Defining trained immunity and its role in health and disease

Mihai G Netea, Jorge Domínguez-Andrés, Luis B Barreiro, Triantafyllos Chavakis, Maziar Divangahi, Elaine Fuchs, Leo A B Joosten, Jos W M van der Meer, Musa M Mhlanga, Willem J M Mulder, Niels P Riksen, Andreas Schlitzer, Joachim L Schultze, Christine Stabell Benn, Joseph C Sun, Ramnik J Xavier, Eicke Latz, Mihai G Netea, Jorge Domínguez-Andrés, Luis B Barreiro, Triantafyllos Chavakis, Maziar Divangahi, Elaine Fuchs, Leo A B Joosten, Jos W M van der Meer, Musa M Mhlanga, Willem J M Mulder, Niels P Riksen, Andreas Schlitzer, Joachim L Schultze, Christine Stabell Benn, Joseph C Sun, Ramnik J Xavier, Eicke Latz

Abstract

Immune memory is a defining feature of the acquired immune system, but activation of the innate immune system can also result in enhanced responsiveness to subsequent triggers. This process has been termed 'trained immunity', a de facto innate immune memory. Research in the past decade has pointed to the broad benefits of trained immunity for host defence but has also suggested potentially detrimental outcomes in immune-mediated and chronic inflammatory diseases. Here we define 'trained immunity' as a biological process and discuss the innate stimuli and the epigenetic and metabolic reprogramming events that shape the induction of trained immunity.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Trained immunity and tolerance: two…
Fig. 1. Trained immunity and tolerance: two opposite functional programmes of innate immunity.
Infections or sterile tissue triggers induce inflammation and the activation of immune effector mechanisms. Concomitant to a proinflammatory response, anti-inflammatory mechanisms are provoked to prevent overshooting inflammation and tissue damage and to limit the inflammatory response in time. Trained immunity involves epigenetic and metabolic reprogramming of the innate immune cells, allowing qualitatively and quantitatively adjusted responses of innate immune cells to subsequent time-delayed heterologous stimulation. Misguided trained immunity responses can contribute to disease progression, resulting in either a chronic hyperinflammatory state or a persistent state of immunological tolerance, a mechanism that dampens the inflammatory response of the host to maintain homeostasis and prevent tissue damage and organ failure, with the subsequent risk of secondary infections and other diseases related to decreased activity of the immune system.
Fig. 2. Central and peripheral trained immunity.
Fig. 2. Central and peripheral trained immunity.
Although trained immunity was first established in cells of the mononuclear phagocyte lineage (that is, monocytes and macrophages), monocytes have a relatively short lifespan and are unlikely to transmit their memory phenotype to their progeny and provide sustainable protection. Thus, current vaccine strategies that directly target monocytes or macrophages may have limited capacity for generating sustained innate immune memory. By contrast, haematopoietic stem cells (HSCs) are long-lived cells with self-renewal properties that reside in the bone marrow. The bone marrow is the site of haematopoiesis where HSCs continually undergo asymmetric division giving rise to the full repertoire of myeloid and lymphoid cell types. HSCs can directly respond to acute and chronic infections. Although the exact mechanisms of precursor proliferation or differentiation are not well understood, persistent activation of HSCs can result in their exhaustion, leading to devastating effects on the systemic immune compartment. Monocytes derived from trained HSCs migrate to peripheral organs, where they give rise to monocyte-derived macrophages with enhanced effector functions against different types of pathogens. Natural killer (NK) cells possess adaptive immune characteristics following infection. On reinfection, these memory NK cells undergo a secondary expansion and can more rapidly degranulate and release cytokines, resulting in a more protective immune response. Epithelial stem cells show memory functions during human allergic inflammatory disease, displaying changes in the chromatin accessibility when the stimulus is withdrawn. BCG, bacillus Calmette–Guérin; CMP, common myeloid progenitor: GMP, granulocyte–macrophage progenitor; MPP, multipotent progenitor.
Fig. 3. Interplay between epigenetics and metabolism.
Fig. 3. Interplay between epigenetics and metabolism.
The correct initiation of the mechanisms necessary for the induction of trained immunity relies on the active interplay between epigenetic and metabolic reprogramming of the innate immune cells on stimulation. During primary challenge, the recognition of specific ligands by pattern recognition receptors triggers a series of intracellular cascades that lead to the upregulation of different metabolic pathways, such as glycolysis, tricarboxylic acid (TCA) cycle and fatty acid metabolism. Certain metabolites derived from these processes, such as fumarate and acetyl coenzyme A (acetyl-CoA), can activate or inhibit a series of enzymes involved in remodelling the epigenetic landscape of cells, such as the histone demethylase lysine-specific demethylase 5 (KDM5) or histone acetyltransferases, leading to specific changes in histone methylation and acetylation of genes involved in the innate immune responses. β-Glucan-mediated activation of dectin 1 signalling also triggers calcium influx, which leads to the dephosphorylation of nuclear factor of activated T cells (NFAT), allowing its translocation into the nucleus, where it may bind to DNA and activate gene transcription. This facilitates the accessibility of the DNA to the transcriptional machinery and gene regulatory elements and specific long non-coding RNAs, promoting and facilitating an enhanced gene transcription on secondary stimulation of the cells. IGF1R, insulin-like growth factor 1 receptor; MLL1, mixed-lineage leukaemia protein 1 (also known as histone-lysine N-methyltransferase 2A); mTOR, mechanistic target of rapamycin; Pol, polymerase; UMLILO, upstream master long non-coding RNA of the inflammatory chemokine locus; WDR5, WD repeat-containing protein 5.
Fig. 4. Epigenetic reprogramming underlies the induction…
Fig. 4. Epigenetic reprogramming underlies the induction of trained immunity.
Stimulation of innate immune cells is accompanied by the deposition of chromatin marks and changes in the DNA methylation status, leading to unfolding of chromatin and facilitating transcription and expression of proinflammatory factors. All of these changes are only partially removed after cessation of the stimulus. This allows quicker and enhanced recruitment of transcription factors and gene expression after secondary challenge with another stimulus. The figure illustrates the chromatin states and epigenetic signatures associated with unstimulated cells, with cells following acute stimulation, with resting ‘trained’ cells and with trained cells following restimulation. H3K27ac, histone 3 lysine 27 acetylation; H3K4me, histone 3 lysine 4 methylation; H3K4me3, histone 3 lysine 4 trimethylation.

References

    1. Medzhitov R, Janeway C. Innate immune recognition: mechanisms and pathways. Immunol. Rev. 2000;173:89–97. doi: 10.1034/j.1600-065X.2000.917309.x.
    1. Lanier LL. NK cell recognition. Annu. Rev. Immunol. 2005;23:225–274. doi: 10.1146/annurev.immunol.23.021704.115526.
    1. Bonilla FA, Oettgen HC. Adaptive immunity. J. Allergy Clin. Immunol. 2010;125:S33–S40. doi: 10.1016/j.jaci.2009.09.017.
    1. Bowdish DME, Loffredo MS, Mukhopadhyay S, Mantovani A, Gordon S. Macrophage receptors implicated in the “adaptive” form of innate immunity. Microbes Infect. 2007;9:1680–1687. doi: 10.1016/j.micinf.2007.09.002.
    1. Netea MG, Quintin J, Van Der Meer JWM. Trained immunity: a memory for innate host defense. Cell Host Microbe. 2011;9:355–361. doi: 10.1016/j.chom.2011.04.006.
    1. Naik S, et al. Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature. 2017;550:475–480. doi: 10.1038/nature24271.
    1. Lay K, et al. Stem cells repurpose proliferation to contain a breach in their niche barrier. eLife. 2018;7:e41661. doi: 10.7554/eLife.41661.
    1. Christ A, et al. Western diet triggers NLRP3-dependent innate immune reprogramming. Cell. 2018;172:162–175. doi: 10.1016/j.cell.2017.12.013.
    1. Kurtz J. Specific memory within innate immune systems. Trends Immunol. 2005;26:186–192. doi: 10.1016/j.it.2005.02.001.
    1. Conrath U, Beckers GJM, Langenbach CJG, Jaskiewicz MR. Priming for enhanced defense. Annu. Rev. Phytopathol. 2015;53:97–119. doi: 10.1146/annurev-phyto-080614-120132.
    1. Gourbal B, et al. Innate immune memory: an evolutionary perspective. Immunol. Rev. 2018;283:21–40. doi: 10.1111/imr.12647.
    1. Netea MG, et al. Trained immunity: a program of innate immune memory in health and disease. Science. 2016;352:aaf1098. doi: 10.1126/science.aaf1098.
    1. Novakovic B, et al. β-glucan reverses the epigenetic state of lps-induced immunological tolerance. Cell. 2016;167:1354–1368.e14. doi: 10.1016/j.cell.2016.09.034.
    1. Nankabirwa V, et al. Child survival and BCG vaccination: a community based prospective cohort study in Uganda. BMC Public Health. 2015;15:175. doi: 10.1186/s12889-015-1497-8.
    1. Dominguez-Andres J, Netea MG. Long-term reprogramming of the innate immune system. J. Leukoc. Biol. 2018;105:329–338. doi: 10.1002/JLB.MR0318-104R.
    1. Berendsen MLT, et al. Maternal priming: bacillus Calmette-Guérin (BCG) vaccine scarring in mothers enhances the survival of their child with a BCG vaccine scar. J. Pediatric Infect. Dis. Soc. 2019 doi: 10.1093/jpids/piy142.
    1. Moore RS, Kaletsky R, Murphy CT. Piwi/PRG-1 argonaute and TGF-β mediate transgenerational learned pathogenic avoidance. Cell. 2019;177:1827–1841. doi: 10.1016/j.cell.2019.05.024.
    1. Hirano M, Das S, Guo P, Cooper MD. The evolution of adaptive immunity in vertebrates. in. Adv. Immunol. 2011;109:125–157. doi: 10.1016/B978-0-12-387664-5.00004-2.
    1. Cooper MD, Alder MN. The evolution of adaptive immune systems. Cell. 2006;124:815–822. doi: 10.1016/j.cell.2006.02.001.
    1. Purvis A, Hector A. Getting the measure of biodiversity. Nature. 2000;405:212–219. doi: 10.1038/35012221.
    1. Milutinović B, Kurtz J. Immune memory in invertebrates. Semin. Immunol. 2016;28:328–342. doi: 10.1016/j.smim.2016.05.004.
    1. Reimer-Michalski E-M, Conrath U. Innate immune memory in plants. Semin. Immunol. 2016;28:319–327. doi: 10.1016/j.smim.2016.05.006.
    1. Kleinnijenhuis J, et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc. Natl Acad. Sci. USA. 2012;109:17537–17542. doi: 10.1073/pnas.1202870109.
    1. Di Luzio NR, Williams DL. Protective effect of glucan against systemic Staphylococcus aureus septicemia in normal and leukemic mice. Infect. Immun. 1978;20:804–810. doi: 10.1128/IAI.20.3.804-810.1978.
    1. Marakalala MJ, et al. Dectin-1 plays a redundant role in the immunomodulatory activities of β-glucan-rich ligands in vivo. Microbes Infect. 2013;15:511–515. doi: 10.1016/j.micinf.2013.03.002.
    1. Krahenbuhl JL, Sharma SD, Ferraresi RW, Remington JS. Effects of muramyl dipeptide treatment on resistance to infection with Toxoplasma gondii in mice. Infect. Immun. 1981;31:716–722. doi: 10.1128/IAI.31.2.716-722.1981.
    1. Ribes S, et al. Intraperitoneal prophylaxis with CpG oligodeoxynucleotides protects neutropenic mice against intracerebral Escherichia coli K1 infection. J. Neuroinflammation. 2014;11:14. doi: 10.1186/1742-2094-11-14.
    1. Muñoz N, et al. Mucosal administration of flagellin protects mice from Streptococcus pneumoniae lung infection. Infect. Immun. 2010;78:4226–4233. doi: 10.1128/IAI.00224-10.
    1. Zhang B, et al. Viral infection. Prevention and cure of rotavirus infection via TLR5/NLRC4-mediated production of IL-22 and IL-18. Science. 2014;346:861–865. doi: 10.1126/science.1256999.
    1. van’t Wout JW, Poell R, van Furth R. The role of BCG/PPD-activated macrophages in resistance against systemic candidiasis in mice. Scand. J. Immunol. 1992;36:713–719. doi: 10.1111/j.1365-3083.1992.tb03132.x.
    1. Tribouley J, Tribouley-Duret J, Appriou M. Effect of bacillus Callmette Guerin (BCG) on the receptivity of nude mice to Schistosoma mansoni. C. R. Seances Soc. Biol. Fil. 1978;172:902–904.
    1. Kaufmann E, et al. BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis. Cell. 2018;172:176–190.e19. doi: 10.1016/j.cell.2017.12.031.
    1. Bromuro C, et al. Interplay between protective and inhibitory antibodies dictates the outcome of experimentally disseminated candidiasis in recipients of a Candida albicans vaccine. Infect. Immun. 2002;70:5462–5470. doi: 10.1128/IAI.70.10.5462-5470.2002.
    1. Polonelli L, et al. Therapeutic activity of an engineered synthetic killer antiidiotypic antibody fragment against experimental mucosal and systemic candidiasis. Infect. Immun. 2003;71:6205–6212. doi: 10.1128/IAI.71.11.6205-6212.2003.
    1. Bistoni F, et al. Evidence for macrophage-mediated protection against lethal Candida albicans infection. Infect. Immun. 1986;51:668–674. doi: 10.1128/IAI.51.2.668-674.1986.
    1. Bistoni F, et al. Immunomodulation by a low-virulence, agerminative variant of Candida albicans. Further evidence for macrophage activation as one of the effector mechanisms of nonspecific anti-infectious protection. J. Med. Vet. Mycol. 1988;26:285–299. doi: 10.1080/02681218880000401.
    1. Quintin J, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe. 2012;12:223–232. doi: 10.1016/j.chom.2012.06.006.
    1. Vecchiarelli A, et al. Protective immunity induced by low-virulence Candida albicans: cytokine production in the development of the anti-infectious state. Cell. Immunol. 1989;124:334–344. doi: 10.1016/0008-8749(89)90135-4.
    1. Tso GHW, et al. Experimental evolution of a fungal pathogen into a gut symbiont. Science. 2018;362:589–595. doi: 10.1126/science.aat0537.
    1. Biering-Sørensen S, et al. Early BCG-Denmark and neonatal mortality among infants weighing <2500 g: a randomized controlled trial. Clin. Infect. Dis. 2017;65:1183–1190. doi: 10.1093/cid/cix525.
    1. Rieckmann A, et al. Vaccinations against smallpox and tuberculosis are associated with better long-term survival: a Danish case-cohort study 1971–2010. Int. J. Epidemiol. 2017;46:695–705.
    1. Aaby P, et al. Vaccinia scars associated with better survival for adults. An observational study from Guinea-Bissau. Vaccine. 2006;24:5718–5725. doi: 10.1016/j.vaccine.2006.04.045.
    1. Aaby P, et al. Randomized trial of BCG vaccination at birth to low-birth-weight children: beneficial nonspecific effects in the neonatal period? J. Infect. Dis. 2011;204:245–252. doi: 10.1093/infdis/jir240.
    1. Aaby P, et al. Non-specific beneficial effect of measles immunisation: analysis of mortality studies from developing countries. BMJ. 1995;311:481–485. doi: 10.1136/bmj.311.7003.481.
    1. Aaby P, et al. Non-specific effects of standard measles vaccine at 4.5 and 9 months of age on childhood mortality: randomised controlled trial. BMJ. 2010;341:c6495. doi: 10.1136/bmj.c6495.
    1. Lund N, et al. The effect of oral polio vaccine at birth on infant mortality: a randomized trial. Clin. Infect. Dis. 2015;61:1504–1511. doi: 10.1093/cid/civ617.
    1. Andersen A, et al. National immunization campaigns with oral polio vaccine reduce all-cause mortality: a natural experiment within seven randomized trials. Front. Public Health. 2018;6:13. doi: 10.3389/fpubh.2018.00013.
    1. Benn CS, Netea MG, Selin LK, Aaby P. A small jab - a big effect: nonspecific immunomodulation by vaccines. Trends Immunol. 2013;34:431–439. doi: 10.1016/j.it.2013.04.004.
    1. Kleinnijenhuis J, et al. BCG-induced trained immunity in NK cells: role for non-specific protection to infection. Clin. Immunol. 2014;155:213–219. doi: 10.1016/j.clim.2014.10.005.
    1. Jensen KJ, et al. Heterologous immunological effects of early BCG vaccination in low-birth-weight infants in Guinea-Bissau: a randomized-controlled trial. J. Infect. Dis. 2015;211:956–967. doi: 10.1093/infdis/jiu508.
    1. Freyne B, et al. Neonatal BCG vaccination influences cytokine responses to toll-like receptor ligands and heterologous antigens. J. Infect. Dis. 2018;217:1798–1808. doi: 10.1093/infdis/jiy069.
    1. Aaby P, et al. Differences in female-male mortality after high-titre measles vaccine and association with subsequent vaccination with diphtheria-tetanus-pertussis and inactivated poliovirus: reanalysis of West African studies. Lancet. 2003;361:2183–2188. doi: 10.1016/S0140-6736(03)13771-3.
    1. Blok BA, et al. Interacting non-specific immunological effects of BCG and TDAPF vaccinations: an explorative randomized trial. Clin. Infect. Dis. 2019;70:455–463.
    1. Arts RJW, et al. BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe. 2018;23:89–100. doi: 10.1016/j.chom.2017.12.010.
    1. Walk J, et al. Outcomes of controlled human malaria infection after BCG vaccination. Nat. Commun. 2019;10:874. doi: 10.1038/s41467-019-08659-3.
    1. McCall MBB, et al. Plasmodium falciparum infection causes proinflammatory priming of human TLR responses. J. Immunol. 2007;179:162–171. doi: 10.4049/jimmunol.179.1.162.
    1. Ataide MA, et al. Malaria-induced NLRP12/NLRP3-dependent caspase-1 activation mediates inflammation and hypersensitivity to bacterial superinfection. PLoS Pathog. 2014;10:e1003885. doi: 10.1371/journal.ppat.1003885.
    1. Fitzgerald KA, et al. Cutting edge: Plasmodium falciparum induces trained innate immunity. J. Immunol. 2018;6:1243–1248.
    1. Redelman-Sidi G, Glickman MS, Bochner BH. The mechanism of action of BCG therapy for bladder cancer—a current perspective. Nat. Rev. Urol. 2014;11:153–162. doi: 10.1038/nrurol.2014.15.
    1. Stewart JH, Levine EA. Role of bacillus Calmette-Guérin in the treatment of advanced melanoma. Expert. Rev. Anticancer. Ther. 2011;11:1671–1676. doi: 10.1586/era.11.163.
    1. Powles RL, et al. Maintenance of remission in acute myelogenous leukaemia by a mixture of B.C.G. and irradiated leukaemia cells. Lancet. 1977;2:1107–1110. doi: 10.1016/S0140-6736(77)90549-9.
    1. Villumsen M, et al. Risk of lymphoma and leukaemia after bacille Calmette-Guérin and smallpox vaccination: a Danish case-cohort study. Vaccine. 2009;27:6950–6958. doi: 10.1016/j.vaccine.2009.08.103.
    1. Buffen K, et al. Autophagy controls BCG-induced trained immunity and the response to intravesical BCG therapy for bladder cancer. PLoS Pathog. 2014;10:e1004485. doi: 10.1371/journal.ppat.1004485.
    1. Foster SL, Hargreaves DC, Medzhitov R. Gene-specific control of inflammation by TLR-induced chromatin modifications. Nature. 2007;447:972–978. doi: 10.1038/nature05836.
    1. Saeed S, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science. 2014;345:1251086. doi: 10.1126/science.1251086.
    1. Chen F, et al. Neutrophils prime a long-lived effector macrophage phenotype that mediates accelerated helminth expulsion. Nat. Immunol. 2014;15:938–946. doi: 10.1038/ni.2984.
    1. Hole CR, et al. Induction of memory-like dendritic cell responses in vivo. Nat. Commun. 2019;10:2955. doi: 10.1038/s41467-019-10486-5.
    1. Barton ES, et al. Herpesvirus latency confers symbiotic protection from bacterial infection. Nature. 2007;447:326–329. doi: 10.1038/nature05762.
    1. Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature. 2009;457:557–561. doi: 10.1038/nature07665.
    1. Sun JC, et al. Proinflammatory cytokine signaling required for the generation of natural killer cell memory. J. Exp. Med. 2012;209:947–954. doi: 10.1084/jem.20111760.
    1. Björkström NK, et al. Rapid expansion and long-term persistence of elevated NK cell numbers in humans infected with hantavirus. J. Exp. Med. 2011;208:13–21. doi: 10.1084/jem.20100762.
    1. Della Chiesa M, et al. Phenotypic and functional heterogeneity of human NK cells developing after umbilical cord blood transplantation: a role for human cytomegalovirus? Blood. 2012;119:399–410. doi: 10.1182/blood-2011-08-372003.
    1. Foley B, et al. Human cytomegalovirus (CMV)-induced memory-like NKG2C+ NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J. Immunol. 2012;189:5082–5088. doi: 10.4049/jimmunol.1201964.
    1. Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002;296:1323–1326. doi: 10.1126/science.1070884.
    1. Brown MG, et al. Vital involvement of a natural killer cell activation receptor in resistance to viral infection. Science. 2001;292:934–937. doi: 10.1126/science.1060042.
    1. Adams NM, et al. Cytomegalovirus infection drives avidity selection of natural killer cells. Immunity. 2019;50:1381–1390.e5. doi: 10.1016/j.immuni.2019.04.009.
    1. Grassmann S, et al. Distinct surface expression of activating receptor Ly49H drives differential expansion of NK cell clones upon murine cytomegalovirus infection. Immunity. 2019;50:1391–1400.e4. doi: 10.1016/j.immuni.2019.04.015.
    1. Sun JC, Lanier LL. NK cell development, homeostasis and function: parallels with CD8+ T cells. Nat. Rev. Immunol. 2011;11:645–657. doi: 10.1038/nri3044.
    1. Min-Oo G, Lanier LL. Cytomegalovirus generates long-lived antigen-specific NK cells with diminished bystander activation to heterologous infection. J. Exp. Med. 2014;211:2669–2680. doi: 10.1084/jem.20141172.
    1. Rölle A, et al. IL-12–producing monocytes and HLA-E control HCMV-driven NKG2C+ NK cell expansion. J. Clin. Invest. 2014;124:5305–5316. doi: 10.1172/JCI77440.
    1. Hammer Q, et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 2018;19:453–463. doi: 10.1038/s41590-018-0082-6.
    1. Karo JM, Schatz DG, Sun JC. The RAG recombinase dictates functional heterogeneity and cellular fitness in natural killer cells. Cell. 2014;159:94–107. doi: 10.1016/j.cell.2014.08.026.
    1. Vivier E, et al. Innate or adaptive immunity? The example of natural killer cells. Science. 2011;331:44–49. doi: 10.1126/science.1198687.
    1. Cooper MA, et al. Cytokine-induced memory-like natural killer cells. Proc. Natl Acad. Sci. USA. 2009;106:1915–1919. doi: 10.1073/pnas.0813192106.
    1. Romee R, et al. Cytokine activation induces human memory-like NK cells. Blood. 2012;120:4751–4760. doi: 10.1182/blood-2012-04-419283.
    1. Romee R, et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci. Transl Med. 2016;8:357ra123. doi: 10.1126/scitranslmed.aaf2341.
    1. Madera S, et al. Type I IFN promotes NK cell expansion during viral infection by protecting NK cells against fratricide. J. Exp. Med. 2016;213:225–233. doi: 10.1084/jem.20150712.
    1. Madera S, Sun JC. Cutting edge: stage-specific requirement of IL-18 for antiviral NK cell expansion. J. Immunol. 2015;194:1408–1412. doi: 10.4049/jimmunol.1402001.
    1. Nabekura T, Girard J-P, Lanier LL. IL-33 receptor ST2 amplifies the expansion of NK cells and enhances host defense during mouse cytomegalovirus infection. J. Immunol. 2015;194:5948–5952. doi: 10.4049/jimmunol.1500424.
    1. Weizman O-E, et al. Mouse cytomegalovirus-experienced ILC1s acquire a memory response dependent on the viral glycoprotein m12. Nat. Immunol. 2019;20:1004–1011. doi: 10.1038/s41590-019-0430-1.
    1. Martinez-Gonzalez I, et al. Allergen-experienced group 2 innate lymphoid cells acquire memory-like properties and enhance allergic lung inflammation. Immunity. 2016;45:198–208. doi: 10.1016/j.immuni.2016.06.017.
    1. Paust S, et al. Critical role for the chemokine receptor CXCR6 in NK cell–mediated antigen-specific memory of haptens and viruses. Nat. Immunol. 2010;11:1127–1135. doi: 10.1038/ni.1953.
    1. Gamliel M, et al. Trained memory of human uterine NK cells enhances their function in subsequent pregnancies. Immunity. 2018;48:951–962. doi: 10.1016/j.immuni.2018.03.030.
    1. Gonzales KAU, Fuchs E. Skin and its regenerative powers: an alliance between stem cells and their niche. Dev. Cell. 2017;43:387–401. doi: 10.1016/j.devcel.2017.10.001.
    1. Naik S, Larsen SB, Cowley CJ, Fuchs E. Two to tango: dialog between immunity and stem cells in health and disease. Cell. 2018;175:908–920. doi: 10.1016/j.cell.2018.08.071.
    1. Biton M, et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell. 2018;175:1307–1320. doi: 10.1016/j.cell.2018.10.008.
    1. Lindemans CA, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528:560–564. doi: 10.1038/nature16460.
    1. von Moltke J, Ji M, Liang H-E, Locksley RM. Tuft-cell-derived IL-25 regulates an intestinal ILC2–epithelial response circuit. Nature. 2016;529:221–225. doi: 10.1038/nature16161.
    1. Kamada R, et al. Interferon stimulation creates chromatin marks and establishes transcriptional memory. Proc. Natl Acad. Sci. USA. 2018;115:E9162–E9171. doi: 10.1073/pnas.1720930115.
    1. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 2013;31:563–604. doi: 10.1146/annurev-immunol-020711-074950.
    1. Yona S, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 2013;38:79–91. doi: 10.1016/j.immuni.2012.12.001.
    1. Patel AA, et al. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J. Exp. Med. 2017;214:1913–1923. doi: 10.1084/jem.20170355.
    1. Chavakis T, Mitroulis I, Hajishengallis G. Hematopoietic progenitor cells as integrative hubs for adaptation to and fine-tuning of inflammation. Nat. Immunol. 2019;20:802–811. doi: 10.1038/s41590-019-0402-5.
    1. Mitroulis I, et al. Modulation of myelopoiesis progenitors is an integral component of trained immunity. Cell. 2018;172:147–161. doi: 10.1016/j.cell.2017.11.034.
    1. Ordovas-Montanes J, et al. Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature. 2018;560:649–654. doi: 10.1038/s41586-018-0449-8.
    1. Machiels B, et al. A gammaherpesvirus provides protection against allergic asthma by inducing the replacement of resident alveolar macrophages with regulatory monocytes. Nat. Immunol. 2017;18:1310–1320. doi: 10.1038/ni.3857.
    1. Yao Y, et al. Induction of autonomous memory alveolar macrophages requires T cell help and is critical to trained immunity. Cell. 2018;175:1634–1650.e17. doi: 10.1016/j.cell.2018.09.042.
    1. Smale ST, Tarakhovsky A, Natoli G. Chromatin contributions to the regulation of innate immunity. Annu. Rev. Immunol. 2014;32:489–511. doi: 10.1146/annurev-immunol-031210-101303.
    1. Ghisletti S, et al. Identification and characterization of enhancers controlling the inflammatory gene expression program in macrophages. Immunity. 2010;32:317–328. doi: 10.1016/j.immuni.2010.02.008.
    1. Fanucchi S, et al. Immune genes are primed for robust transcription by proximal long noncoding RNAs located in nuclear compartments. Nat. Genet. 2019;51:138–150. doi: 10.1038/s41588-018-0298-2.
    1. Natoli G, Ostuni R. Adaptation and memory in immune responses. Nat. Immunol. 2019;20:783–792. doi: 10.1038/s41590-019-0399-9.
    1. Luetke-Eversloh M, et al. Human cytomegalovirus drives epigenetic imprinting of the IFNG locus in NKG2Chi natural killer cells. PLoS Pathog. 2014;10:e1004441. doi: 10.1371/journal.ppat.1004441.
    1. Lau CM, et al. Epigenetic control of innate and adaptive immune memory. Nat. Immunol. 2018;19:963–972. doi: 10.1038/s41590-018-0176-1.
    1. Rapp M, et al. Core-binding factor β and Runx transcription factors promote adaptive natural killer cell responses. Sci. Immunol. 2017;2:eaan3796. doi: 10.1126/sciimmunol.aan3796.
    1. Geary CD, et al. Non-redundant ISGF3 components promote NK cell survival in an auto-regulatory manner during viral infection. Cell Rep. 2018;24:1949–1957.e6. doi: 10.1016/j.celrep.2018.07.060.
    1. Lam VC, Folkersen L, Aguilar OA, Lanier LL. KLF12 regulates mouse NK cell proliferation. J. Immunol. 2019;203:981–989. doi: 10.4049/jimmunol.1900396.
    1. Adams NM, et al. Transcription factor IRF8 orchestrates the adaptive natural killer cell response. Immunity. 2018;48:1172–1182.e6. doi: 10.1016/j.immuni.2018.04.018.
    1. Madera S, et al. Cutting edge: divergent requirement of T-box transcription factors in effector and memory NK cells. J. Immunol. 2018;200:1977–1981. doi: 10.4049/jimmunol.1700416.
    1. Zawislak CL, et al. Stage-specific regulation of natural killer cell homeostasis and response against viral infection by microRNA-155. Proc. Natl Acad. Sci. USA. 2013;110:6967–6972. doi: 10.1073/pnas.1304410110.
    1. Lu L-F, et al. A single miRNA-mRNA interaction affects the immune response in a context- and cell-type-specific manner. Immunity. 2015;43:52–64. doi: 10.1016/j.immuni.2015.04.022.
    1. O’Connell RM, Chaudhuri AA, Rao DS, Baltimore D. Inositol phosphatase SHIP1 is a primary target of miR-155. Proc. Natl Acad. Sci. USA. 2009;106:7113–7118. doi: 10.1073/pnas.0902636106.
    1. Saz-Leal P, et al. Targeting SHIP-1 in myeloid cells enhances trained immunity and boosts response to infection. Cell Rep. 2018;25:1118–1126. doi: 10.1016/j.celrep.2018.09.092.
    1. Verma D, et al. Anti-mycobacterial activity correlates with altered DNA methylation pattern in immune cells from BCG-vaccinated subjects. Sci. Rep. 2017;7:12305. doi: 10.1038/s41598-017-12110-2.
    1. Das J, Verma D, Gustafsson M, Lerm M. Identification of DNA methylation patterns predisposing for an efficient response to BCG vaccination in healthy BCG-naïve subjects. Epigenetics. 2019;14:589–601. doi: 10.1080/15592294.2019.1603963.
    1. Penkov S, Mitroulis I, Hajishengallis G, Chavakis T. Immunometabolic crosstalk: an ancestral principle of trained immunity? Trends Immunol. 2019;40:1–11. doi: 10.1016/j.it.2018.11.002.
    1. Norata GD, et al. The cellular and molecular basis of translational immunometabolism. Immunity. 2015;43:421–434. doi: 10.1016/j.immuni.2015.08.023.
    1. Domínguez-Andrés J, Joosten LA, Netea MG. Induction of innate immune memory: the role of cellular metabolism. Curr. Opin. Immunol. 2019;56:10–16. doi: 10.1016/j.coi.2018.09.001.
    1. Donohoe DR, Bultman SJ. Metaboloepigenetics: interrelationships between energy metabolism and epigenetic control of gene expression. J. Cell. Physiol. 2012;227:3169–3177. doi: 10.1002/jcp.24054.
    1. Cheng S-C, et al. mTOR- and HIF-1-mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345:1250684–1250684. doi: 10.1126/science.1250684.
    1. Arts RJW, et al. Immunometabolic pathways in BCG-induced trained immunity. Cell Rep. 2016;17:2562–2571. doi: 10.1016/j.celrep.2016.11.011.
    1. Arts RJW, et al. Glutaminolysis and fumarate accumulation integrate immunometabolic and epigenetic programs in trained immunity. Cell Metab. 2016 doi: 10.1016/j.cmet.2016.10.008.
    1. Liu P-S, et al. α-Ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 2017;18:985–994. doi: 10.1038/ni.3796.
    1. Tannahill GM, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496:238–242. doi: 10.1038/nature11986.
    1. Lampropoulou V, et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 2016;24:158–166. doi: 10.1016/j.cmet.2016.06.004.
    1. Cordes T, et al. Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. J. Biol. Chem. 2016;291:14274–14284. doi: 10.1074/jbc.M115.685792.
    1. Mills EL, et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. 2018;556:113–117. doi: 10.1038/nature25986.
    1. Bambouskova M, et al. Electrophilic properties of itaconate and derivatives regulate the IκBζ–ATF3 inflammatory axis. Nature. 2018;556:501–504. doi: 10.1038/s41586-018-0052-z.
    1. Dominguez-Andres J, et al. The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab. 2018;29:211–220. doi: 10.1016/j.cmet.2018.09.003.
    1. Bekkering S, et al. Metabolic induction of trained immunity through the mevalonate pathway. Cell. 2018;172:135–146.e9. doi: 10.1016/j.cell.2017.11.025.
    1. Yvan-Charvet L, et al. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science. 2010;328:1689–1693. doi: 10.1126/science.1189731.
    1. Murphy AJ, et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J. Clin. Invest. 2011;121:4138–4149. doi: 10.1172/JCI57559.
    1. Christ A, Latz E. The Western lifestyle has lasting effects on metaflammation. Nat. Rev. Immunol. 2019;19:267–268. doi: 10.1038/s41577-019-0156-1.
    1. Musher DM, Abers MS, Corrales-Medina VF. Acute infection and myocardial infarction. N. Engl. J. Med. 2019;380:171–176. doi: 10.1056/NEJMra1808137.
    1. Leentjens J, et al. Trained innate immunity as a novel mechanism linking infection and the development of atherosclerosis. Circ. Res. 2018;122:664–669. doi: 10.1161/CIRCRESAHA.117.312465.
    1. Bekkering S, et al. Oxidized low-density lipoprotein induces long-term proinflammatory cytokine production and foam cell formation via epigenetic reprogramming of monocytes. Arterioscler. Thromb. Vasc. Biol. 2014;34:1731–1738. doi: 10.1161/ATVBAHA.114.303887.
    1. Braza MS, et al. Inhibiting inflammation with myeloid cell-specific nanobiologics promotes organ transplant acceptance. Immunity. 2018;49:819–828. doi: 10.1016/j.immuni.2018.09.008.
    1. van der Valk FM, et al. Oxidized phospholipids on lipoprotein(a) elicit arterial wall inflammation and an inflammatory monocyte response in humans. Circulation. 2016;134:611–624. doi: 10.1161/CIRCULATIONAHA.116.020838.
    1. Bekkering S, et al. Treatment with statins does not revert trained immunity in patients with familial hypercholesterolemia. Cell Metab. 2019;30:1–2. doi: 10.1016/j.cmet.2019.05.014.
    1. Bekkering S, et al. Innate immune cell activation and epigenetic remodeling in symptomatic and asymptomatic atherosclerosis in humans in vivo. Atherosclerosis. 2016;254:228–236. doi: 10.1016/j.atherosclerosis.2016.10.019.
    1. Shirai T, et al. The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease. J. Exp. Med. 2016;213:337–354. doi: 10.1084/jem.20150900.
    1. Zimmermann P, Finn A, Curtis N. Does BCG vaccination protect against nontuberculous mycobacterial infection? A systematic review and meta-analysis. J. Infect. Dis. 2018;218:679–687. doi: 10.1093/infdis/jiy207.
    1. Leentjens J, et al. BCG vaccination enhances the immunogenicity of subsequent influenza vaccination in healthy volunteers: a randomized, placebo-controlled pilot study. J. Infect. Dis. 2015;212:1930–1938. doi: 10.1093/infdis/jiv332.
    1. Ransohoff RM. How neuroinflammation contributes to neurodegeneration. Science. 2016;353:777–783. doi: 10.1126/science.aag2590.
    1. Wendeln A-C, et al. Innate immune memory in the brain shapes neurological disease hallmarks. Nature. 2018;556:332–338. doi: 10.1038/s41586-018-0023-4.
    1. Frost PS, et al. Neonatal infection leads to increased susceptibility to Aβ oligomer-induced brain inflammation, synapse loss and cognitive impairment in mice. Cell Death Dis. 2019;10:323. doi: 10.1038/s41419-019-1529-x.
    1. Low A, Mak E, Rowe JB, Markus HS, O’Brien JT. Inflammation and cerebral small vessel disease: a systematic review. Ageing Res. Rev. 2019;53:100916. doi: 10.1016/j.arr.2019.100916.
    1. Noz MP, et al. Trained immunity characteristics are associated with progressive cerebral small vessel disease. Stroke. 2018;49:2910–2917. doi: 10.1161/STROKEAHA.118.023192.
    1. Franceschi C, et al. Immunobiography and the heterogeneity of immune responses in the elderly: a focus on inflammaging and trained immunity. Front. Immunol. 2017;8:982. doi: 10.3389/fimmu.2017.00982.
    1. Qian F, et al. Age-associated elevation in TLR5 leads to increased inflammatory responses in the elderly. Aging Cell. 2012;11:104–110. doi: 10.1111/j.1474-9726.2011.00759.x.
    1. Metcalf TU, et al. Global analyses revealed age-related alterations in innate immune responses after stimulation of pathogen recognition receptors. Aging Cell. 2015;14:421–432. doi: 10.1111/acel.12320.
    1. Hoogeboom R, et al. A mutated B cell chronic lymphocytic leukemia subset that recognizes and responds to fungi. J. Exp. Med. 2013;210:59–70. doi: 10.1084/jem.20121801.
    1. Jurado-Camino T, et al. Chronic lymphocytic leukemia: a paradigm of innate immune cross-tolerance. J. Immunol. 2015;194:719–727. doi: 10.4049/jimmunol.1402272.
    1. Pauken KE, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354:1160–1165. doi: 10.1126/science.aaf2807.
    1. Lee SH, et al. TNFα enhances cancer stem cell-like phenotype via Notch-Hes1 activation in oral squamous cell carcinoma cells. Biochem. Biophys. Res. Commun. 2012;424:58–64. doi: 10.1016/j.bbrc.2012.06.065.
    1. Hodge DR, Hurt EM, Farrar WL. The role of IL-6 and STAT3 in inflammation and cancer. Eur. J. Cancer. 2005;41:2502–2512. doi: 10.1016/j.ejca.2005.08.016.
    1. Rabold K, Netea MG, Adema GJ, Netea-Maier RT. Cellular metabolism of tumor-associated macrophages - functional impact and consequences. FEBS Lett. 2017;591:3022–3041. doi: 10.1002/1873-3468.12771.
    1. Cubas P, Vincent C, Coen E. An epigenetic mutation responsible for natural variation in floral symmetry. Nature. 1999;401:157–161. doi: 10.1038/43657.
    1. Greer EL, et al. A histone methylation network regulates transgenerational epigenetic memory in C. elegans. Cell Rep. 2014;7:113–126. doi: 10.1016/j.celrep.2014.02.044.
    1. Tate AT, Andolfatto P, Demuth JP, Graham AL. The within-host dynamics of infection in trans-generationally primed flour beetles. Mol. Ecol. 2017;26:3794–3807. doi: 10.1111/mec.14088.
    1. No Authors Listed. Meeting of the Strategic Advisory Group of Experts on immunization, April 2014 - conclusions and recommendations. Wkly Epidemiol. Rec. 2014;89:221–236.
    1. Shann F. The heterologous (non-specific) effects of vaccines: implications for policy in high-mortality countries. Trans. R. Soc. Trop. Med. Hyg. 2015;109:5–8. doi: 10.1093/trstmh/tru161.
    1. Benn CS, Fisker AB, Whittle HC, Aaby P. Revaccination with live attenuated vaccines confer additional beneficial nonspecific effects on overall survival: a review. EBioMedicine. 2016;10:312–317. doi: 10.1016/j.ebiom.2016.07.016.
    1. Locht C, Mielcarek N. Live attenuated vaccines against pertussis. Expert. Rev. Vaccines. 2014;13:1147–1158. doi: 10.1586/14760584.2014.942222.
    1. Meyers PA, et al. Osteosarcoma: the addition of muramyl tripeptide to chemotherapy improves overall survival-a report from the Children’s Oncology Group. J. Clin. Oncol. 2008;26:633–638. doi: 10.1200/JCO.2008.14.0095.
    1. Muramatsu D, et al. β-glucan derived from Aureobasidium pullulans is effective for the prevention of influenza in mice. PLoS One. 2012;7:e41399. doi: 10.1371/journal.pone.0041399.
    1. Mulder WJM, Ochando J, Joosten LAB, Fayad ZA, Netea MG. Therapeutic targeting of trained immunity. Nat. Rev. Drug. Discov. 2019;18:553–566. doi: 10.1038/s41573-019-0025-4.
    1. Levine DB. The Hospital for the Ruptured and Crippled: William Bradley Coley, third Surgeon-in-Chief 1925-1933. HSS J. 2008;4:1–9. doi: 10.1007/s11420-007-9063-2.
    1. Coley WB., II. Contribution to the knowledge of sarcoma. Ann. Surg. 1891;14:199–220. doi: 10.1097/00000658-189112000-00015.
    1. Old LJ, Clarke DA, Benacerraf B. Effect of bacillus Calmette-Guérin infection on transplanted tumours in the mouse. Nature. 1959;184:291–292. doi: 10.1038/184291a0.
    1. Ostadrahimi A, et al. Effect of beta glucan on white blood cell counts and serum levels of IL-4 and IL-12 in women with breast cancer undergoing chemotherapy: a randomized double-blind placebo-controlled clinical trial. Asian Pac. J. Cancer Prev. 2014;15:5733–5739. doi: 10.7314/APJCP.2014.15.14.5733.
    1. Bashir KMI, Choi J-S. Clinical and physiological perspectives of β-glucans: the past, present, and future. Int. J. Mol. Sci. 2017;18:1906. doi: 10.3390/ijms18091906.
    1. Vetvicka V, Vannucci L, Sima P, Richter J. Beta glucan: supplement or drug? from laboratory to clinical trials. Molecules. 2019;24:1251. doi: 10.3390/molecules24071251.
    1. Lim WA, June CH. The principles of engineering immune cells to treat. Cancer Cell. 2017;168:724–740.
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61. doi: 10.1126/science.aaa8172.
    1. Routy B, et al. The gut microbiota influences anticancer immunosurveillance and general health. Nat. Rev. Clin. Oncol. 2018;15:382–396. doi: 10.1038/s41571-018-0006-2.
    1. Lameijer M, et al. Efficacy and safety assessment of a TRAF6-targeted nanoimmunotherapy in atherosclerotic mice and non-human primates. Nat. Biomed. Eng. 2018;2:279–292. doi: 10.1038/s41551-018-0221-2.
    1. Netea MG, Schlitzer A, Placek K, Joosten LAB, Schultze JL. Innate and adaptive immune memory: an evolutionary continuum in the host’s response to pathogens. Cell Host Microbe. 2019;25:13–26. doi: 10.1016/j.chom.2018.12.006.
    1. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914.
    1. Domínguez-Andrés J, et al. Bromodomain inhibitor I-BET151 suppresses immune responses during fungal-immune interaction. Eur. J. Immunol. 2019;49:2044–2050. doi: 10.1002/eji.201848081.
    1. Alqahtani A, et al. Bromodomain and extra-terminal motif inhibitors: a review of preclinical and clinical advances in cancer therapy. Future Sci. OA. 2019;5:FSO372. doi: 10.4155/fsoa-2018-0115.

Source: PubMed

3
Abonneren