Postinfectious Epigenetic Immune Modifications - A Double-Edged Sword

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Dan L. Longo, M.D., Editor

Postinfectious Epigenetic Immune


Modifications — A Double-Edged Sword
Andrew R. DiNardo, M.D., Mihai G. Netea, M.D., Ph.D.,
and Daniel M. Musher, M.D.​​

T
he mechanisms by which antigens specifically sensitize lympho- From the Global Tuberculosis Program,
cytes to induce immunity are well recognized. The past decade has wit- William T. Shearer Center for Human Im­
munobiology, Texas Children’s Hospital
nessed an increased awareness of the importance of specific sensitization (A.R.D.), the Immigrant and Global Health
of monocytes, natural killer cells, and dendritic cells. This sensitization results Program, Department of Pediatrics (A.R.D.),
from long-lived epigenetic changes that profoundly enhance or suppress immune and the Departments of Medicine and
Molecular Virology and Microbiology
responses. Stated simply, immunologic stimuli — infections or immunizations (D.M.M.), Baylor College of Medicine,
— can have long-term consequences for the immune responsiveness of the host. and the Medical Care Line, Infectious
Our purpose is to describe the mechanisms by which these epigenetic effects come Disease Section, Michael E. DeBakey Vet­
erans Affairs Medical Center (D.M.M.)
about and their effects on responses to subsequent infection and vaccination. — all in Houston; the Department of In­
Epigenetic therapies have improved clinical outcomes in patients with cancer and ternal Medicine and Radboud Center for
are on the verge of regular clinical applicability in the management of infections Infectious Diseases, Radboud University
Medical Center, Nijmegen, the Nether­
and vaccine administration. lands (M.G.N.); and the Department of
Genomics and Immunoregulation, Life
and Medical Sciences Institute, Univer­
Specifici t y of Humor a l a nd Cel l -Medi ated Im muni t y sity of Bonn, Bonn, Germany (M.G.N.).
Address reprint requests to Dr. DiNardo
In the 1890s, Klemperer and Klemperer showed that inoculating rabbits with at dinardo@bcm.edu.
killed pneumococci protected them against pneumococcal challenge. Infusing N Engl J Med 2021;384:261-70.
serum from immune rabbits to nonimmune ones transferred protection, thus DOI: 10.1056/NEJMra2028358
expressing the concept of humoral immunity. They further discovered that this Copyright © 2021 Massachusetts Medical Society.

inoculation did not protect against challenge with a different pneumococcal


strain, thereby showing that humoral immunity was an immunologically specific
phenomenon.1
The concept of cell-mediated immunity developed more slowly. Koch’s attempts
to immunize against tuberculosis with tuberculin, a set of proteins extracted from
Mycobacterium tuberculosis, were not successful. Von Pirquet showed that, in patients
who had tuberculosis, injection of tuberculin caused a local inflammatory re-
sponse; this delayed-type hypersensitivity was characterized by accumulation of
lymphocytes and macrophages and was antigen-specific. In 1957, Dubos and
Schaedler reported that injecting bacille Calmette–Guérin (BCG), an attenuated
strain of Mycobacterium bovis, into mice enhanced their resistance to Staphylococcus
aureus and M. fortuitum.2 They attributed this non–antigen-specific immunity to
activation of monocytes, a phenomenon that was shown in vitro.3,4 Seminal ex-
periments by Mackaness5,6 showed that infection of mice with intracellular organ-
isms such as brucella or BCG protected them against antigenically unrelated
bacteria such as listeria. The proposed mechanism was an antigenically specific
stimulation that resulted in an enhanced nonspecific bactericidal capacity of mac-
rophages.
Mackaness called this phenomenon acquired cellular resistance. Musher et al.
reported that infection with listeria protected mice against the quintessentially

n engl j med 384;3  nejm.org  January 21, 2021 261


The New England Journal of Medicine
Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 1. Evidence for Persistent, Beneficial Nonspecific Immunity.*

Type of Evidence Epigenetic Modifications Reference


In vitro: incubating monocytes with BCG or BCG increases H3K4me3 at TNF and IL6 promoter ; BCG14
Kleinnijenhuis et al.,14 Arts
β-glucan enhances microbicidal activity increases H3K27ac in immune pathways related to et al.,15 Fanucchi et al.16
against candida, Escherichia coli, and Fc epsilon signaling, chemokine signaling, and Fc
Staphylococcus aureus gamma–mediated phagocytosis15; training occurs
through long noncoding RNA, such as UMLILO, to
rearrange topologically associated domains of chro­
mosomes16
Animal models: infection by intracellular bac­ Historic studies from 1960–1970 did not evaluate epigen­ Dubos and Schaedler,2 Ratzan
teria (brucella, listeria, salmonella, toxo­ etic modifications et al.,8 Spencer et al.,9 Clark
plasma, influenza, or BCG) induces non­ et al.,10 Remington et al.,11
specific, cross-protective immunity against Murray et al.12
other intracellular pathogens
Animal models: memory-like cells capable of After infection, memory NK cells and CD8+ T cells share a Lau et al.17
protective recall develop in mice infected common opening in chromatin accessibility in genes
with MCMV related to immune memory (Tcf7 and Bach2) and
immune-effector function (granzymes and perforin)17
Animal models: infection with intracellular MTBVAC (attenuated, live Mycobacterium tuberculosis vac­ Dubos and Schaedler,2
organisms induces nonspecific, cross- cine) increases H3K4me3 of TNF and IL6 promoters18; Mackaness,5 Mackaness,6
protective immunity against extracellular candida β-glucan increases H3K27ac and H3K4me3 Musher et al.,7 Kleinnijenhuis
pathogens (E. coli, S. aureus, candida, and of genes related to TNF, mTOR, and glycolysis (hexo­ et al.,14 Tarancón et al.,18
Streptococcus pneumoniae) kinase, pyruvate kinase, TNF, IKKB, TSC1, and ribo­ Cheng et al.,19 Arts et al.,20
somal protein S6)19; BCG increases H3K4me3 of TNF Kleinnijenhuis et al.,21 Walk
and IL6 and decreases H3K9me3 of genes related to et al.,22 Kaufmann et al.23
glycolysis (GLUD, GLS, HK2, and MTOR)20
Humans: injection of BCG or β-glucan in­ Elderly persons vaccinated with BCG have increased Arts et al.,15 Walk et al.,22
creases antibody responses to vaccines, H3K27ac of the TNF and IL6 promoters24; β-glucan Giamarellos-Bourboulis
increases clearance of yellow fever viremia, increases epigenome-wide chromatin accessibility et al.,24 Saeed et al.25
and increases symptoms after experimen­ (DNase accessibility assay), H3K27ac, and H3K4me3
tally induced malaria infection in genes related to cytokines and chemokines25
Epidemiologic studies: BCG decreases neo­ Humans vaccinated with BCG have increased H3K4me3 Kleinnijenhuis et al.,14 Benn
natal sepsis, pneumonia, and all-cause at TNF and IL6 promoters14 et al.26
mortality in developing countries
Humans: β-glucan reverses immune tolerance β-glucan is able to restore beneficial H3K27ac marks in Novakovic et al.27
promoters and enhancers after LPS-induced tolerance27

* BCG denotes bacille Calmette–Guérin, H3K4me3 trimethylation of histone 3 at lysine 4, H3K27ac acetylation of histone 3 at lysine 27, LPS
lipopolysaccharide, MCMV murine cytomegalovirus, mTOR mammalian target of rapamycin, NK natural killer, and TNF tumor necrosis
factor.

extracellular organism Streptococcus pneumoniae7 stood to be due to long-lasting epigenetic changes


and attributed this effect to increased oxidative in macrophages.
metabolism in listeria-infected macrophages.8
Nonspecific acquired cellular resistance pro- Epigene t ic s a nd A n t igen-
tected experimental animals against challenge Nonspecific T r a ined Im muni t y
with intracellular bacteria (listeria),6 viruses (in-
fluenza virus),9 and protozoa (babesia and plas- The mechanisms by which innate cells — mac-
modium,10 toxoplasma,11 and leishmania12). A rophages, dendritic cells, natural killer cells, and
crucial set of observations in Mackaness’s ex- reticuloendothelial cells — can become activated
periments went unexplained. Macrophages from by acquiring antigen-nonspecific immune mem-
mice that were previously infected with listeria, ory have been elucidated in the past decade. This
brucella, or BCG retained enhanced antimicro- adaptive characteristic, one that is independent
bial capacity after the original infecting organ- of lymphocytes, has been termed “trained im-
isms had been eradicated, without reintroduc- munity.”13 The two most commonly studied
tion of the sensitizing antigen and independent of models for inducing trained immunity are im-
lymphocytes. This phenomenon is now under- munization with BCG or with β-glucan (a cell-

262 n engl j med 384;3  nejm.org  January 21, 2021

The New England Journal of Medicine


Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
postinfectious epigenetic immune modifications

wall component of candida). The lymphocyte- which controls the binding of transcription fac-
independent nature of this training and antigen tors to gene promoters, and the release of non-
nonspecificity have been shown in a broad array coding RNAs, which also regulate transcrip-
of experiments (Table 1). Seven days after puri- tional processes.
fied macrophages were incubated with BCG or This reorganization, not of the DNA itself but
with β-glucan in vitro, exposure to lipopolysac- “above” (epi) the DNA, is referred to as epigen-
charide, candida, or staphylococcus caused them etic modification. These modifications restruc-
to generate greater amounts of tumor necrosis ture accessibility to promoters and, therefore,
factor than control macrophages.14 When mice accessibility to gene transcription (Fig. 1). The
that had severe combined immunodeficiency net result is a reorganization of the interactions
(SCID) and, therefore, lacked B or T cells were within topologically associated domains that
infected with BCG, their macrophages had more inhibits or stimulates expression of some genes;
robust antigen-nonspecific secondary responses thus, the genes and domains become available
and they were able to survive challenge with a for rapid coexpression on cellular activation.16
lethal dose of candida.14 Mice that were vacci- After stimulation, innate cells undergo epigene-
nated with β-glucan had increased survival when tic rewiring with increased chromatin accessibil-
given a lethal dose of S. aureus.19 Three months ity that persists and allows the cell to respond
after humans were vaccinated with BCG, their more quickly and robustly to a second stimulus
macrophages generated greater amounts of tu- that is either similar or antigenically different.
mor necrosis factor and interleukin-6 after expo- “Trained immunity” is the term given to these
sure to staphylococcus or candida.14 Other ex- antigen-nonspecific, cross-protective, and long-
periments in laboratory animals have shown lived innate immune responses.
that macrophages can acquire antigen-nonspe- When BCG or β-glucan stimulates innate cells,
cific and cross-protective trained immunity nucleotide-binding oligomerization domain–like
against an array of pathogens, including bacteria receptors, C-type lectin receptors, and other
(Escherichia coli, S. pneumoniae, and S. aureus),14,19-21 pattern-recognition receptors induce a shift in
viruses (yellow fever and influenza),9,15 parasites cellular metabolism, which increases glycolysis,
(plasmodia),22 and fungi (Candida albicans).14,20,22,23 glutaminolysis, and cholesterol synthesis20 and
Within 5 days after initial exposure to BCG or triggers epigenetic changes.25,28 The best-described
β-glucan, innate cells begin to exhibit more ro- changes to date are increases in chromatin ac-
bust responses that persisted for at least 1 year, cessibility and histone 3 lysine trimethylation
long after the initial stimulus had been eradi- (H3K4me3) in interleukin-1β, interleukin-6, and
cated.21 Clearly, the trained immunity described tumor necrosis factor. The use of chemical inhibi-
in this body of experimental data is distinct tors of histone methylation blocks BCG-induced
from B-cell–mediated and T-cell–mediated anti- epigenetic changes and trained immunity, which
gen-specific adaptive immunity. establishes a mechanistic relationship between
Trained immunity occurs through epigenetic epigenetic changes and trained immunity.14,15,19
changes, a reconfiguration of how the genome In addition to BCG and β-glucan, exposure of
is organized. Each human cell contains approxi- mice to another live, attenuated vaccine derived
mately 2 m of double-stranded DNA that is from M. tuberculosis (called MTBVAC) induced
packed and tightly wrapped into the nucleus in similar epigenetic-mediated innate training and
nucleosomes consisting of groups of eight his- decreased mortality when vaccinated mice were
tones. The packing of the DNA is not haphazard; challenged with S. pneumoniae.18 Epigenetic-medi-
rather, it is organized into a three-dimensional ated innate training was still detected 1 year
structure such that the ability of the gene-­ after initial sensitization, even though termi-
expression machinery to access a specific gene nally differentiated innate cells are thought to
promoter depends on the its location within the live only a few weeks. Preliminary evidence sug-
chromatin network. The chromatin can be reor- gests that transcriptional, metabolic, and epi-
ganized by acetylating or methylating histones genetic changes in hematopoietic stem cells yield
or methylating the 5′ cytosine residues of DNA terminally differentiated monocytes with in-
(Fig. 1). Other layers of epigenetic regulation of creased antimicrobial killing capacity,23,29-31
transcription are methylation of DNA itself, which establishes a potential mechanism for

n engl j med 384;3  nejm.org  January 21, 2021 263


The New England Journal of Medicine
Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A
Methylation
K4 K4
K9 K9
Chromosomes
Methylation K14 S10 K14 S10
K27 K27

Chromatin S28 S28


K36 K36
H3 H3

DNA Histone
Closed Heterochromatin Open Euchromatin

B Open Euchromatin Other Epigenetic Marks


DNA hypomethylation Ubiquination
H3K4me3 Butyrylation
H3K27Ac Crotonylation
Lactylation
?
Hypusination

Bivalent, “poised” epigenetic marks RNA Methylation


H3K4me3
H3K27me3

Closed Heterochromatin lncRNA


DNA hypermethylation lncRNA
H3K9me3
H3K27me3

Scaffold for multiple modifying


complexes and topologically associated domains

C D
BCG
β-glucan
Pattern-
recognition Dectin-1
receptor

NOD2 mTOR ↑ PD-1,


NFAT CTLA4, and TIM3
MACROPHAGE LYMPHOCYTE
NF-κB NFAT

H3K27me3
H3K4me3
H3K9me3
IFNG and TNF
Global DNA
Promoter IL6 and TNF hypermethylation

↑ Interleukin-6 and TNF response ↓ Interferon-γ and TNF response

264 n engl j med 384;3 nejm.org January 21, 2021

The New England Journal of Medicine


Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
postinfectious epigenetic immune modifications

Figure 1 (facing page). Epigenetic Mechanisms of Antigen- measles vaccine, or oral polio vaccine — have
Nonspecific Immune Memory (Trained Immunity). decreased all-cause mortality.26 (Some nonlive
DNA is condensed into closed heterochromatin and vaccines have been suggested to negate the ben-
open euchromatin by wrapping DNA around histone efit of live vaccines and increase the risk of un-
proteins (Panel A). Histone proteins have tails that can related, antigen-nonspecific infections, especial-
be modified, often with marks that make genes more
ly among girls.26) Results have been mixed, but
accessible. Some common post-translational modifi­
cations include trimethylation of histone 3 at lysine 4 with follow-up of 8 months to 5 years, measles
(H3K4me3), which promotes open chromatin. In con­ or BCG vaccination decreased mortality from
trast, trimethylation of histone 3 at lysine 27 (H3K27me3) nontarget-related infections.26 A randomized,
promotes heterochromatin. The gene-expression impli­ controlled trial showed that, in the year after
cations of certain epigenetic marks are well established
BCG vaccination of South African adolescents,
(Panel B). DNA methylation decreases transcription-
factor binding and decreases gene expression. In con­ the risk of upper respiratory tract infections was
trast, H3K27 acetylation increases chromatin accessibil­ reduced by 72%.32 A similarly designed trial in-
ity, which increases the capacity for gene expression. volving elderly adults showed that BCG vaccina-
Genes can also be “poised” in a bivalent state simulta­ tion augmented antigen-nonspecific innate immu-
neously with both activating (i.e., H3K4me3) and re­
nity that corresponded with beneficial epigenetic
pressive (i.e., H3K27me3) modifications. Epigenetic
marks other than acetylation and methylation are not as changes and that vaccination was associated
well studied and are less understood. Long noncoding with a 75% decrease in respiratory infections.24
RNA (lncRNA) acts as a scaffolding to connect topo­ The ability of BCG, the oral polio vaccine, and
logically associated domains of genes many kilobases the measles vaccine to improve antigen-unrelated
apart. The role of histone ubiquination, crotonylation,
immunity and decrease nonspecific infections is
butyrylation, or lactylation and of RNA acetylation and
methylation in long-lasting immune function is still being broadly evaluated as a means to decrease
­unknown. In macrophages (Panel C), bacille Calmette– coronavirus disease 2019–related morbidity and
Guérin (BCG) and β-glucan induce specific ligand–cell mortality in multiple studies.33
signaling pathways through nucleotide-binding oligo­
merization domain–containing protein 2 (NOD2) and
mammalian target of rapamycin (mTOR), respectively, De t r imen ta l Epigene t ic Sc a r s
which results in increased H3K4me3 at the TNF and IL6 in Im mune Cel l s
promoter. Thereby, when macrophages are exposed to
an antigenically different, nonspecific stimulation, there Unfortunately, epigenetic changes can serve as a
is increased interleukin-6 and tumor necrosis factor double-edged sword, because the same mecha-
(TNF) response. In both monocytes and lymphocytes
nisms that drive antigen-nonspecific, cross-pro-
(Panel D), sepsis, pneumonia, and chronic infections
induce detrimental epigenetic scars, such as DNA hy­ tective beneficial immunity can also underlie
permethylation and closed chromatin conformation detrimental, antigen-nonspecific immune sup-
that results in decreased effector function when the pression. In order to prevent exuberant, patho-
cells are challenged. CTLA-4 denotes cytotoxic T-lym­ logic immunity, epigenetic machinery rearranges
phocyte–associated protein 4, H3K9me3 trimethylation
chromatin organization to limit immune-induced
of histone 3 at lysine 9, H3K27ac acetylation of histone
3 at lysine 27, NFAT nuclear factor of activated T cells, collateral damage to host tissue. This was illus-
NF-κB nuclear factor κB, PD-1 programmed death 1, trated in experiments in which blocking these
and TIM3 T-cell immunoglobulin mucin 3. epigenetic changes led to exuberant immune
pathologic responses.34,35 Limiting exuberant im-
munity in the short term has advantages, but in
the long-lived nature of antigen-nonspecific the long term, the persistent immune suppres-
training. sion renders the host susceptible to a variety of
microbial pathogens. Previous infections may
increase the risk of future antigen-nonspecific
Epidemiol o gy of A n t igen-
Nonspecific Im muni t y infections. For example, in 1909 von Pirquet
observed clinical flares of tuberculosis during
These observations now explain epidemiologic recovery from measles and showed that this
studies that have shown a survival benefit from clinical finding correlated with suppressed re-
live, attenuated vaccines that is not specific to sponses to tuberculin on delayed-type hypersen-
the targeted pathogen. In developing countries, sitivity skin tests.36 Measles-induced immune
children vaccinated with live vaccines — BCG, suppression also increases the risk of secondary

n engl j med 384;3  nejm.org  January 21, 2021 265


The New England Journal of Medicine
Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

bacterial otitis media, tracheitis, and pneumo- dritic cells, thereby diminishing type 1 helper
nia.37 Similarly, hepatitis B and C have been as- T-cell immunity and skewing immunity toward
sociated with an increased risk of staphylococcal a type 2 helper T-cell response (decreased inter­
and pneumococcal infections.38-40 feron-γ and increased interleukin-4, -5, and -13).57
After recovery from the acute phase of sepsis, Experimental E. coli or influenza pneumonia in
adaptive and innate immune function may be mice causes long-lasting epigenetic changes that
profoundly suppressed for a prolonged period, tolerize macrophage function and decrease the
which can result in an increased risk of second- capacity to phagocytose antigen-nonspecific, un-
ary bacterial infections.41,42 A propensity-matched related bacteria.41 One mechanism of the long-
cohort study showed that for at least 2 years af- lived nature of epigenetic scarring is the devel-
ter recovery from sepsis, the risk of death is in- opment of detrimental epigenetic marks in
creased 10 to 22%.43 Similarly, chronic helminth progenitor, stem-cell memory populations.53,56,61
infections perturb host immunity and increase Specifically, chronic LCMV induces epigenetic-
the risk of tuberculosis by a factor of 444 and the mediated exhausted memory T cells.53,56 Chronic
risk of human immunodeficiency virus (HIV) by M. avium infection in mice induces dysfunction
a factor of 2 to 645,46 and cause a loss of vaccine in stem-cell differentiation,61 and macrophages
immunogenicity.47 When Sandvall et al. reported derived after tolerization of hematopoietic stem
that pneumococcal pneumonia is followed by a cells have decreased production of tumor necro-
persistent increase in the risk of death that was sis factor on secondary stimulation.62
proportional to the severity of the initial infec- Just as with beneficial epigenetic marks, detri-
tion,48 their interpretation was that the initial mental epigenetic scars are also persistently re-
infection was a marker of increased susceptibil- tained. After bacterial pneumonia or sepsis, epi-
ity41; long-lasting detrimental epigenetic changes genetic reprogramming inhibits macrophage
that result from severe infection appear to offer phagocytic capacity for at least 6 months.41
a better explanation. Similarly, after successful Similarly, 6 months after children have been
treatment of tuberculosis, an increase in all- successfully treated for schistosomiasis, they
cause risk of infection-related death and non– retain detrimental DNA hypermethylation marks
infection-related death is noted.49,50 in CD4+ T cells that ablate immune responses to
Mechanistic insight into these suppressed BCG.58 Despite successful antituberculosis ther-
immune responses has been derived from a wide apy, detrimental epigenetic scars persist in both
variety of experiments in which detrimental epi- adaptive and innate immune cells for at least
genetic scars in both innate and adaptive im- 6 months.60 After 2 years of successful antiretro-
mune cells have been shown. The evidence for viral therapy, HIV-infected persons retain detri-
such changes is overwhelming (Table 2). In mice, mental epigenetic marks in the genes encoding
infection due to chronic lymphocytic chorio- interleukin-2 and programmed death 1 in CD8+
meningitis virus (LCMV) blunts lymphocyte pro- T cells.59
duction of interleukin-2, tumor necrosis factor, Detrimental epigenetic scars also occur as a
and interferon-γ with a consequent decrease in consequence of physiologic aging. With aging,
T-cell proliferative capacity, delayed-type hyper- immune cells increase global DNA methylation
sensitivity, and antigen-nonspecific microbial and closed chromatin conformation. These age-
killing capacity.51,54 Chronic LCMV infection in- related epigenetic changes occur preferentially
duces DNA hypermethylation marks in the pro- in aspects of immunity that are required for the
moter region of IFNG that inhibits CD8+ T-cell development of new immunologic memory63;
immunity.52,55 Exposure of monocytes to lipo- DNA hypermethylation and chromatin closure at
polysaccharide induces a closed chromatin con- the interleukin-7 receptor and T-cell factor 1
formation that epigenetically silences the gene (TCF1) appear to be responsible. Interleukin-7 is
encoding interleukin-6 (IL6), which results in required for the survival of memory cells, and
decreased production of interleukin-6 (and other TCF1 is a transcription factor necessary for the
proinflammatory cytokines) on restimulation.25 development of effector memory immune forma-
In animal models of sepsis, 12 weeks after liga- tion. Therefore, detrimental epigenetic scars,
tion of the colon, persistent epigenetic perturba- whether from chronic infections or from aging,
tions inhibit interleukin-12 production in den- inhibit the development of new immune memory.

266 n engl j med 384;3  nejm.org  January 21, 2021

The New England Journal of Medicine


Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
postinfectious epigenetic immune modifications

Table 2. Evidence for Persistent, Detrimental Effects on Nonspecific Immunity.*

Type of Evidence Epigenetic Modifications Reference


In vitro: LPS causes decreased TNF and inter­ LPS-tolerized monocytes decrease H3K4me3 and Saeed et al.,25 Novakovic et al.27
leukin-6 production after exposure of mono­ H3K27ac marks in proinflammatory genes and tran­
cytes to diverse challenges scription factors (IRFs, STAT, and HIF-1α)27; LPS
induces epigenome loss of H3K27ac marks25
Animal models: chronic LCMV infection inhibits CD8+ T cells from chronic LCMV–infected mice have in­ Seo et al.,51 Ghoneim et al.,52
TNF, interleukin-2, and interferon-γ produc­ creased DNA methylation and decreased chromatin Miller et al.,53 Ahmed et al.,54
tion; decreases antigen-induced proliferation, accessibility in Ifng, Myc, and genes related to T-cell Bengsch et al.,55 Yao et al.56
and decreases nonspecific microbicidal receptor signaling and interleukin-7 and -2 signaling;
activity increased chromatin accessibility in genes related
to immune-checkpoint inhibition (PD-1, TIM3, and
LAG3)51-53
Animal models: sepsis causes prolonged epi­ Six weeks after colon ligation–induced sepsis, interleu­ Roquilly et al.,41 Wen et al.57
genetic inhibition of interleukin-12 and de­ kin-12 and both p35 and p40 promoters of dendritic
creased microbial killing capacity cells have decreased H3K4me3 and increased
H3K27me2; PRC remains bound and inhibits the
­interleukin-12 promoter57
Animal models: infection with E. coli or influenza Infection-cured mice have decreased H3K27ac of TLR5 Roquilly et al.41
virus induces detrimental epigenetic marks and epigenomic overlap with human monocytes
and inhibits phagocytosis of unrelated or­ tolerized by LPS41
ganisms
Humans: measles increases the risk of TB and No studies have yet evaluated whether epigenetic per­ Turk,36 Perry and Halsey37
other bacterial infections turbations are part of the postmeasles ablation of
adaptive immunity
Humans: recovery from sepsis is followed by a No human studies have documented postsepsis in­ van der Poll et al.,42 Prescott
persistent increased risk of death duced immune tolerance and immune exhaustion, et al.43
but no epigenetic studies involving humans post
sepsis have yet been completed
Humans: hepatitis B or C virus infection in­ Patients with hepatitis have DNA hypermethylation Musher and McKenzie,38 Marrie
creases the risk of bacteremia or bacterial and closed chromatin conformation that resembles et al.39
pneumonia chronic LCMV–induced immune exhaustion
Humans: helminths ablate vaccine immuno­ Six months after successful deworming, CD4+ T cells Labeaud et al.,47 DiNardo et al.58
genicity from children who had had schistosomiasis retained
DNA hypermethylation of interleukin-12–interferon-γ
signaling and multiple transcription factors58
Humans: chronic helminth infection increases Memory CD4+ T cells in patients with HIV have DNA Kroidl et al.,45 Downs et al.,46
the risk of TB or HIV infection hypermethylation of interleukin-2; 2 years after suc­ Youngblood et al.59
cessful aviremia, CD8+ T cells retain unmethylated
DNA marks at PD-159
Humans: after successful therapy, patients with After TB therapy, NK cells, monocytes, and CD8+ and Verver et al.,49 Romanowski
TB are at increased risk for recurrent TB and CD4+ T cells retain DNA hypermethylation of the et al.,50 DiNardo et al.60
other infections and retain detrimental DNA interferon-γ, mTOR, TNF–NF-κB, and PI3K–AKT
hypermethylation marks signaling pathways60

* HIF-1α denotes hypoxia-inducible factor 1α, IRF interferon regulatory factor, LAG3 lymphocyte-activation gene 3, LCMV lymphocytic cho­
riomeningitis virus, NF-κB nuclear factor κB, PD-1 programmed death 1, PI3K phosphatidylinositol 3-kinase, PRC polycomb repressive
complex, STAT signal transducer and activator of transcription, TB tuberculosis, TIM3 T-cell immunoglobulin mucin 3, and TLR5 toll-like
receptor 5.

These observations may explain why elderly per- H a r ne ssing Epigene t ic s t o He a l


sons lack strong immunologic responses, as De t r imen ta l Sc a r s
shown by their increased susceptibility to infec-
tion and decreased responses to immunization. An understanding of epigenetic mechanisms
The implications of epigenetic scars for tumor shows why, in vaccine development, investiga-
immunology and susceptibility to autoimmune tors cannot simply extrapolate results from ex-
disease are apparent, but these problems are perimental animals or healthy humans to results
beyond the scope of the present article. in countries where chronic infections are preva-

n engl j med 384;3  nejm.org  January 21, 2021 267


The New England Journal of Medicine
Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

lent. An important question is whether it is pos- nomenon by which administration of nonlive


sible to use knowledge of epigenetics to mend vaccines hinders immunity and increases the
detrimental scars. Immune cells, and macro- risk of nonrelated infections, particularly among
phages in particular, have plasticity in their fate young women.26
and function,64 and initial evidence preliminarily
suggests that detrimental epigenetic scars are C onclusions a nd Impl ic at ions
mendable. In ex vivo studies, human monocytes for the F u t ur e
that had decreased responsiveness to lipopoly-
saccharide were rescued by β-glucan, resulting Studies over the past decade have revealed a new
in reversal of detrimental epigenetic chromatin epigenetic basis for acquired immunity in both
marks and restoration of the production of tu- innate and adaptive immune cells. These studies
mor necrosis factor.27 In mice, decitabine, a hypo- provide a mechanistic explanation for the anti-
methylating agent, reversed chronic LCMV–­ gen-nonspecific immune enhancement that has
induced detrimental DNA hypermethylating marks been shown in numerous studies during the past
in CD8+ T cells and restored immune function- 60 years. They show that innate training works
ality.52 Additional evidence that epigenetic scars in concert with, and augments, specific T- and
can be reversed comes from a trial involving elder­ B-cell responses. The metaphor of a double-
ly adults after hospital discharge, a population edged sword reflects the fact that those same
expected to have substantial epigenetic scars. mechanisms that underlie beneficial epigenetic
This trial showed a benefit from BCG vaccina- innate training also underlie detrimental epigen-
tion, as indicated by increased candida-induced etic scarring that occurs after severe infections
cytokine production and decreased all-cause in- or with aging. Identifying safe mechanisms to
fections (especially respiratory infections), chang- reverse detrimental epigenetic scars, for example
es that were associated with epigenetic rewiring.24 by injection of β-glucan or BCG vaccination,
could have broad implications for decreasing
mortality after serious infections, protecting el-
Impl ic at ions of Epigene t ic s
for Vac cine De v el opmen t derly persons against infection, improving treat-
ment approaches for tuberculosis and other
Ideally, vaccine development would simultane- chronic infections, and implementing effective
ously harness classic B-cell and T-cell adaptive vaccines in developing countries.
immunity, as well as antigen-nonspecific innate Reversing detrimental epigenetic scarring and
immune training. With BCG bioengineered to applying beneficial innate training are on the
lack urease production and include lysteriolysin verge of clinical applicability but require contin-
O, a toxin that enables listeria to escape the ued basic and translational research. Cellular
phagosome, the VPM1002 strain of BCG in- metabolic shifts trigger the epigenetic changes
creases T-cell immunogenicity while maintain- that underlie both beneficial innate training and
ing innate training.65 Similarly, MTBVAC en- detrimental epigenetic scarring. Identifying the
hances T-cell immunity while also inducing metabolic processes that drive epigenetic chang-
epigenetic-mediated trained immunity.18 Vacci- es will clarify methods to manipulate the system
nation with M. tuberculosis–specific antigens and identify potential means to overcome im-
(Rv0125 and Rv1196) together with the adjuvant mune suppression.51,55,67 The differences in epi-
AS01E induces both robust innate and adaptive genetic effects according to sex suggest that we
immunity and provides substantial protection will need to clarify how hormones influence
against progression of tuberculosis.66 Previous epigenetic changes before they can be broadly
administration of a live vaccine augments hu- used.26,68 Detrimental scars that are induced by
moral responses to influenza, hepatitis B, pneu- chronic LCMV in mice are well-characterized,
mococcal, and meningococcal vaccines.26 Clari- and this understanding needs to be applied to
fying and harnessing the mechanisms for these chronic infections in humans. The documenta-
effects might help to enhance the efficacy of all tion of detrimental and beneficial epigenetic
existing vaccines. In contrast, the timing of vac- marks is only beginning. Oncologists have es-
cine administration needs to address the phe- tablished public database repositories (the Can-

268 n engl j med 384;3  nejm.org  January 21, 2021

The New England Journal of Medicine


Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
postinfectious epigenetic immune modifications

cer Genome Atlas and Gene Expression Omni- increases susceptibility to other infections and
bus) that allow data to be merged and mined, decreases vaccine immunogenicity; how this can
thereby aiding in the identification of new be reversed is unknown. To date, vaccine devel-
therapeutic options. Because of similarities be- opment has been focused nearly exclusively on
tween chronic infection and cancer-induced im- the vaccine and the adjuvant, not on the host
mune suppression,51 tools identified to reverse receiving the vaccine. Detrimental epigenetic
cancer-induced detrimental epigenetic scars69 scars that occur after chronic infection or with
have the potential to reverse postinfectious epi- aging will probably inhibit robust vaccine im-
genetic scars. munogenicity. Finding a mechanism to reverse
In 1966, Fazekas de St. Groth and Webster detrimental epigenetic immune scarring may be
presciently stated: “Response to vaccine depends the key to better treatment of infections in el-
not only on the nature of the antigen itself but derly persons and for successful application of
also on the immunological history of the recipi- vaccines in countries where malaria, helminthic
ent.”70 Up to one third of the world’s population infection, tuberculosis, and other acute or
has been affected by malaria, helminth infec- chronic infections are so unfortunately wide-
tions, HIV infection, tuberculosis, or another spread.
chronic infection. The effect of the persistent Disclosure forms provided by the authors are available with
epigenetic scars from these chronic infections the full text of this article at NEJM.org.

References
1. Gray G, Musher DM. The history of 10. Clark IA, Allison AC, Cox FE. Protec- culosis vaccine MTBVAC induces trained
pneumococcal disease. Washington, DC:​ tion of mice against Babesia and Plasmo- immunity and confers protection against
ASM Press, 2008. dium with BCG. Nature 1976;​259:​309-11. experimental lethal pneumonia. PLoS
2. Dubos RJ, Schaedler RW. Effects of 11. Remington JS, Krahenbuhl JL, Men- Pathog 2020;​16(4):​e1008404.
cellular constituents of mycobacteria on denhall JW. A role for activated macro- 19. Cheng SC, Quintin J, Cramer RA,
the resistance of mice to heterologous in- phages in resistance to infection with et al. mTOR- and HIF-1α-mediated aero-
fections I. Protective effects. J Exp Med Toxoplasma. Infect Immun 1972;​6:​829-34. bic glycolysis as metabolic basis for trained
1957;​106:​703-17. 12. Murray HW, Rubin BY, Rothermel CD. immunity. Science 2014;​345:​1250684.
3. Loebel RO, Shorr E, Richardson HB. Killing of intracellular Leishmania don- 20. Arts RJW, Carvalho A, La Rocca C,
The influence of adverse conditions upon ovani by lymphokine-stimulated human et al. Immunometabolic pathways in BCG-
the respiratory metabolism and growth of mononuclear phagocytes: evidence that induced trained immunity. Cell Rep 2016;​
human tubercle bacilli. J Bacteriol 1933;​ interferon-gamma is the activating lym- 17:​2562-71.
26:​167-200. phokine. J Clin Invest 1983;​72:​1506-10. 21. Kleinnijenhuis J, Quintin J, Preijers F,
4. Elberg SS, Schneider P, Fong J. Cross- 13. Netea MG, Quintin J, van der Meer et al. Long-lasting effects of BCG vaccina-
immunity between Brucella melitensis JW. Trained immunity: a memory for in- tion on both heterologous Th1/Th17 re-
and Mycobacterium tuberculosis; intra- nate host defense. Cell Host Microbe sponses and innate trained immunity.
cellular behavior of Brucella melitensis in 2011;​9:​355-61. J Innate Immun 2014;​6:​152-8.
monocytes from vaccinated animals. J Exp 14. Kleinnijenhuis J, Quintin J, Preijers F, 22. Walk J, de Bree LCJ, Graumans W,
Med 1957;​106:​545-54. et al. Bacille Calmette-Guerin induces et al. Outcomes of controlled human ma-
5. Mackaness GB. Cellular resistance to NOD2-dependent nonspecific protection laria infection after BCG vaccination. Nat
infection. J Exp Med 1962;​116:​381-406. from reinfection via epigenetic repro- Commun 2019;​10:​874.
6. Mackaness GB. The immunological gramming of monocytes. Proc Natl Acad 23. Kaufmann E, Sanz J, Dunn JL, et al.
basis of acquired cellular resistance. J Exp Sci U S A 2012;​109:​17537-42. BCG educates hematopoietic stem cells
Med 1964;​120:​105-20. 15. Arts RJW, Moorlag SJCFM, Novakovic to generate protective innate immunity
7. Musher DM, Ratzan KR, Weinstein L. B, et al. BCG vaccination protects against against tuberculosis. Cell 2018;​172(1-2):​
The effect of Listeria monocytogenes on experimental viral infection in humans 176-190.e19.
resistance to pneumococcal infection. through the induction of cytokines asso- 24. Giamarellos-Bourboulis EJ, Tsilika M,
Proc Soc Exp Biol Med 1970;​135:​557-60. ciated with trained immunity. Cell Host Moorlag S, et al. Activate: randomized
8. Ratzan KR, Musher DM, Keusch GT, Microbe 2018;​23(1):​89-100.e5. clinical trial of BCG vaccination against
Weinstein L. Correlation of increased 16. Fanucchi S, Fok ET, Dalla E, et al. Im- infection in the elderly. Cell 2020;​183(2):​
metabolic activity, resistance to infection, mune genes are primed for robust tran- 315-323.e9.
enhanced phagocytosis, and inhibition of scription by proximal long noncoding 25. Saeed S, Quintin J, Kerstens HHD, et al.
bacterial growth by macrophages from RNAs located in nuclear compartments. Epigenetic programming of monocyte-to-
Listeria- and BCG-infected mice. Infect Nat Genet 2019;​51:​138-50. macrophage differentiation and trained
Immun 1972;​5:​499-504. 17. Lau CM, Adams NM, Geary CD, et al. innate immunity. Science 2014;​ 345:​
9. Spencer JC, Ganguly R, Waldman RH. Epigenetic control of innate and adaptive 1251086.
Nonspecific protection of mice against in- immune memory. Nat Immunol 2018;​19:​ 26. Benn CS, Fisker AB, Rieckmann A,
fluenza virus infection by local or systemic 963-72. Sørup S, Aaby P. Vaccinology: time to
immunization with Bacille Calmette- 18. Tarancón R, Domínguez-Andrés J, change the paradigm? Lancet Infect Dis
Guérin. J Infect Dis 1977;​136:​171-5. Uranga S, et al. New live attenuated tuber- 2020;​20(10):​e274-e283.

n engl j med 384;3  nejm.org  January 21, 2021 269


The New England Journal of Medicine
Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.
postinfectious epigenetic immune modifications

27. Novakovic B, Habibi E, Wang S-Y, et al. tic targets. Nat Rev Immunol 2017;​17:​407- gle-cell RNA-seq reveals TOX as a key
β-Glucan reverses the epigenetic state of 20. regulator of CD8+ T cell persistence in
LPS-induced immunological tolerance. Cell 43. Prescott HC, Osterholzer JJ, Langa chronic infection. Nat Immunol 2019;​20:​
2016;​167(5):​1354-1368.e14. KM, Angus DC, Iwashyna TJ. Late mortal- 890-901.
28. Verma D, Parasa VR, Raffetseder J, ity after sepsis: propensity matched cohort 57. Wen H, Dou Y, Hogaboam CM, Kun-
et al. Anti-mycobacterial activity correlates study. BMJ 2016;​353:​i2375. kel SL. Epigenetic regulation of dendritic
with altered DNA methylation pattern in 44. Elias D, Mengistu G, Akuffo H, Brit- cell-derived interleukin-12 facilitates im-
immune cells from BCG-vaccinated sub- ton S. Are intestinal helminths risk fac- munosuppression after a severe innate
jects. Sci Rep 2017;​7:​12305. tors for developing active tuberculosis? immune response. Blood 2008;​111:​1797-
29. Mitroulis I, Ruppova K, Wang B, et al. Trop Med Int Health 2006;​11:​551-8. 804.
Modulation of myelopoiesis progenitors 45. Kroidl I, Saathoff E, Maganga L, et al. 58. DiNardo AR, Nishiguchi T, Mace EM,
is an integral component of trained im- Effect of Wuchereria bancrofti infection et al. Schistosomiasis induces persistent
munity. Cell 2018;​172(1-2):​147-161.e12. on HIV incidence in southwest Tanzania: DNA methylation and tuberculosis-spe-
30. de Laval B, Maurizio J, Kandalla PK, a prospective cohort study. Lancet 2016;​ cific immune changes. J Immunol 2018;​
et al. C/EBPβ-dependent epigenetic mem- 388:​1912-20. 201:​124-33.
ory induces trained immunity in hemato- 46. Downs JA, van Dam GJ, Changalucha 59. Youngblood B, Noto A, Porichis F,
poietic stem cells. Cell Stem Cell 2020;​ JM, et al. Association of schistosomiasis et al. Cutting edge: prolonged exposure
26(5):​657-674.e8. and HIV infection in Tanzania. Am J Trop to HIV reinforces a poised epigenetic pro-
31. Cirovic B, de Bree LCJ, Groh L, et al. Med Hyg 2012;​87:​868-73. gram for PD-1 expression in virus-specific
BCG vaccination in humans elicits trained 47. Labeaud AD, Malhotra I, King MJ, CD8 T cells. J Immunol 2013;​191:​540-4.
immunity via the hematopoietic progeni- King CL, King CH. Do antenatal parasite 60. DiNardo AR, Rajapakshe K, Nishigu-
tor compartment. Cell Host Microbe 2020;​ infections devalue childhood vaccination? chi T, et al. DNA hypermethylation during
28(2):​322-334.e5. PLoS Negl Trop Dis 2009;​3(5):​e442. tuberculosis dampens host immune re-
32. Nemes E, Geldenhuys H, Rozot V, et al. 48. Sandvall B, Rueda AM, Musher DM. sponsiveness. J Clin Invest 2020;​130:​3113-
Prevention of M. tuberculosis infection with Long-term survival following pneumo- 23.
H4:IC31 vaccine or BCG revaccination. coccal pneumonia. Clin Infect Dis 2013;​ 61. Matatall KA, Jeong M, Chen S, et al.
N Engl J Med 2018;​379:​138-49. 56:​1145-6. Chronic infection depletes hematopoietic
33. Mantovani A, Netea MG. Trained in- 49. Verver S, Warren RM, Beyers N, et al. stem cells through stress-induced termi-
nate immunity, epigenetics, and Covid-19. Rate of reinfection tuberculosis after suc- nal differentiation. Cell Rep 2016;​ 17:​
N Engl J Med 2020;​383:​1078-80. cessful treatment is higher than rate of 2584-95.
34. Hu L, Yu Y, Huang H, et al. Epigenetic new tuberculosis. Am J Respir Crit Care 62. Yáñez A, Hassanzadeh-Kiabi N, Ng
regulation of interleukin 6 by histone Med 2005;​171:​1430-5. MY, et al. Detection of a TLR2 agonist by
acetylation in macrophages and its role in 50. Romanowski K, Baumann B, Basham hematopoietic stem and progenitor cells
paraquat-induced pulmonary fibrosis. CA, Ahmad Khan F, Fox GJ, Johnston JC. impacts the function of the macrophages
Front Immunol 2017;​7:​696. Long-term all-cause mortality in people they produce. Eur J Immunol 2013;​ 43:​
35. Zhang Q, Zhao K, Shen Q, et al. Tet2 treated for tuberculosis: a systematic re- 2114-25.
is required to resolve inflammation by re- view and meta-analysis. Lancet Infect Dis 63. Márquez EJ, Chung C-H, Marches R,
cruiting Hdac2 to specifically repress IL-6. 2019;​19:​1129-37. et al. Sexual-dimorphism in human im-
Nature 2015;​525:​389-93. 51. Seo H, Chen J, González-Avalos E, mune system aging. Nat Commun 2020;​
36. Turk JL. Von Pirquet, allergy and in- et al. TOX and TOX2 transcription factors 11:​751.
fectious diseases: a review. J R Soc Med cooperate with NR4A transcription fac- 64. Marakalala MJ, Martinez FO, Plüdde­
1987;​80:​31-3. tors to impose CD8+ T cell exhaustion. mann A, Gordon S. Macrophage hetero-
37. Perry RT, Halsey NA. The clinical sig- Proc Natl Acad Sci U S A 2019;​116:​12410-5. geneity in the immunopathogenesis of tu-
nificance of measles: a review. J Infect Dis 52. Ghoneim HE, Fan Y, Moustaki A, et al. berculosis. Front Microbiol 2018;​9:​1028.
2004;​189:​Suppl 1:​S4-S16. De novo epigenetic programs inhibit PD-1 65. Kaufmann SHE. Vaccination against
38. Musher DM, McKenzie SO. Infections blockade-mediated T cell rejuvenation. tuberculosis: revamping BCG by molecu-
due to Staphylococcus aureus. Medicine Cell 2017;​170(1):​142-157.e19. lar genetics guided by immunology. Front
(Baltimore) 1977;​56:​383-409. 53. Miller BC, Sen DR, Al Abosy R, et al. Immunol 2020;​11:​316.
39. Marrie TJ, Tyrrell GJ, Majumdar SR, Subsets of exhausted CD8+ T cells differ- 66. Tait DR, Hatherill M, Van Der Meeren
Eurich DT. Concurrent infection with entially mediate tumor control and re- O, et al. Final analysis of a trial of M72/
hepatitis C virus and Streptococcus pneu- spond to checkpoint blockade. Nat Im- AS01E vaccine to prevent tuberculosis.
moniae. Emerg Infect Dis 2017;​23:​1118- munol 2019;​20:​326-36. N Engl J Med 2019;​381:​2429-39.
23. 54. Ahmed R, Salmi A, Butler LD, Chiller 67. Ryan DG, O’Neill LAJ. Krebs cycle re-
40. Kaka AS, Filice GA, Kuskowski M, JM, Oldstone MB. Selection of genetic born in macrophage immunometabolism.
Musher DM. Does active hepatitis C virus variants of lymphocytic choriomeningitis Annu Rev Immunol 2020;​38:​289-313.
infection increase the risk for infection virus in spleens of persistently infected 68. Koeken VA, de Bree LCJ, Mourits VP,
due to Staphylococcus aureus? Eur J Clin mice: role in suppression of cytotoxic et al. BCG vaccination in humans inhibits
Microbiol Infect Dis 2017;​36:​1217-23. T lymphocyte response and viral persis- systemic inflammation in a sex-dependent
41. Roquilly A, Jacqueline C, Davieau M, tence. J Exp Med 1984;​160:​521-40. manner. J Clin Invest 2020;​130:​5591-602.
et al. Alveolar macrophages are epigeneti- 55. Bengsch B, Johnson AL, Kurachi M, 69. Bates SE. Epigenetic therapies for
cally altered after inflammation, leading et al. Bioenergetic insufficiencies due to cancer. N Engl J Med 2020;​383:​650-63.
to long-term lung immunoparalysis. Nat metabolic alterations regulated by the in- 70. Fazekas de St Groth S, Webster RG.
Immunol 2020;​21:​636-48. hibitory receptor PD-1 are an early driver Disquisitions on original antigenic sin. II.
42. van der Poll T, van de Veerdonk FL, of CD8(+) T cell exhaustion. Immunity Proof in lower creatures. J Exp Med 1966;​
Scicluna BP, Netea MG. The immunopa- 2016;​45:​358-73. 124:​347-61.
thology of sepsis and potential therapeu- 56. Yao C, Sun H-W, Lacey NE, et al. Sin- Copyright © 2021 Massachusetts Medical Society.

270 n engl j med 384;3  nejm.org  January 21, 2021

The New England Journal of Medicine


Downloaded from nejm.org at MONASH UNIVERSITY LIBRARY on February 14, 2021. For personal use only. No other uses without permission.
Copyright © 2021 Massachusetts Medical Society. All rights reserved.

You might also like