Data are mean ± SEM, and significance was determined by two-tailed Student’s t-test or one-way ANOVA with Fisher’s LSD post-hoc analysis.
Extended Data Fig. 7 Related to Figs. 6 and 7: Additional in-vivo and in-vitro data on the role of AhR in efferocytosis-induced resolution.
a, BMDMs were incubated for 1 h with 100 μM Kyn alone or with control (non-PS) or PS beads ± Kyn and then assayed for Cyp1a1 and Ido1 after a 3-h chase (n = 4 samples/group). b, BMDMs incubated with ACs, PS-beads (PS), Kyn, or PS-beads + Kyn were assayed for Tgfb1 after a 6-h chase (n = 4 samples/group). c, BMDMs incubated with 50 μM Trp, PS-beads (PS), Trp and PS-beads, or 100 μM Kyn and PS-beads for 1 h were assayed for Cyp1a1 and Il10 after a 3-h or 6-h chase, respectively (n = 3 biological replicates/group). d, BMDMs pre-treated ± U0126 were incubated with ACs for 1 h and then immunoblotted for AhR and ARNT after a 3-h chase (n = 3 samples/group). e, BMDMs treated with PS-beads (PS) and Kyn ± U0126 for 1 h were assayed for Tgfb1 or Ido1 after a 3-h chase or 6-h chase, respectively (n = 3 biological replicates/group). f, BMDMs incubated ± ACs ± CH223191 for 1 h were assayed Hsp90 or Xap2 after a 3-h chase (n = 3 biological replicates/group). g, Scr-, siHsp90-, or siXap2-transfected BMDMs were incubated with PKH26-labelled ACs and quantified for percent PKH26+ macrophages (arrows). Scale bar, 50 μm (n = 3 biological replicates/group). h, Proposed pathway (created using BioRender.com): Trp form an efferocytosed AC (AC1) is transported into the macrophage by SLC36A4 and then converted to Kyn by IDO1. Kyn and activated ERK induce Hsp90 and Xap2, leading to AhR-ARNT-mediated transcription of Tgfb1, Il10, and Ido1 and Rac1-mediated AC2 internalization (continuing efferocytosis). i-k, The thymi of the mice from Fig. 7 were immunostained for AhR (red) and Mac2 (green) and quantified for AhR MFI in Mac2+ areas (arrows). Also shown are thymus weight and F4/80+ macrophages/thymus (n = 8 mice/group). All mRNA data are expressed relative to the indicated control groups. Data are mean ± SEM, and significance was determined by two-tailed Student’s t-test or one-way ANOVA with Fisher’s LSD post-hoc analysis.
Extended Data Fig. 8 Related to Fig. 8: Systemic and lesional parameters in control and Mϕ-IDO1-iKO BMT Ldlr−/− mice.
Ldlr−/− mice were transplanted with BM from Ido1fl/fl (Control) or Ido1fl/flCx3cr1creERT2+/− (Mϕ-IDO1-iKO) mice and then fed the Western diet for 16 weeks. One cohort from each group was harvested (Baseline), and the rest of the mice were switched to chow diet, injected with HDAd-LDLR virus, and given tamoxifen. After 7 weeks, the mice were harvested (Regression). a-k, Body weight, total plasma cholesterol, fasting blood glucose, complete blood count (n = 9–10 mice/group). WBC, white blood cell; NE, neutrophils; LY, lymphocytes; MO, monocytes; EO, eosinophils; BA, basophils, RBC, red blood cells; PLT, platelets. l, Immunostaining of IDO1 (red) and Mac2 (green) in regressing aortic root lesions, with quantification of IDO1 MFI in Mac2+ and Mac2− areas. Arrows, examples of IDO1-Mac2 co-localization. DAPI was used for nuclear staining. Scale bar, 25 μm (n = 10 mice/group). m, Quantification of lesion area, based on H&E staining of the aortic root lesions (n = 10 mice/group). n, The regressing aortic root lesions of Control and Mϕ-IDO1-iKO groups were immunostained for Mac2 (macrophages; green) and TGF-β1 or IL-10. (red) Arrows, examples of colocalization of Mac2 and TGF-β1 (top) and Mac2 and IL-10 (bottom). DAPI (blue) was used for nuclear staining. Scale bar, 50 μm. o, The total number of Mac2+ cells per lesion section was quantified in regressing aortic root lesions (n = 10 mice/group). The data are expressed as mean ± SEM, and significance was determined by one-way ANOVA with Fisher’s LSD post-hoc analysis for panels a-k and m, and by Student’s t-test for panels l and o.
Supplementary information
Supplementary Tables 1–5
Supplementary Table 1: Macrophage efferocytosis drives tryptophan metabolism. Supplementary Table 2: The average and s.d. of retention time and m/z of tryptophan metabolites using LC–MS. Supplementary Table 3: Macrophage efferocytosis drives tryptophan metabolism in an SLC36A4-dependent manner. Supplementary Table 4: List of antibodies. Supplementary Table 5: Primer sequences and RNAi list.
Source data
Source Data Figs. 1, 5, and 6
Unprocessed western blots for Figs. 1, 5, and 6.
Source Data Extended Data Fig. 4–7
Unprocessed western blots for Extended Data Figs. 4–7.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Sukka, S.R., Ampomah, P.B., Darville, L.N.F. et al. Efferocytosis drives a tryptophan metabolism pathway in macrophages to promote tissue resolution. Nat Metab 6, 1736–1755 (2024). https://doi.org/10.1038/s42255-024-01115-7
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s42255-024-01115-7
This article is cited by
-
Dys-regulated phosphatidylserine externalization as a cell intrinsic immune escape mechanism in cancer
Cell Communication and Signaling (2025)
-
The systemic inflammation response index as risks factor for all-cause and cardiovascular mortality among individuals with respiratory sarcopenia
BMC Pulmonary Medicine (2025)
-
Microglia efferocytosis: an emerging mechanism for the resolution of neuroinflammation in Alzheimer’s disease
Journal of Neuroinflammation (2025)
-
Regulation of inflammatory processes by caspases
Nature Reviews Molecular Cell Biology (2025)
-
Anti-Atherogenic Mechanisms and Therapies
Current Atherosclerosis Reports (2025)