Bekkering S, Domà nguez-Andrés J, Joosten LAB, Riksen NP, Netea MG. Trained Immunity Reprogramming Innate Immunity in Health and Disease
Bekkering S, Domà nguez-Andrés J, Joosten LAB, Riksen NP, Netea MG. Trained Immunity Reprogramming Innate Immunity in Health and Disease
Bekkering S, Domà nguez-Andrés J, Joosten LAB, Riksen NP, Netea MG. Trained Immunity Reprogramming Innate Immunity in Health and Disease
Trained Immunity:
Reprogramming Innate
Immunity in Health
Access provided by 2806:2f0:7021:d9b7:f588:453c:1b73:c93e on 10/18/23. For personal use only.
and Disease
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
667
HISTORICAL PERSPECTIVE
As early as in 1931, in the Swedish province of Norrbotten, Carl Naeslund discovered that after
the introduction of bacillus Calmette-Guérin (BCG) vaccination in neonates—who are known to
have immature adaptive immune responses—mortality in the region dropped significantly. This
drop was more than could be explained by a decrease in tuberculosis alone, and he was “tempted
to find an explanation for this much lower mortality among vaccinated children in the idea that
BCG provokes a nonspecific immunity. . .” (1, p. 629). It took almost a century to understand
that the innate immune system can indeed provide protection against reinfection by building a
heterologous immunological memory in innate immune cells, a process that ten years ago was
termed trained immunity (2).
In plants and invertebrate animals, which both lack an adaptive immune system, the innate
Access provided by 2806:2f0:7021:d9b7:f588:453c:1b73:c93e on 10/18/23. For personal use only.
immune system is long known to present memory characteristics. Plants that survive an infection
obtain long-term protection against reinfection, a phenomenon called systemic acquired resis-
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
tance (SAR) (3). SAR spreads from the site of infection throughout the entire plant through sig-
naling molecules, subsequently inducing an enhanced expression of pattern-recognition receptors
(PRRs) and the secretion of antimicrobial proteins (4). This is mediated via epigenetic changes and
can even be even transmitted to the progeny through seeds (5). In invertebrates such as Drosophila
melanogaster, a sublethal dose of Streptococcus pneumoniae primes macrophages and protects the an-
imal for life against a secondary, otherwise lethal reinfection with the same pathogen, despite the
fact that the organism does not have an adaptive immune system (6). A similar phenomenon was
observed in beetles and was shown to go beyond even the first generation (7, 8). Even though the
definition and mechanisms of innate immune memory in vertebrates were only unraveled in the
last decade, older studies showed evidence of BCG protecting athymic nude mice against a sec-
ondary lethal infection with Candida albicans (9), through a process driven by macrophages (10).
In humans, multiple epidemiological studies have shown that live attenuated vaccines can protect
against heterologous microorganisms different from the target pathogen (11, 12). In conclusion,
plants, invertebrates, and vertebrates are all capable of developing adaptive characteristics in in-
nate immune cells, a de facto memory, which results in a nonspecific increased effector function of
the innate immune cell itself. The mechanisms and characteristics of this innate immune memory,
or trained immunity, substantially differ from those of the specific memory induced in cells of the
adaptive immune system by gene recombination.
Initial
stimulation
with A
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
Trained
immunity
Homeostasis
Tolerance
Acetyl CoA
Epigenetic and metabolic
reprogramming
Oxaloacetate Citric acid
NADH
NAD+
Malate Isocitric acid
H2O CO2
Krebs cycle NAD+
Fumarate
FADH2 NADH
FAD α-Ketoglutaric acid
Succinate CO2
NAD+
GTP Succinyl CoA NADH
GDP
Figure 1
Trained immunity versus tolerance. Microbial or endogenous stimuli (stimulus A) can activate innate
immune cells and induce a primary response. Innate immune cells will produce cytokines or activate other
effector functions. Depending on the dose and stimulus, subsequent anti-inflammatory mechanisms can be
induced, and a secondary stimulation (stimulus B) will not lead to another proinflammatory response, so as
to limit tissue damage (tolerance). However, when trained immunity is induced, this leads to enhanced innate
immune effector function upon secondary stimulation (compared to the initial response). Both training and
tolerance are mediated via metabolic and epigenetic changes, although each phenotype has its own signature:
Different genes and pathways will be activated or repressed by different epigenetic and metabolic changes.
Figure adapted from image created with BioRender.com.
wall component β-glucan (21) were among the first described inducers of trained immunity in
vitro as well as in vivo and are the most used in experimental studies on trained immunity. More
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
recently the malaria pathogen Plasmodium falciparum (22) and hepatitis B virus (11) were added to
the growing list of microbial inducers of trained immunity. Immune tolerance, on the other hand,
can be induced by lipopolysaccharide (LPS) endotoxins from Escherichia coli (23) and other micro-
bial ligands (24), although at lower concentration LPS is also able to induce trained immunity as
well (24). This dichotomy of dose-dependent training or tolerance is described for more danger-
associated molecular patterns and pathogen-associated molecular patterns (24). Nonmicrobial en-
dogenous inducers of trained immunity include lipoproteins such as oxidized low-density lipopro-
tein (oxLDL) (25), lipoprotein(a) [Lp(a)] (26), and activators of the liver X receptor pathways (27,
28); uric acid (29, 30); catecholamines (31); aldosterone (32); S100-alarmin (33); and interferons
(34). Importantly, all inducers of trained immunity affect the inflammatory functions of myeloid
cells, although with different potential. In general, microbial inducers of trained immunity such
as β-glucan and BCG have a greater potential to induce the inflammatory effector function upon
secondary stimulation than endogenous stimuli such as lipids. Importantly, besides its capacity to
induce trained immunity, BCG is also a strong inducer of T helper type 1 (Th1) responses and
has been employed as a therapeutic alternative in pathologies characterized by exacerbated Th2
responses, such as allergies and asthma (35, 36). Furthermore, due to its strong immunogenicity,
BCG has also been used as an adjuvant to enhance the protective capacity of different types of
vaccines, such as veterinary vaccines (37) and cancer vaccines (38).
Fatty acid
synthesis
Acetyl-CoA Cholesterol cAMP
Mevalonate synthesis
Oxaloacetate
OXPHOS Citrate Acetyl-CoA PKA
ATP TCA
Fumarate cycle
α-KG Glutamine
Succinate HATs
Me1 Me3 Ac
KDM5 TF TF Proinflammatory
Enhancer Promoter genes
Figure 2
Molecular mechanisms of trained immunity. Inducers of trained immunity, such as β-glucan, BCG, uric acid, and several (oxidized)
lipids, activate intracellular metabolic pathways, each via different receptors. The most common pathway is the Akt-HIF1α-mTOR
pathway, which ultimately leads to the upregulation of glycolysis, the TCA cycle, and also the cholesterol synthesis pathway.
Metabolites from these pathways can in turn regulate epigenetic remodeling of histones; for example, acetyl-CoA serves as an acetyl
donor for histone acetyltransferases. Fumarate, on the other hand, a TCA cycle metabolite, inhibits lysine-demethylase KDM5, thereby
increasing histone methylation, another epigenetic mark associated with trained immunity. Catecholamines, via activation of the
beta-adrenergic receptor, are inducing epigenetic and metabolic changes via the cAMP-PKA pathway. Abbreviations: Ac, acetylation;
ATP, adenosine triphosphate; α-KG, α-ketoglutarate; BCG, bacillus Calmette-Guérin; cAMP, cyclic adenosine monophosphate;
GLUT1, glucose transporter 1; HAT, histone acetyl transferase; HIF1α, hypoxia-inducible factor 1 alpha; IGF1R, insulin-like growth
factor 1 receptor; KDM5, lysin-specific demethylase 5; Lp(a), lipoprotein(a); Me1, monomethylation; Me3, trimethylation; mTOR,
mammalian target of rapamycin; NOD2, nucleotide-binding oligomerization domain–containing protein 2; OPV, oral polio vaccine;
oxLDL, oxidized low-density lipoprotein; OXPHOS, oxidative phosphorylation; oxPL, oxidized phospholipid; PKA, protein kinase A;
TCA, tricarboxylic acid; TF, transcription factor; TLR, Toll-like receptor. Figure adapted from image created with BioRender.com.
evidence listed here comes from studies using an in vitro protocol for inducing trained immu-
nity (40), although the majority of findings are confirmed in in vivo animal models or human
studies.
Glycolysis plays a central role in cellular metabolism, and its upregulation is essential during
the activation of macrophages (41). Upregulation of glycolysis results in an increased uptake of
glucose, which is subsequently converted into pyruvate and transformed into lactate, which is then
induces a shift from OXPHOS to glycolysis, also known as the Warburg effect (42). Instead of
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
providing energy by ATP production, there is an anabolic repurposing of TCA metabolites (43).
Glutamine replenishment of the TCA cycle leads to the accumulation of fumarate, which can in-
duce trained immunity by inhibiting the activity of histone demethylase KDM5. This is followed
by persistence of histone methylation and opening of the chromatin that is needed for gene tran-
scription and the increased production of proinflammatory cytokines upon restimulation. Sub-
sequent studies showed that the balance between OXPHOS and glycolysis is dependent on the
specific concentration of β-glucan used in the studies: A lower dose of β-glucan that was also as-
sociated with a hyperresponsive trained phenotype induced an upregulation of both OXPHOS
and glycolysis in trained monocytes (47). This activation of OXPHOS is due to enrichment of the
activating histone modification H3K4me1 on the enhancers of specific metabolic enzymes by the
histone methyltransferase Set7 (47). In BCG- and oxLDL-trained monocytes, a similar upregula-
tion of both OXPHOS and glycolysis is also observed (45, 48). Other intermediates from the TCA
cycle such as α-ketoglutarate, a cofactor for JmjC demethylases (49); succinate; and acetyl-CoA
play important roles in the regulation of inflammation as well. Succinate can stabilize HIF1α,
thereby promoting IL-1β transcription (50). Acetyl-CoA serves as an acetyl donor for histone
acetyl transferases (51), which are upregulated in the context of trained immunity. Examples of
acetylated genes in trained immunity are genes for hexokinase and lactate dehydrogenase, which
in turn promote glycolysis (52). Finally, itaconate, another TCA cycle metabolite, is an important
node between trained immunity and tolerance in that it inhibits inflammation and trained immu-
nity. In this sense, itaconate is able to mimic the tolerizing effects of LPS and inhibit the induction
of trained immunity by β-glucan through the inhibition of mitochondrial metabolism, leading to
decreased responsiveness after secondary stimulation (53).
During the induction of trained immunity by β-glucan, in addition to upregulation of glycolysis
and OXPHOS and repurposing of the TCA cycle, an upregulation of cholesterol synthesis path-
way genes was observed (23). Inhibition of the cholesterol synthesis pathway by statins prevented
the induction of training for β-glucan, BCG, and oxidized LDL, indicating that the cholesterol
synthesis pathway is essential for trained immunity (54). Further characterization of the pathway
revealed that accumulation of mevalonate is responsible for the induction and further amplifica-
tion of trained immunity through the IGF1R-Akt-mTOR pathway. Further evidence for a role for
mevalonate in trained immunity comes from the characterization of monocytes from patients with
deleterious mutations in mevalonate kinase who subsequently accumulate high levels of meval-
onate. These patients with the hyperimmunoglobulin D syndrome (HIDS) are characterized by
recurrent attacks of sterile inflammation and fever, and their monocyte phenotype resembles an
endogenous trained immune phenotype, with increased cytokine production, upregulation of gly-
colysis, and epigenetic changes (54).
and TNF-α. Preincubation of aldosterone-trained cells with a fatty acid synthesis inhibitor before
restimulation prevented augmented cytokine production, showing a role for fatty acid synthesis in
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
the secondary effector function. Monocyte-derived macrophages from patients with hyperaldos-
teronism, who also have increased arterial wall inflammation, showed a similar increase in TNF-α
production upon stimulation compared to trained cells in vitro (57). In contrast, monocytes from
patients with sepsis, characterized by immune tolerance, showed lower proinflammatory cytokine
production accompanied by impaired beta-oxidation.
that was dependent on IFN-γ signaling (67). Bone marrow–derived macrophages of BCG-trained
mice subsequently acquired protection against infection with M. tuberculosis, which was accompa-
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
nied by epigenetic changes such as H3K4me3 and H3K27ac. Transplantation of the bone marrow
of BCG-trained mice into naive mice then conferred protection against reinfection, confirming
long-term reprogramming of the bone marrow. These observations were confirmed in a study
with human volunteers in which vaccination with BCG led to transcriptional and epigenetic repro-
gramming of the bone marrow hematopoietic stem cell progenitors up to 90 days after initial vacci-
nation, resulting in a myeloid bias and increased protection against reinfection (68, 69). Combined
transcriptional and genetic analysis demonstrated an important role of transcription factors such
as hepatic nuclear factors 1a and 1b in this process (69). In Ldlr−/− mice, a four-week Western diet
followed by a four-week chow diet induced similar reprogramming of the bone marrow via tran-
scriptomic and epigenetic changes (70). This was mediated via the NLRP3 inflammasome and IL-
1β and resulted in increased proliferation and enhanced innate immune responses in the mice. The
role of bone marrow reprogramming in other settings of trained immunity is under investigation.
High-cholesterol
Western diet
Vaccine/adjuvant
BCG or β-glucan
Access provided by 2806:2f0:7021:d9b7:f588:453c:1b73:c93e on 10/18/23. For personal use only.
Nonimmune Immune
Peripheral trained immunity
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
Endothelial cells
NK cells
ILCs
Macrophage
Epithelial cells
Enhanced
VSMC DC effector function
Fibroblast
Figure 3
Central versus peripheral trained immunity. Although trained immunity was first discovered in circulating
cells such as monocytes and NK cells, these cells have a short life span; it is shorter than the duration of
trained immunity in humans in vivo. We now know that progenitor cells of the bone marrow can also obtain
a memory phenotype, for example through vaccination or a Western diet, that is then transmitted to their
progeny, the cells in circulation. Eventually, these circulating cells migrate to tissues, where they have
enhanced effector functions as tissue macrophages and NK cells. Additionally, other immune cells in the
tissues, such as DCs and ILCs, are capable of obtaining a trained immunity phenotype as well. Finally,
nonimmune cells such as endothelial cells, epithelial cells, fibroblasts, and VSMCs are described to have
memory capacities as well (78, 79, 94–96). Abbreviations: BCG, bacillus Calmette-Guérin; CMP, common
myeloid progenitor; DC, dendritic cell; GMP, granulocyte-macrophage progenitor; HSC, hematopoietic
stem cell; ILC, innate lymphoid cell; MPP, myeloid progenitor population; NK, natural killer; VSMC,
vascular smooth muscle cell. Figure adapted from image created with BioRender.com.
models of Alzheimer disease and stroke, both innate immune training and tolerance were shown in
brain-resident microglia via epigenetic reprogramming. This memory lasted for at least six months
(76) but is also thought to be transgenerational (77). The important role played by chronic inflam-
mation in neurodegenerative processes makes trained immunity a potential important therapeutic
target in Alzheimer and Parkinson diseases.
The field of DC memory is relatively new, although there are indications that DCs can obtain
a memory phenotype as well. In the lung, local modulation of DCs after the resolution of pneu-
monia resulted in long-term susceptibility to secondary infections, indicating DC tolerance (78).
This was further studied by Hole et al., who showed that DCs can be trained to exhibit enhanced
transcriptional activation upon secondary stimulation, a process driven by epigenetic modifica-
tions (79). The induction of DC memory might be useful in optimizing vaccination strategies
for better clearance of infections. Indeed, one can argue that the adjuvanticity effect on antigen-
presenting cells is a functional program similar to induction of trained immunity.
NK cell training. A combination of IL-12, IL-15, and IL-18 was shown to induce memory-like
NK cells, resulting in effector cells with superior control of leukemic cells (87) and enhanced
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
IFN-γ production (88). This affects tumor immune surveillance of NK cells and might be very
significant for clinical immunotherapy. In the uterus, training of NK cells and subsequent epige-
netic modifications are essential for their function in successful placentation (89). Mechanistically,
both metabolic and epigenetic reprogramming are shown to underlie NK cell memory, similar to
trained immunity in monocytes (90–93).
ILC memory research is still in its infancy, but recent studies show that liver-resident group
1 ILCs (ILC1s) can be primed in a non-antigen-specific fashion, inducing stable transcriptional,
epigenetic, and phenotypic changes up to one month after CMV infection (94). This is driven by
cytokines. ILC2s can also obtain immune memory characteristics upon stimulation with allergens
(95), similar to hapten-induced NK cell memory, as described above. Further research into the
roles of ILC memory in health and disease is warranted.
that involves epigenetic changes (106, 107). Interestingly, not all fibroblasts can obtain a memory
phenotype, as dermal fibroblasts, for example, were not able to undergo trained immunity (108).
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
Further research is warranted to assess the role of trained immunity in (non)immune cells in vivo.
m
im
d
ine
COVID-19 Organ
tra
transplantation
of ts
De
ffec
trim
Mucosal Allergies
tolerance
ial e
Access provided by 2806:2f0:7021:d9b7:f588:453c:1b73:c93e on 10/18/23. For personal use only.
ental
Benefic
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
Vaccinations
unit
Autoinflammatory
disease
y
Recurrent
infections
Neuroinflammation,
Cancer Cerebral small-vessel
disease
Du
al e
ffects
Figure 4
Trained immunity in health and disease. Trained immunity is associated with both health and disease states,
and therefore there is therapeutic potential in either inducing it or inhibiting it. Trained immunity can
induce increased protection against reinfection, which can be used to optimize vaccination strategies or for
protection against, for example, COVID-19. It is important in the induction of mucosal tolerance, and it is
already being used to treat bladder cancer. On the other hand, several chronic inflammatory diseases are
characterized by an inappropriate trained immunity phenotype, such as cardiovascular diseases, allergies,
transplantation rejection, and autoinflammatory diseases. Here, it will be more beneficial to inhibit trained
immunity. Abbreviation: COVID-19, coronavirus disease 2019. Figure adapted from image created with
BioRender.com.
host. In a recent mouse model, β-glucan training led to protection against Leishmania infection
via upregulated expression of IL-1 and IL-32 (68). A similar IL-1β-dependent protection against
M. tuberculosis infection was shown upon β-glucan training in mice, a process that was also me-
diated via reprogramming of the bone marrow. Mice latently infected with gammaherpesvirus or
CMV show antigen-aspecific resistance to infection with Listeria monocytogenes and Yersinia pestis,
a process that was mediated by increased innate immune activation (119). Intraperitoneal stim-
ulation with CpG dinucleotides protected neutropenic mice against intracerebral Escherichia coli
immune-tolerized states.
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
(140) showed that orally delivered β-glucans can aggravate intestinal inflammation. On the other
hand, mice lacking dectin-1, the receptor for β-glucans, also exhibit increased susceptibility to
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
DSS-induced colitis; this also occurs in humans with specific polymorphisms in dectin-1 (141).
Furthermore, prolonged oral treatment of mice with antifungal drugs increases disease severity
in models of chronic colitis as well as chronic allergic airways disease, indicating the importance
of a healthy fungal community in the gut but also highlighting the influence of gut microbiota on
peripheral immune responses and allergic diseases (142). Further research is warranted to better
understand the roles of trained immunity in the gut in both health and disease.
Cardiometabolic Disease
Monocytes and macrophages play a pivotal role in the disease progression of cardiometabolic dis-
orders, such as atherosclerosis, diabetes, and obesity. Several lines of evidence now indicate that
in these chronic inflammatory cardiometabolic diseases, trained immunity can play a detrimental
role (143–145). First of all, trained immunity may explain the known epidemiological association
between the infectious burden and increased cardiovascular disease risk, as discussed in Refer-
ence 146. Secondly, in addition to microbial stimuli, several endogenous atherogenic stimuli are
able to induce trained innate immunity in monocytes in vitro, such as oxLDL (25), Lp(a) (26),
catecholamines (31), and aldosterone (32).
Furthermore, hyperglycemia can induce innate immune memory via long-term epigenetic
changes in both monocytes and endothelial cells (101, 147, 148). In vitro, training of mono-
cytes with oxLDL in high-glucose conditions boosted cytokine production capacity compared
to oxLDL training in normoglycemic conditions (46). Recently, it was shown that diabetes also
induces enhanced proliferation of HSPCs in the bone marrow and a myeloid bias, leading to
augmented circulating myeloid cell numbers and enhanced atherosclerosis (149, 150). This was
regulated via the interaction between endothelial cells and bone marrow progenitor cells. Fur-
thermore, in an atherosclerosis-prone Ldlr−/− mouse model, a four-week Western diet induced
long-term reprogramming of cells of the myeloid lineage, a process that originated in changes in
bone marrow progenitor cells (70). These inflammatory changes persisted for at least four weeks
after reversal of the diet to normal chow.
In human observational studies, monocytes from patients with symptomatic atherosclerosis
exhibited a trained immunity phenotype with increased proinflammatory cytokine production,
accompanied by metabolic and epigenetic changes compared to healthy controls (61). This was
confirmed in another study by Shirai et al. (151), who showed that monocytes from patients
with coronary atherosclerosis have a proinflammatory phenotype and enhanced glycolytic activ-
ity, even after differentiation into macrophages in vitro. In treatment-naive patients with familial
hypercholesterolemia, monocytes showed a trained immunity phenotype, in contrast to healthy
And indeed, trained immunity was confirmed by epigenetic analysis showing long-term enriched
H3K4me3 in promoter regions of proinflammatory genes (31).
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
In a small proof-of-principle study, it was shown that lifestyle intervention might be an inter-
esting therapeutic target for decreasing inflammation in patients at risk for cardiovascular diseases.
In a group of obese and hypertensive patients, sedentary behavior was reduced over a 16-week pe-
riod, which induced a reduction in cytokine production capacity upon secondary stimulation. This
was accompanied by anti-inflammatory changes in intracellular metabolism, such as a decrease in
glycolysis and OXPHOS. It would be interesting to study whether these observed changes were
also accompanied by long-term epigenetic changes. Lifestyle changes are increasingly seen as a
potential therapy for cardiovascular disease and the underlying innate immune memory, and it
will be interesting to study the effects on trained immunity (154).
Neurodegenerative Disease
Microglia are the resident macrophages of the central nervous system and play a vital role in nor-
mal brain function and pathologies. Microglial immune memory has now been suggested as an
underlying cause of neuroinflammation (75). As mentioned above, microglia can be trained, so as
to result in metabolic and epigenetic changes. This was first shown by Wendeln et al. (76), who
showed that both immune training and tolerance can be induced in brain-resident macrophages
via changes in H3K4me1 and H3K27ac, which lasted for at least six months. The imprinted mem-
ory, in the case of trained immunity induction, resulted in exacerbated cerebral inflammation and
beta-amyloidosis in a mouse model of Alzheimer disease. When tolerance was induced, on the
other hand, disease pathology was alleviated. Similar context-dependent observations in the mi-
croglia were made by Datta et al. (155), who showed that this process was dependent on histone
acetyltransferases HDAC1/2. We now know that microglial immune memory can be induced by
both microbial (156) and endogenous stimuli such as stress (157), and it is thought to be trans-
generational (77) or at least long-term, with an important role for IL-1β in early life (158–160).
In the brain, vascular diseases such as arteriolosclerosis underlie cognitive decline and demen-
tia. Here, trained immunity might also contribute to the pathology of disease. In an elderly cohort
of individuals with cerebral small-vessel disease, monocytes showed a proinflammatory phenotype
and disease progression associated with increased cytokine production capacity (161).
with soluble uric acid, leading to increased proinflammatory cytokine production upon restimula-
tion, which is mediated via epigenetic and metabolic reprogramming (29, 30). In murine autoim-
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
mune arthritis, myeloid skewing occurs in HSPCs, resulting in increased inflammation. Whether
this is mediated via metabolic and epigenetic changes, however, requires further investigation
(165). The therapeutic potential of inducing trained immunity or tolerance in autoinflammatory
diseases was elegantly studied by Jeljeli et al. (166). They showed that the induction of LPS tol-
erance alleviated fibrosis in a mouse model of systemic sclerosis, whereas induction of training by
BCG exacerbated disease progression. The induction of two opposing immune programs resulted
in opposing disease states, collectively mediated via innate immune activation (166).
Recently, a potential role for trained immunity and epigenetic changes was hypothesized to
underlie food allergy (167). Allergic children have an increased innate immune response com-
pared to nonallergic children (168) that is already present in early life (169). Monocytes from
children with food allergy show changes in innate immune function resembling a trained pheno-
type (170), although the underlying epigenetic changes and memory upon removal of the stimulus
are still under investigation. The development of food allergy later in life, however, can be pre-
dicted from monocyte immune responses at birth, indicating some sort of programmed phenotype
(171). However, the induction of a balanced state of trained immunity has also been suggested to
prevent (food) allergy. Several studies have shown the beneficial effects of vaccines on the develop-
ment of (food) allergies (172). Prior BCG vaccination was shown to suppress allergic sensitization
in an animal model of allergic airway disease (173). This was confirmed in another study in which
gammaherpesvirus infection provided protection against allergic asthma via activation of the in-
nate immune system (71). In a similar fashion, whole-cell pertussis vaccination decreased the risk
of IgE-mediated food allergy (174). Large randomized clinical trials are being conducted to study
whether BCG vaccination at birth prevents the development of food allergy in countries with low
infectious burden (175). To improve prevention or treatment strategies, more research is needed
to understand the difference between the potential of trained immunity to prevent food allergy
and the detrimental effects of trained immunity once the allergy is established. On the other hand,
these data could also mirror different programs of trained immunity. Indeed, trained immunity
is a means by which innate immune cells gain a different function through long-term epigenetic
changes, rather than a specific transcriptional program. Some of these changes can be protective in
certain circumstances (a protective trained immunity program), while other changes can represent
the molecular substrate of disease (a deleterious trained immunity program).
Organ Transplantation
The role of innate immune cells in organ transplantation is poorly studied, and the focus has
long been on the role of the adaptive immune system. However, recent advances have shown
secondary effector function upon a first stimulation, but we are increasingly realizing that the un-
derlying mechanisms and subsequent secondary phenotype can differ for each stimulus or in dif-
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
ferent health or disease states. This might also impact the way we need to boost trained immunity
to achieve health, such as by using nonspecific effects of vaccines, or inhibit it in chronic inflam-
matory diseases. We have only begun to unravel the role of trained immunity in (inflamm)aging.
Further research is therefore warranted to study the diversity of stimuli and disease pathologies,
the accompanying intracellular metabolic and epigenetic changes that are induced, and the sub-
sequent increased inflammatory response upon secondary stimulation, in order to optimize the
therapeutic potential that trained immunity encompasses (178).
Ideal targets for designing novel approaches for therapeutics are the central mechanisms un-
derlying trained immunity, such as metabolic and epigenetic changes. Both these processes are
amenable for therapeutic targeting, as has been suggested in several reviews (178–180). Epige-
netic enzymes such as histone demethylase KDM5 and histone methyltransferase Set7 are known
to regulate trained immunity (43, 47), and Set7 is even implicated in bone marrow reprogramming
by β-glucan. These two enzymes are therefore interesting targets for preventing trained immu-
nity. Another group of potential epigenetic modifiers is bromodomain and extraterminal domain
inhibitors (BETis). BETis are increasingly recognized for their therapeutic potential in inflam-
mation (181), as they interfere with the recognition of acetylated histone marks, areas that could
well be primed by trained immunity for increased transcription upon secondary stimulation. An
example of a BETi that is under investigation is iBET151. iBET151 was shown to suppress the
immune response during a fungal infection and prevent the induction of trained immunity (182).
iBET151 was also shown to reduce chronic inflammation in rheumatoid arthritis and diabetes
(183, 184). Several other BETis are now studied for their anti-inflammatory potential. Next to
dampening the effects of trained immunity and the written epigenetic marks, it would be interest-
ing to reverse trained immunity and remove the accompanying epigenetic marks. Would simply
removing the stimulus for long enough be sufficient to reverse trained immunity and the accom-
panying epigenetic changes? Further research is much warranted to study reversal of a trained
phenotype and the enzymes that remove histone marks.
In addition to epigenetic enzymes, metabolic changes are the second interesting therapeu-
tic target in trained immunity. Even though different metabolic pathways have been described
for different trained immunity stimuli, most stimuli share the activation of the PI3K/Akt/mTOR
pathway and subsequent induction of glycolysis (Figure 2), which would therefore be an interest-
ing first target. Several small molecules have been shown to reduce glycolysis in vivo, for example
3-PO, a partial glycolysis inhibitor that can significantly reduce atherosclerosis development in an
atherosclerotic mouse model (185). Interestingly, glycolysis is not completely blocked by 3-PO,
thereby guaranteeing normal immune function of the innate immune cell when needed for fight-
ing infections. Secondly, rapamycin-loaded HDL-nanoparticles have been shown to specifically
tory agent. This interaction can either induce long-term improvement of host defense and pro-
tection against heterologous infections when induced adaptively by infections or vaccinations or
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
DISCLOSURE STATEMENT
M.G.N. and L.A.B.J. are scientific founders of TTxD and are owners of two patents on modulation
of trained immunity. L.A.B.J. is an SAB member of Olatec Pharmaceuticals.
ACKNOWLEDGMENTS
This work was supported by IN-CONTROL CVON grants (CVON2012-03 and CVON2018-
27 to N.P.R., M.G.N., and L.A.B.J.). S.B. is supported by the Dutch Heart Foundation (Dekker
grant 2018-T028). M.G.N. is supported by a European Research Council (ERC) Advanced Grant
(ERC 833247) and a Netherlands Organization for Scientific Research Spinoza Grant (NWO SPI
94-212). L.A.B.J. is supported by a Competitiveness Operational Program grant of the Romanian
Ministry of European Funds (HINT, ID P_37_762; MySMIS 103587). N.P.R. is recipient of a
grant of the ERA-CVD Joint Transnational Call 2018, which is supported by the Dutch Heart
Foundation ( JTC2018, project MEMORY; 2018T093).
LITERATURE CITED
1. Naeslund C. 1931. Résultats des expériences de vaccination par le BCG poursuivies dans le Norrbotten
(Suède) (Septembre 1927–Décembre 1931). Rev. Tuberc. 12:617–36
2. Netea M, Quintin J, van der Meer J. 2011. Trained immunity: a memory for innate host defense. Cell
Host Microbe 9(5):355–61
3. Durrant WE, Dong X. 2004. Systemic acquired resistance. Annu. Rev. Phytopathol. 42(1):185–209
4. Fu ZQ, Dong X. 2013. Systemic acquired resistance: turning local infection into global defense. Annu.
Rev. Plant Biol. 64(1):839–63
5. Reimer-Michalski EM, Conrath U. 2016. Innate immune memory in plants. Semin. Immunol. 28(4):319–
27
6. Pham LN, Dionne MS, Shirasu-Hiza M, Schneider DS. 2007. A specific primed immune response in
Drosophila is dependent on phagocytes. PLOS Pathog. 3(3):e26
7. Tate AT, Andolfatto P, Demuth JP, Graham AL. 2017. The within-host dynamics of infection in trans-
generationally primed flour beetles. Mol. Ecol. 26(14):3794–807
13. Kleinnijenhuis J, Quintin J, Preijers F, Joosten LAB, Ifrim DC, et al. 2012. Bacille Calmette-Guerin
induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands
in sterile inflammation. Nat. Immunol. 14(8):812–20
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
56. Mitroulis I, Ruppova K, Wang B, Chen L-S, Grzybek M, et al. 2018. Modulation of myelopoiesis pro-
genitors is an integral component of trained immunity. Cell 172(1–2):147–61.e12
57. van der Heijden CD, Smeets EM, Aarntzen EH, Noz MP, Monajemi H, et al. 2020. Arterial wall inflam-
mation and increased hematopoietic activity in patients with primary aldosteronism. J. Clin. Endocrinol.
Metab. 105(5):e1967–80
58. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, et al. 2020. Defining trained
immunity and its role in health and disease. Nat. Rev. Immunol. 20(6):375–88
59. Novakovic B, Habibi E, Wang SY, Arts RJW, Davar R, et al. 2016. β-Glucan reverses the epigenetic
state of LPS-induced immunological tolerance. Cell 167(5):1354–68.e14
60. Bekkering S, Stiekema LCA, Bernelot Moens S, Verweij SL, Novakovic B, et al. 2019. Treatment with
statins does not revert trained immunity in patients with familial hypercholesterolemia. Cell Metab.
30(1):1–2
61. Bekkering S, van den Munckhof I, Nielen T, Lamfers E, Dinarello C, et al. 2016. Innate immune cell
activation and epigenetic remodeling in symptomatic and asymptomatic atherosclerosis in humans in
vivo. Atherosclerosis 254:228–36
62. Fok ET, Davignon L, Fanucchi S, Mhlanga MM. 2019. The lncRNA connection between cellular
metabolism and epigenetics in trained immunity. Front. Immunol. 29(9):3184
63. Fanucchi S, Mhlanga MM. 2019. Lnc-ing trained immunity to chromatin architecture. Front. Cell Dev.
Biol. 7:2
64. Verma D, Parasa VR, Raffetseder J, Martis M, Mehta RB, et al. 2017. Anti-mycobacterial activity cor-
relates with altered DNA methylation pattern in immune cells from BCG-vaccinated subjects. Sci. Rep.
7(1):12305
65. Das J, Verma D, Gustafsson M, Lerm M. 2019. Identification of DNA methylation patterns predisposing
for an efficient response to BCG vaccination in healthy BCG-naïve subjects. Epigenetics 14(6):589–601
66. Kleinnijenhuis J, Quintin J, Preijers F, Joosten LAB, Jacobs C, et al. 2014. BCG-induced trained immu-
nity in NK cells: role for non-specific protection to infection. Clin. Immunol. 155(2):213–19
67. Kaufmann E, Sanz J, Dunn JL, Khan N, Mendonça LE, et al. 2018. BCG educates hematopoietic stem
cells to generate protective innate immunity against tuberculosis. Cell 172(1–2):176–90.e19
68. dos Santos JC, Barroso de Figueiredo AM, Teodoro Silva MV, Cirovic B, de Bree LCJ, et al. 2019. β-
Glucan-induced trained immunity protects against Leishmania braziliensis infection: a crucial role for
IL-32. Cell Rep. 28(10):2659–72.e6
69. Cirovic B, de Bree LCJ, Groh L, Blok BA, Chan J, et al. 2020. BCG vaccination in humans elicits trained
immunity via the hematopoietic progenitor compartment. Cell Host Microbe 28(2):322–34.e5
70. Christ A, Günther P, Lauterbach MAR, Duewell P, Biswas D, et al. 2018. Western diet triggers NLRP3-
dependent innate immune reprogramming. Cell 172(1–2):162–75.e14
71. Machiels B, Dourcy M, Xiao X, Javaux J, Mesnil C, et al. 2017. A gammaherpesvirus provides protection
against allergic asthma by inducing the replacement of resident alveolar macrophages with regulatory
monocytes. Nat. Immunol. 18(12):1310–20
10(4):217–24
78. Roquilly A, McWilliam HEG, Jacqueline C, Tian Z, Cinotti R, et al. 2017. Local modulation of antigen-
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
presenting cell development after resolution of pneumonia induces long-term susceptibility to secondary
infections. Immunity 47(1):135–47.e5
79. Hole CR, Wager CML, Castro-Lopez N, Campuzano A, Cai H, et al. 2019. Induction of memory-like
dendritic cell responses in vivo. Nat. Commun. 10(1):2955
80. Placek K, Schultze JL, Netea MG. 2019. Immune memory characteristics of innate lymphoid cells. Curr.
Opin. Infect. Dis. 32(3):196–203
81. O’Sullivan TE, Sun JC, Lanier LL. 2015. Natural killer cell memory. Immunity 43(4):634–45
82. Dou Y, Fu B, Sun R, Li W, Hu W, et al. 2015. Influenza vaccine induces intracellular immune memory
of human NK cells. PLOS ONE 10(3):e0121258
83. Nielsen CM, White MJ, Bottomley C, Lusa C, Rodríguez-Galán A, et al. 2015. Impaired NK cell re-
sponses to pertussis and H1N1 influenza vaccine antigens in human cytomegalovirus-infected individ-
uals. J. Immunol. 194(10):4657–67
84. McCall MBB, Roestenberg M, Ploemen I, Teirlinck A, Hopman J, et al. 2010. Memory-like IFN-γ
response by NK cells following malaria infection reveals the crucial role of T cells in NK cell activation
by P. falciparum. Eur. J. Immunol. 40(12):3472–77
85. O’Leary JG, Goodarzi M, Drayton DL, von Andrian UH. 2006. T cell- and B cell-independent adaptive
immunity mediated by natural killer cells. Nat. Immunol. 7(5):507–16
86. Sun JC, Ma A, Lanier LL. 2009. Cutting edge: IL-15-independent NK cell response to mouse cy-
tomegalovirus infection. J. Immunol. 183(5):2911–14
87. Romee R, Maximillian R, Berrien-Elliott MM, Wagner JA, Jewell BA, et al. 2015. Human cytokine-
induced memory-like NK cells exhibit in vivo anti-leukemia activity in xenografted NSG mice and in
patients with acute myeloid leukemia (AML). Blood 126(23):101
88. Cooper MA, Elliott JM, Keyel PA, Yang L, Carrero JA, Yokoyama WM. 2009. Cytokine-induced
memory-like natural killer cells. PNAS 106(6):1915–19
89. Gamliel M, Goldman-Wohl D, Isaacson B, Gur C, Stein N, et al. 2018. Trained memory of human
uterine NK cells enhances their function in subsequent pregnancies. Immunity 48(5):951–62.e5
90. Donnelly RP, Loftus RM, Keating SE, Liou KT, Biron CA, et al. 2014. mTORC1-dependent metabolic
reprogramming is a prerequisite for NK cell effector function. J. Immunol. 193(9):4477–84
91. Schlums H, Cichocki F, Tesi B, Theorell J, Beziat V, et al. 2015. Cytomegalovirus infection drives
adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity
42(3):443–56
92. Cichocki F, Wu CY, Zhang B, Felices M, Tesi B, et al. 2018. ARID5B regulates metabolic programming
in human adaptive NK cells. J. Exp. Med. 215(9):2379–95
93. Rasid O, Chevalier C, Camarasa TMN, Fitting C, Cavaillon JM, Hamon MA. 2019. H3K4me1 supports
memory-like NK cells induced by systemic inflammation. Cell Rep. 29(12):3933–45.e3
94. Weizman O, Song E, Adams NM, Hildreth AD, Riggan L, et al. 2019. Mouse cytomegalovirus-
experienced ILC1s acquire a memory response dependent on the viral glycoprotein m12. Nat. Immunol.
20(8):1004–11
101. El-Osta A, Brasacchio D, Yao D, Pocai A, Jones PL, et al. 2008. Transient high glucose causes persis-
tent epigenetic changes and altered gene expression during subsequent normoglycemia. J. Exp. Med.
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
205(10):2409–17
102. Schnitzler JG, Hoogeveen RM, Ali L, Prange KHM, Waissi F, et al. 2020. Atherogenic lipoprotein(a)
increases vascular glycolysis, thereby facilitating inflammation and leukocyte extravasation. Circ. Res.
126(10):1346–59
103. Schnack L, Sohrabi Y, Lagache SMM, Kahles F, Bruemmer D, et al. 2019. Mechanisms of trained innate
immunity in oxLDL primed human coronary smooth muscle cells. Front. Immunol. 10:13
104. Ospelt C, Reedquist KA, Gay S, Tak PP. 2011. Inflammatory memories: Is epigenetics the missing link
to persistent stromal cell activation in rheumatoid arthritis? Autoimmun. Rev. 10(9):519–24
105. Ara T, Kurata K, Hirai K, Uchihashi T, Uematsu T, et al. 2009. Human gingival fibroblasts are critical
in sustaining inflammation in periodontal disease. J. Periodontal Res. 44(1):21–27
106. Sohn C, Lee A, Qiao Y, Loupasakis K, Ivashkiv LB, Kalliolias GD. 2015. Prolonged tumor necrosis
factor α primes fibroblast-like synoviocytes in a gene-specific manner by altering chromatin. Arthritis
Rheumatol. 67(1):86–95
107. Klein K, Frank-Bertoncelj M, Karouzakis E, Gay RE, Kolling C, et al. 2017. The epigenetic architecture
at gene promoters determines cell type-specific LPS tolerance. J. Autoimmun. 83:122–33
108. Crowley T, O’Neil JD, Adams H, Thomas AM, Filer A, et al. 2017. Priming in response to pro-
inflammatory cytokines is a feature of adult synovial but not dermal fibroblasts. Arthritis Res. Ther.
19(1):35
109. Stensballe LG, Nante E, Jensen IP, Kofoed PE, Poulsen A, et al. 2005. Acute lower respiratory tract
infections and respiratory syncytial virus in infants in Guinea-Bissau: a beneficial effect of BCG vacci-
nation for girls; community based case-control study. Vaccine 23(10):1251–57
110. Aaby P, Roth A, Ravn H, Napirna BM, Rodrigues A, et al. 2011. Randomized trial of BCG vaccination
at birth to low-birth-weight children: beneficial nonspecific effects in the neonatal period? J. Infect. Dis.
204(2):245–52
111. Aaby P, Mogensen SW, Rodrigues A, Benn CS. 2018. Evidence of increase in mortality after the intro-
duction of diphtheria-tetanus-pertussis vaccine to children aged 6–35 months in Guinea-Bissau: a time
for reflection? Front. Public Health 6:79
112. Nemes E, Geldenhuys H, Rozot V, Rutkowski KT, Ratangee F, et al. 2018. Prevention of M. tuberculosis
infection with H4:IC31 vaccine or BCG revaccination. N. Engl. J. Med. 379(2):138–49
113. Arts RJW, Moorlag SJCFM, Novakovic B, Li Y, Wang SY, et al. 2018. BCG vaccination protects against
experimental viral infection in humans through the induction of cytokines associated with trained im-
munity. Cell Host Microbe 23(1):89–100.e5
114. Blok BA, Arts RJW, van Crevel R, Aaby P, Joosten LAB, et al. 2020. Differential effects of BCG vaccine
on immune responses induced by vi polysaccharide typhoid fever vaccination: an explorative randomized
trial. Eur. J. Clin. Microbiol. Infect. Dis. 39(6):1177–84
115. Blok BA, de Bree LCJ, Diavatopoulis DA, Langereis JD, Joosten LA, et al. 2020. Interacting, nonspecific,
immunological effects of Bacille Calmette-Guérin and tetanus-diphtheria-pertussis inactivated polio
vaccinations: an explorative, randomized trial. Clin. Infect. Dis. 70(3):455–63
J. Neuroinflamm. 11:14
121. Li R, Lim A, Phoon MC, Narasaraju T, Ng JKW, et al. 2010. Attenuated Bordetella pertussis protects
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
against highly pathogenic influenza A viruses by dampening the cytokine storm. J. Virol. 84(14):7105–
13
122. van der Poll T, Opal SM. 2008. Host-pathogen interactions in sepsis. Lancet Infect. Dis. 8(1):32–43
123. Cheng SC, Scicluna BP, Arts RJW, Gresnigt MS, Lachmandas E, et al. 2016. Broad defects in the energy
metabolism of leukocytes underlie immunoparalysis in sepsis. Nat. Immunol. 17(4):406–13
124. Netea MG, Giamarellos-Bourboulis EJ, Domínguez-Andrés J, Curtis N, van Crevel R, et al. 2020.
Trained immunity: a tool for reducing susceptibility and severity of SARS-CoV-2 infection. Cell
181(5):969–77
125. Gursel M, Gursel I. 2020. Is global BCG vaccination-induced trained immunity relevant to the progres-
sion of SARS-CoV-2 pandemic? Allergy 75(7):1815–19
126. Faust L, Huddart S, MacLean E, Svadzian A. 2020. Universal BCG vaccination and protec-
tion against COVID-19: critique of an ecological study. Nature Research Microbiology Community.
https://naturemicrobiologycommunity.nature.com/users/36050-emily-maclean/posts/64892-
universal-bcg-vaccination-and-protection-against-covid-19-critique-of-an-ecological-study
127. O’Neill LAJ, Netea MG. 2020. BCG-induced trained immunity: Can it offer protection against
COVID-19? Nat. Rev. Immunol. 20(6):335–37
128. Curtis N, Sparrow A, Ghebreyesus TA, Netea MG. 2020. Considering BCG vaccination to reduce the
impact of COVID-19. Lancet 395(10236):1545–46
129. Aaby P, Benn CS, Flanagan KL, Klein SL, Kollmann TR, et al. 2020. The non-specific and sex-
differential effects of vaccines. Nat. Rev. Immunol. 20(8):464–70
130. Sylvester RJ, van der Meijden APM, Lamm DL. 2002. Intravesical bacillus Calmette-Guerin reduces the
risk of progression in patients with superficial bladder cancer: a meta-analysis of the published results
of randomized clinical trials. J. Urol. 168(5):1964–70
131. Morton DL, Eilber FR, Holmes EC, Hunt JS, Ketcham AS, et al. 1974. BCG immunotherapy of ma-
lignant melanoma: summary of a seven year experience. Ann. Surg. 180(4):635–43
132. Sokal JE, Aungst CW, Snyderman M, Sokal PJ. 1974. Delay in progression of malignant lymphoma after
BCG vaccination. N. Engl. J. Med. 291(23):1226–30
133. Buffen K, Oosting M, Quintin J, Ng A, Kleinnijenhuis J, et al. 2014. Autophagy controls BCG-induced
trained immunity and the response to intravesical BCG therapy for bladder cancer. PLOS Pathog.
10(10):e1004485
134. Kar UK, Joosten LAB. 2020. Training the trainable cells of the immune system and beyond. Nat.
Immunol. 21(2):115–19
135. Netea MG, Joosten LAB, van der Meer JWM. 2017. Hypothesis: stimulation of trained immunity as
adjunctive immunotherapy in cancer. J. Leukoc. Biol. 102(6):1323–32
136. Owens BMJ. 2015. Inflammation, innate immunity, and the intestinal stromal cell niche: opportunities
and challenges. Front. Immunol. 6:319
137. Foster SL, Hargreaves DC, Medzhitov R. 2007. Gene-specific control of inflammation by TLR-induced
chromatin modifications. Nature 447(7147):972–78
144. Bekkering S, Saner C, Riksen NP, Netea MG, Sabin MA, et al. 2020. Trained immunity: linking obesity
and cardiovascular disease across the life-course? Trends Endocrinol. Metab. 31(5):378–89
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
145. Thiem K, Stienstra R, Riksen NP, Keating ST. 2019. Trained immunity and diabetic vascular disease.
Clin. Sci. 133(2):195–203
146. Leentjens J, Bekkering S, Joosten LAB, Netea MG, Burgner DP, Riksen NP. 2018. Trained innate immu-
nity as a novel mechanism linking infection and the development of atherosclerosis. Circ. Res. 122(5):664–
69
147. Brasacchio D, Okabe J, Tikellis C, Balcerczyk A, George P, et al. 2009. Hyperglycemia induces a dynamic
cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic
marks that coexist on the lysine tail. Diabetes 58(5):1229–36
148. Yun JM, Jialal I, Devaraj S. 2011. Epigenetic regulation of high glucose-induced proinflammatory cy-
tokine production in monocytes by curcumin. J. Nutr. Biochem. 22(5):450–58
149. Hoyer FF, Zhang X, Coppin E, Vasamsetti SB, Modugu G, et al. 2020. Bone marrow endothelial cells
regulate myelopoiesis in diabetes mellitus. Circulation 142(3):244–58
150. Barman PK, Urao N, Koh TJ. 2019. Diabetes induces myeloid bias in bone marrow progenitors associ-
ated with enhanced wound macrophage accumulation and impaired healing. J. Pathol. 249(4):435–46
151. Shirai T, Nazarewicz RR, Wallis BB, Yanes RE, Watanabe R, et al. 2016. The glycolytic enzyme PKM2
bridges metabolic and inflammatory dysfunction in coronary artery disease. J. Exp. Med. 213(3):337–54
152. Viney NJ, van Capelleveen JC, Geary RS, Xia S, Tami JA, et al. 2016. Antisense oligonucleotides tar-
geting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-
controlled, dose-ranging trials. Lancet 388(10057):2239–53
153. Schnitzler JG, Poels K, Stiekema LCA, Yeang C, Tsimikas S, et al. 2020. Short-term regulation of
hematopoiesis by lipoprotein(a) results in the production of pro-inflammatory monocytes. Int. J. Cardiol.
315:81–85
154. Schloss MJ, Swirski FK, Nahrendorf M. 2020. Modifiable cardiovascular risk, hematopoiesis, and innate
immunity. Circ. Res. 126(9):1242–59
155. Datta M, Staszewski O, Raschi E, Frosch M, Hagemeyer N, et al. 2018. Histone deacetylases 1 and
2 regulate microglia function during development, homeostasis, and neurodegeneration in a context-
dependent manner. Immunity 48(3):514–29.e6
156. Püntener U, Booth SG, Perry VH, Teeling JL. 2012. Long-term impact of systemic bacterial infection
on the cerebral vasculature and microglia. J. Neuroinflamm. 9:146
157. Ramirez K, Shea DT, McKim DB, Reader BF, Sheridan JF. 2015. Imipramine attenuates neuroinflam-
matory signaling and reverses stress-induced social avoidance. Brain Behav. Immun. 46:212–20
158. Knuesel I, Chicha L, Britschgi M, Schobel SA, Bodmer M, et al. 2014. Maternal immune activation and
abnormal brain development across CNS disorders. Nat. Rev. Neurol. 10(11):643–60
159. Bilbo SD, Biedenkapp JC, Der-Avakian A, Watkins LR, Rudy JW, Maier SF. 2005. Neonatal infection-
induced memory impairment after lipopolysaccharide in adulthood is prevented via caspase-1 inhibition.
J. Neurosci. 25(35):8000–9
160. Williamson LL, Sholar PW, Mistry RS, Smith SH, Bilbo SD. 2011. Microglia and memory: modulation
by early-life infection. J. Neurosci. 31(43):15511–21
167. Imran S, Neeland MR, Shepherd R, Messina N, Perrett KP, et al. 2020. A potential role for epigenetically
mediated trained immunity in food allergy. iScience 23(6):101171
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
168. Tulic MK, Hodder M, Forsberg A, McCarthy S, Richman T, et al. 2011. Differences in innate immune
function between allergic and nonallergic children: new insights into immune ontogeny. J. Allergy Clin.
Immunol. 127(2):470–78.e1
169. Neeland MR, Koplin JJ, Dang TD, Dharmage SC, Tang ML, et al. 2018. Early life innate immune
signatures of persistent food allergy. J. Allergy Clin. Immunol. 142(3):857–64.e3
170. Neeland MR, Andorf S, Manohar M, Dunham D, Lyu SC, et al. 2020. Mass cytometry reveals cellular
fingerprint associated with IgE+ peanut tolerance and allergy in early life. Nat. Commun. 11(1):1091
171. Zhang Y, Collier F, Naselli G, Saffery R, Tang ML, et al. 2016. Cord blood monocyte-derived inflamma-
tory cytokines suppress IL-2 and induce nonclassic “TH 2-type” immunity associated with development
of food allergy. Sci. Transl. Med. 8(321):321ra8
172. Arnoldussen DL, Linehan M, Sheikh A. 2011. BCG vaccination and allergy: a systematic review and
meta-analysis. J. Allergy Clin. Immunol. 127(1):246–53
173. Herz U, Gerhold K, Grüber C, Braun A, Wahn U, et al. 1998. BCG infection suppresses allergic sen-
sitization and development of increased airway reactivity in an animal model. J. Allergy Clin. Immunol.
102(5):867–74
174. Estcourt MJ, Campbell DE, Gold MS, Richmond P, Allen KJ, et al. 2019. Whole-cell pertussis vac-
cination and decreased risk of IgE-mediated food allergy: a nested case-control study. J. Allergy Clin.
Immunol. Pract. 8(6):2004–14
175. Messina NL, Gardiner K, Donath S, Flanagan K, Ponsonby AL, et al. 2019. Study protocol for the Mel-
bourne Infant Study: BCG for Allergy and Infection Reduction (MIS BAIR), a randomised controlled
trial to determine the non-specific effects of neonatal BCG vaccination in a low-mortality setting. BMJ
Open 9(12):e032844
176. Ochando J, Fayad ZA, Madsen JC, Netea MG, Mulder WJM. 2020. Trained immunity in organ trans-
plantation. Am. J. Transplant. 20(1):10–18
177. Braza MS, van Leent MMT, Lameijer M, Sanchez-Gaytan BL, Arts RJW, et al. 2018. Inhibiting in-
flammation with myeloid cell-specific nanobiologics promotes organ transplant acceptance. Immunity
49(5):819–28.e6
178. Mulder WJM, Ochando J, Joosten LAB, Fayad ZA, Netea MG. 2019. Therapeutic targeting of trained
immunity. Nat. Rev. Drug Discov. 18(7):553–66
179. van der Heijden CDCC, Noz MP, Joosten LAB, Netea MG, Riksen NP, Keating ST. 2018. Epigenetics
and trained immunity. Antioxid. Redox Signal. 29(11):1023–40
180. Rodriguez RM, Suarez-Alvarez B, Lopez-Larrea C. 2019. Therapeutic epigenetic reprogramming of
trained immunity in myeloid cells. Trends Immunol. 40(1):66–80
181. Nicodeme E, Jeffrey KL, Schaefer U, Beinke S, Dewell S, et al. 2010. Suppression of inflammation by a
synthetic histone mimic. Nature 468(7327):1119–23
182. Domínguez-Andrés J, Ferreira AV, Jansen T, Smithers N, Prinjha RK, et al. 2019. Bromodomain in-
hibitor I-BET151 suppresses immune responses during fungal-immune interaction. Eur. J. Immunol.
49(11):2044–50
Annual Review of
Immunology
Contents
Access provided by 2806:2f0:7021:d9b7:f588:453c:1b73:c93e on 10/18/23. For personal use only.
Annu. Rev. Immunol. 2021.39:667-693. Downloaded from www.annualreviews.org
viii
IY39_FrontMatter ARjats.cls March 30, 2021 15:54
Contents ix
IY39_FrontMatter ARjats.cls March 30, 2021 15:54
Indexes
Cumulative Index of Contributing Authors, Volumes 29–39 p p p p p p p p p p p p p p p p p p p p p p p p p p p 819
Cumulative Index of Article Titles, Volumes 29–39 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 826
Errata
An online log of corrections to Annual Review of Immunology articles may be found at
http://www.annualreviews.org/errata/immunol
x Contents