You are on page 1of 11

ll

Perspective
Standing on the shoulders of mice
Kwat Medetgul-Ernar2 and Mark M. Davis1,*
1Howard Hughes Medical Institute, Institute for Immunity, Transplantation and Infection, Department of Microbiology and Immunology,

Stanford University School of Medicine, Palo Alto, CA 94304, USA


2Immunology Program, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA

94304, USA
*Correspondence: mmdavis@stanford.edu
https://doi.org/10.1016/j.immuni.2022.07.008

SUMMARY

While inbred mice have informed most of what we know about the immune system in the modern era, they
have clear limitations with respect to their ability to be informative regarding genetic heterogeneity or micro-
bial influences. They have also not been very predictive as models of human disease or vaccination results.
Although there are concerted attempts to compensate for these flaws, the rapid rise of human studies, driven
by both technical and conceptual advances, promises to fill in these gaps, as well as provide direct informa-
tion about human diseases and vaccination responses. Work on human immunity has already provided
important additional perspectives on basic immunology such as the importance of clonal deletion to self-
tolerance, and while many challenges remain, it seems inevitable that ‘‘the human model’’ will continue to
inform us about the immune system and even allow for the discovery of new mechanisms.

INTRODUCTION some of the deficiencies in the inbred mouse model, humanized


mice, collaborative cross mice, pet shop mice, etc. and how
While the ‘‘Delbru €ck School’’ of bacteriophage (Judson, 1996) these have definite advantages and yet significant difficulties in
and the structure of DNA were key to the rapid advancement terms of wide adoption and utility, which is not to say that human
of molecular biology, inbred mice have long been the Rosetta immunology exists in isolation, because in most cases one must
Stone for immunologists in the modern era, allowing decades continually go to mice to rigorously test hypotheses. An excep-
of immunological phenomena to be deciphered and reduced tion to this is human genetic lesions that affect the immune sys-
to mechanistic insights, or at least workable hypotheses. Power- tem, where there is a clear cause and effect, although pheno-
ful methodologies have been developed and used to understand types can vary widely. Human genetics has even led the way in
antibody and T cell receptor diversity, major histocompatibility some cases, such as cloning the autoimmune regulator (AIRE)
complex (MHC) restriction, and the differences between innate gene in patients with a rare autoimmune disease (Nagamine
and adaptive detection of self and non-self antigens and ensuing et al., 1997; Aaltonen et al., 1997), leading to an explosion of
effector systems. Methods like monoclonal antibodies, the dis- work on the equivalent gene in mice and the elucidation of its
covery of various inflammatory pathways, and checkpoint inhibi- key role in immune tolerance (Mathis and Benoist, 2007). Simi-
tion developed in mice have been widely adopted as therapeu- larly, the identification of another key gene in tolerance, FoxP3,
tics for cancer, infectious disease, and autoimmunity as well as was discovered in humans (Bennett et al., 2001) just slightly
being a mainstay of ongoing basic research in many areas of before the mouse equivalent was identified (Brunkow et al.,
biology. But immunology is one of the least static of any area 2001), and an even more prodigious amount of work occurred
of modern biology, and so one must always ask what’s next. in defining role of this gene (Rudensky, 2011). In any case, the
The purpose of this perspective is to suggest that human immu- technology to analyze human immune responses is advancing
nology is the answer not as a replacement to mice, but as an rapidly and has tremendous potential because there is so
increasingly rich field in itself; scientists are taking advantage much unexplored territory. But also important is the way human
of many new technologies and events (like the ongoing immunology provides a different and consequently broader
pandemic) to expand our immunological universe into places perspective on basic immune mechanisms. Longer term, the
that mice cannot go, such as the interplay of the immune system many differences seen between mice and human immune re-
with thousands of human diseases, genetic and environmental sponses and the structural differences in skin and spleens high-
diversity, and the effects of the many medical treatments on peo- lighted here suggest that new mechanisms may emerge from
ple’s immune systems (Pulendran and Davis, 2020). There is also human work, especially as they relate to the lengths infectious
the distinct possibility that entirely new immunological mecha- pathogens will go to evade the human immune system.
nisms will be discovered in human beings given the many unique
genes that are known and the fact that human infants have to Human immunology provides a valuable alternative
survive many more years than mice in sufficient numbers to allow perspective
the species to survive. Within this context, we will discuss the The first time you visit a country and culture outside your own,
various ways immunologists have tried to compensate for you immediately become more aware of the many normally

Immunity 55, August 9, 2022 ª 2022 Published by Elsevier Inc. 1343


ll
Perspective

‘‘hidden assumptions’’ about your own country and culture and is not important, what takes precedence? Following Dobzhan-
thus develop a more informed perspective. In a similar way, sky’s dictum that ‘‘[n]othing in biology makes sense except in
because studies of inbred mice have been so successful and the light of evolution’’ (Dobzhansky, 1973) leads to the realization
have dominated the field for over sixty years, we have not had that autoimmunity could not be a driver of TCR repertoire
much empirical input from other species to shed light on what because it has such a modest effect on selection in children
might be important hidden assumptions in contemporary immu- and young adults (<1%). In contrast, infectious diseases, before
nology. Here, human studies are an ideal complement because widespread vaccination, antibiotics, and sanitation, have killed
they have the genetic diversity that inbred mice lack, and they almost half of children under 5 and many older children and
have the continuous exposure to infectious diseases that are young adults as well (Casanova and Abel, 2005; Su et al.,
scrupulously avoided in modern mouse facilities. A very vivid 2013). This was the case for many generations and probably
example of this to one of the authors of this perspective started soon after the invention of agriculture when people
(M.M.D.) was when Yu Wong asked what the pre-immune fre- started living and trading in fixed, dense communities. This de-
quency of pathogen-specific CD8+ T cells might be in blood gree of child mortality was even worse in the plague years. If in-
bank donors following the groundbreaking work on mice from fectious diseases are the primary driver of immune repertoire,
Moon et al. (2007), who used a sensitive tetramer staining enrich- then it makes sense to have peripheral T cells expressing as
ment method (Day et al., 2003) to quantitate the pre-immune many specificities as they can and finding other ways to hold
T cell receptor (TCR) repertoire. As a control, we also analyzed self-specific T cells in check, unless they are needed, as Yu
self-specific T cells and to our surprise found that the fre- et al. (2015) provided evidence for. Indeed, a number of classic
quencies were very similar (Yu et al., 2015; Su et al., 2013). We papers in murine immunology show that this is the case—that
also looked at the Y chromosome-encoded SMCY antigen-spe- self-specific T cells are quiescent unless triggered by an infec-
cific T cells in males and females and found a 2% to 50%–65% tion that includes their cognate antigen (Ohashi et al., 1991;
reduction in the former versus the latter and almost no difference Röcken et al., 1992).
between male and female mice (using the famous H-Y antigen). There are other examples, but the lesson here is that even
These results were confirmed a short time later with results in when the basic immune mechanisms are the same between
mice by Moon and colleagues (Legoux et al., 2015), who identi- mice and humans, the fact that we often must approach human
fied Cre-specific CD4+ T cells and showed that if Cre was immunology in ways that are different than mice can add an
expressed ubiquitously, there was only a 50% reduction in fre- important perspective. In some cases, such as the one cited,
quency, but in cases of organ-specific expression, there was this can completely change our understanding of the phe-
no detectable deletion. These results have been shocking nomenon.
because of classic experiments with super antigens, and espe-
cially TCR transgenics in the late 1980s, that have shown that Mouse models of disease
self-specific T cells are massively deleted in the thymus of Since the development of inbred mouse strains by Snell in the late
mice and thus are a major component of what was called ‘‘cen- 1940s (Gorer et al., 1948), mouse models of cancer have been a
tral tolerance’’ (Herman et al., 1991; Von Boehmer, 1990). While major activity by immunologists, soon joined by those interested
there was also persistent evidence of clonal anergy (Goodnow in autoimmunity and infectious diseases. By the 1980s, the advent
et al., 1988, 1989; Miller, 1993; Miller and Morahan, 1992; Pike of recombinant DNA methodology and transgenics and then ge-
et al., 1982) being a factor, negative selection has been widely netic ablation, etc. made mouse models of disease the premier
accepted as the major mechanism of self-tolerance for more route to understanding the role of the immune system in virtually
than 25 years and seems to confirm Burnet’s earlier speculation every disease where this was thought to be relevant. And it
that in order to avoid autoimmunity, self-reactive immune cells became a given that if you were doing serious science, and espe-
are eliminated during fetal development (Burnet, 1959). cially medically relevant ‘‘translation’’ of that science, you had to
But clearly, while negative thymic selection is a real phenom- be working on a mouse model. There also arose the prejudice
enon, as confirmed by the papers cited, the magnitude of its ef- that good science was ‘‘hypothesis driven and mechanistic’’
fects has been greatly exaggerated by implanting specific TCRs and that ‘‘descriptive’’ became an epithet to describe work that
into the mice, which causes the TCRs to be expressed earlier in was primitive and hopelessly old fashioned. This implicitly
thymic development. For the superantigen results, this early discouraged work on human beings because there were few
expression was probably due to the abundance of these partic- methods that could be applied ethically to those studies; most
ular self-antigens and the triggering of many TCRs at once. work could only be descriptive and correlative. But this ignores
It is important to note, though, that these experiments were the the fact that all science starts with description, and close and thor-
only way this issue could be addressed at the time. Tetramers, ough observation is key to discovering new phenomena and
and the sensitivity needed, were many years in the future. But formulating useful hypotheses. A mechanistic understanding is
most importantly, these results force a re-evaluation of how of course the ultimate goal, but it can take many years of observa-
important negative selection is to prevent autoimmunity. Clearly, tion and experiments to get there. Even then, an actual mecha-
it’s not as important as it was thought to be because the evi- nism can be elusive. Even in physics one often settles for an accu-
dence shows that even healthy people have a significant fraction rate formula. The failure of many of these mouse translational
of self-specific T cells, and not just of a low affinity, because models was noted many years ago in autoimmunity (von Herrath
many can bind peptide-MHC tetramers perfectly well. What and Nepom, 2005), cancer immunology (Steinman and Mellman,
this re-evaluation has led to is the fundamental question of 2004), and stem cell biology (Payne and Crooks, 2007). But
what shapes the TCR repertoire—if efficient negative selection also, aside from these critiques, in the academic world, one rarely

1344 Immunity 55, August 9, 2022


ll
Perspective

hears about the many failures of mouse models to "translate" into differently: at least 169 genes, using very stringent criteria. This
effective clinical treatments. This is because this work is almost is not a trivial number and may contribute to the many differences
entirely done by biotech or pharmaceutical companies, and fail- seen phenotypically between the species. Also, it appears that
ures are rarely published because no one wants to spend the there are differences in the magnitude of expression that could
time on them, and journals or reviewers don’t welcome them. make a substantial difference, as indicated by the activation of
This results in a classic "publication bias" where only the good CD4+ T cells between the two species (Shay et al., 2013). There
news gets out there. But those of us who advise companies widely may also be major differences in posttranslational modifications,
(M.M.D.) know that mouse models are viewed with great skepti- but that is much more difficult to assess comprehensively. In
cism by the people who actually need to find useful drugs. Earlier addition, many immunological cues in the form of cytokines
on, these models were almost the only source of information, but and chemokines are provided by non-immunological tissues,
recent advances in technology have created other options. I so this is another potential source of divergence to be investi-
asked Frank Nestle, global head of research and chief scientific gated, which may be particularly relevant to differences in tissue
officer at Sanofi, to comment on how mouse models are viewed or organ architecture, as discussed below.
in biotech and pharma: To this point, the structure of tissues and organs relevant to the
The translational drug discovery paradigm is changing from a immune system between humans and mice suggests functional
mouse-first to a human-first approach. We are in the age of hu- differences. One of these is skin, a major starting point and target
man immunology, where we are tapping into unprecedented of many pathogens. As shown in Figure 1 (Zomer and Trentin,
orthogonal data sets (genetics, single cell transcriptomics, pro- 2018), human skin is much thicker than mice skin (43) and
teomics, metabolomics, real world clinical data, [etc.]) to drive has 5–10 cellular layers, versus 2–3 for mice (Zomer and Trentin,
our target discovery and translational research engine. While 2018). The epidermal layers of skin in both species contain den-
some mechanism-based mouse models retain relevance, dis- dritic, antigen-presenting Langerhans together with CD4+ and
ease-mimicking mouse models are becoming more and more CD8+ T cells, but only mice have the mysterious dendritic
the tools of the last decade. epidermal T cells (DETCs), with their uniform gd T cell receptors
Nevertheless, even though mouse models of disease have (Allison and Havran, 1991; Johnson et al., 2020) apparently per-
often been disappointing as a seamless path to human treat- forming a specific sensory function, but also lacking in ab T cells.
ments, as noted earlier, they have been a gold mine of information The dermis layer in both species contains a broader mix of im-
about how the immune system works and will continue to be of mune cells, ab and gd T cells, innate lymphocytic cells (ILCs),
great value, especially as more details of human diseases are un- DCs, and macrophages. The thickness of human skin also allows
covered, and we can determine exactly where the systems a direct connection to the lymphatic system (Pasparakis et al.,
converge. Progress on finding these convergences informatically 2014). Another striking difference in structure has emerged
has been reported by Shen-Orr and colleagues (Normand et al., from the analysis of human spleens compared with mice
2018). Even better would be a broad program of comparing hu- spleens, which show some striking anatomical differences.
man immune deficiencies and their detail phenotypes with mice Figure 2 from Lewis et al. (2019), with data also drawn from Stei-
with the same genetic lesions; while a number of discordances niger (2015), shows how anatomically different human spleens
have been noted, there has been no systematic program to date. are from mice spleens. While both types of spleens have red
pulp and white pulp regions, in humans, the lymphocyte-rich
Similarities and differences areas (white) (where most of the adaptive immune response oc-
Humans and mice share many, although not all, of the genes and curs) have a very different distribution of T cells, which are
cell types we know as important in human function: the same diffused in human germinal centers (GCs) versus discrete clus-
types of V, D, and J gene segments in T cell receptors and immu- ters in mice; human spleens also have an absence of a clear
noglobulin genes, as well as the same rearrangement apparatus bridging outer layer. Whether these striking differences have
that goes back to early vertebrates, plus many of the same or any impact on immune responses is not clear.
similar cytokine receptors and innate apparatus toll-like recep- DeBoer and colleagues (van Hoeven et al., 2017) have also
tors (TLRs), etc. and MHC molecules, along with basically all noted a striking difference in thymic output and T cell half-life be-
the nuts and bolts of adaptive and innate immunology that we tween mice and humans, with the mice having a much shorter
know about at this time, but with some exceptions. One such half-life than humans; this makes sense in terms of the huge dif-
well-known example is the expression of class II human leuko- ference in life spans between the species. And if other immuno-
cyte antigen (HLA) molecules on activated human T cells, which logical properties are speeded up this way in mice, it means that
is not seen in mice. This suggests that these human T cells can human responses might play out over a much longer timescale.
serve as ‘‘professional antigen-presenting cells’’ and act on Other anomalies have emerged from the study of human immune
CD4+ T cells to promote specific responses independently of B deficiencies in cases where they can be directly compared with
cells or dendritic cells (DCs). Whether this has any major conse- the equivalent mouse model. In the case of CD28, a key co-stim-
quences remains to be seen. Benoist and colleagues at the ulatory molecule, mice lacking this gene have very severe im-
Immunological Genome Project (IMMGEN) consortium have mune dysfunction with susceptibility to multiple pathogens,
done a deep dive into the thousands of genes that they and whereas humans with this deficiency are largely healthy and
others have identified as being expressed in immune cells apparently only susceptible to skin papilloma viruses (Béziat
beyond those having generic functions and find that the vast ma- et al., 2021). Another discordant gene is ISG15, an interferon-
jority of homologs are expressed in the same cell subsets (Shay inducible, ubiquitin-like intracellular protein that is necessary
et al., 2013). However, a considerable number are expressed for protection against viruses in mice; human beings deficient

Immunity 55, August 9, 2022 1345


ll
Perspective
Figure 1. A comparison of mouse and
human skin (Zomer and Trentin, 2018)
Human skin is much thicker and is more complex
than mouse skin. In their immune composition,
mice and humans differ in their epidermal layer,
whereas mice have the unique dendritic epidermal
T cells (DETC) cells with their uniform gd T cell
T cell receptors but share with humans Langer-
hans and CD4 and CD8 T cells. Adapted with
permission from Nature (Pasparakis et al., 2014),
copyright 2014.

These efforts make human compari-


sons problematic, especially for genetics,
where most of the many gene-manipu-
lated mice are within the same strain,
C57 BL/6. A modern fix to this is the
collaborative cross system (Welsh et al.,
2012), which consists of five inbred strains
of mice and three from wild mice that have
been crossed and then backcrossed
in this gene seem to have reasonably effective anti-viral re- extensively to fix each of the scrambled genomes into a separate,
sponses, but deficient microbial defenses (Bogunovic et al., homozygous strain (Iraqi et al., 2008; Shorter et al., 2019). Over
2012; Zhang et al., 2015). 160 separate strains have been produced; with most of these hav-
These comparisons of the same genetic lesions in both spe- ing their genomes sequenced (Shorter et al., 2019), these strains
cies give us some of the clearest evidence we have of impor- have been very valuable in showing how different gene polymor-
tant functional differences between the species but with the phisms in mice can alter infectious disease responses (Noll
important caveat that humans lead much more complex lives et al., 2020; Martin et al., 2020). This system has also been used
in terms of environmental exposure than inbred mice. But in to show how some of these strains mimic human drug responses
many cases, pursuing information about a gene from humans (Zeiss et al., 2019). These are very valuable studies that show how
to mice has been very fruitful and shed new light and context the standard inbred strains give us only a very limited view of ge-
as to its function. This is particularly true in the case of netic influences on important phenomena, but they involve
GCN2, whose expression unexpectedly has arisen in an anal- analyzing sometimes over a hundred different mice (Martin
ysis of a human cohort exposed to a yellow fever vaccine et al., 2020), so it’s a major commitment with no guarantee of rele-
and correlated with an enhanced CD8+ T cell response (Querec vance to human immunology. Research on these mice also
et al., 2009). Analyzing the effects of deleting the murine homo- cannot benefit from the many examples of genetic ablation
log, Pulendran and colleagues have found that this gene is models and other manipulations without a great deal of effort.
involved in the amino acid starvation response and the sup- The lack of constant microbial exposure is also a problem, as
pression of intestinal inflammation, which leads to an enhanced clearly demonstrated by Masopust and colleagues, who very
immune response (Ravindran et al., 2016) and insights into the cleverly introduced mice obtained from pet shops to cohabitate
control of autophagy and reactive oxygen species (ROS). Thus, with C57BL/6 mice and have shown that those that survived had
the mouse system can provide much greater depth to a human acquired tissue resident T cells (Beura et al., 2016). But this
result when the systems are in alignment. Currently, there are would be difficult to do in modern mouse houses, as it would
485 known immunodeficiency genes that have been identified require a separate containment facility so as not to endanger
in human beings (Tangye et al., 2022). In addition, it has many of the users, animals, and studies.
become clear that a number of older individuals make autoan- Finally, in 1988, McCune et al. (1988) introduced severe com-
tibodies against particular cytokines, which attenuate pathogen bined immunodeficiency-Human (SCID-Hu) mice, a novel effort
responses that depend on those cytokines with sometimes to reconstitute immunodeficient mice with a human immune sys-
fatal results, as shown in recent work on COVID-19 patients tem in order to create ‘‘humanized’’ mice that could faithfully model
(Bastard et al., 2020; Chang et al., 2021). human responses. Despite many improvements over the years
(McCune, 1996; Yong et al., 2018; Rongvaux et al., 2013), there
The effects of environment and genetic diversity are still issues with getting good B cell responses, although many
To reduce genetic variation, immunologists have embraced aspects seem to model human immune activities adequately.
inbred mice, originally developed by Snell et al. (Gorer et al., Again, this approach is not able to employ the power of genetic
1948) in the 1950s. More recently, since mouse disease outbreaks modifications that have made inbred mouse strains so powerful.
can wreak havoc on immunological experiments, increasingly
cleaner and more stringent mouse husbandry methods have Do humans have novel immune mechanisms?
been introduced, up to and including ultraclean methodologies There is a reasonable hope that human beings might have unique
that involve rederiving all mouse lines by cesarian delivery. immune mechanisms because they are much less fecund and

1346 Immunity 55, August 9, 2022


ll
Perspective

take much longer to reach sexual maturity. Thus, it’s fine if the functions related to increased longevity and low fecundity. Also
average mouse lives six months or so, but it’s a major problem noteworthy is the large number of unique genes in human DCs,
for survival of the species if enough humans don’t make it to neutrophils, and basophils, which may correlate with the diffi-
25–30 years. This means that if the risk of a fatal infectious dis- culty encountered in modeling allergy and other human pathol-
ease is relatively constant, then humans are 503 more vulner- ogies involving those cells in mice.
able over the average lifetime. Thus, we may find additional Also, this list should not be considered the last word; we in fact
‘‘immunological defense’’ mechanisms in human beings if we missed intercellular adhesion molecule-3 (ICAM3) and TLR10 in
look hard enough. How might we find these if they exist? One this survey, which came up anecdotally, so there are likely
way might be the repurposing of genes that are shared between others.
mice and humans, as discussed above. And thus, those 169 The entire workflow of identifying the human genes, matching
genes with different expression patterns between mice and hu- with immune population genes, and running blastp can be
mans might mediate novel functions (Shay et al., 2013) cited found in GitHub repositories (https://github.com/KwatMDPhD/
earlier. But another way to look for human or primate-specific immune_populations.pro and https://github.com/KwatMDPhD/
mechanisms would be to identify human-specific genes versus human_genes.pro).
mouse-specific genes. Here we have used the Ensemble It was also beneficial to vet candidate genes on the very helpful
Program to search for likely genes that are unique to humans NHLBI website, which compares particular genes across many
versus mice (Table 1). species.
The program compares sequence homologies to determine In the table, it is interesting to note the relatively large num-
whether a human gene is sufficiently different from mice to be ber of human genes in DCs, neutrophils, and basophils. This
considered unique. The list is not long, and while it may just may be an explanation for why many of the anomalies be-
reflect divergent evolution and minor tweaking of functions that tween mice and humans occur in the innate immune mecha-
are well known from the many intensive studies of mouse immu- nisms that these cells mediate. Specifically, DCs play a pivotal
nology, given the many apparent differences seen for years be- role in gathering and presenting antigens to T cells and have
tween mouse models and clinical outcomes, these genes and long been thought to be somewhat different between humans
those identified by Shay and colleagues (Shay et al., 2013) and mice, but exactly how and what the consequences might
may be well worth following up on for clues regarding possible be are unclear. For example, in humans, TLR7 and 9 are
novel immunological mechanisms. mostly expressed in plasmacytoid DCs, and thus, agonists
Although the above examples show that even with respect for these TLRs can only activate these cells, whereas in
to the same molecules and organs, humans and mice exhibit mice TLRs7 and 9 are more broadly expressed on other sub-
clear differences, another way to ask the question is to identify sets of DCs. The power of recent advances in single-cell tran-
genes that are unique in humans because these may lead to scriptional analysis has great potential in illuminating human-
even more profound differences and mechanisms. Thus, we mouse differences, but the limitations are also evident in a
have taken advantage of a rich dataset of RNA sequencing recent analysis of DCs in the two species by Rudensky and
data for 29 different human immune cell subsets that have colleagues. They note clear differences in subsets, but
been isolated and analyzed for gene expression (Monaco because a functional readout is not readily available, whether
et al., 2019). these differences are consequential is not clear, which is a
The table listing all human genes based on Genome Refer- general problem for all of these genes that don’t have a mouse
ence Consortium human build (GRCh38.p13) was downloaded homolog, pointing to the need for a complete human system
from Ensembl BioMart (Kinsella et al., 2011). Genes whose that can be analyzed. Here, humanized mice or immune orga-
‘‘gene type’’ is ‘‘protein coding’’ were selected from the table. noids are possible answers.
Next, the table listing mouse genes based on GRCm39 and The number of unique neutrophil genes is also interesting and
their human homologs were downloaded from Ensembl may underlie some of the observed discordances between mice
BioMart. Human genes from the former table that do not and humans, especially given the very different composition of
have any mouse homologs in the latter were identified as these cells in circulating white blood cells (60%–70% in humans
candidate-unique human protein-coding genes. In addition, versus only 30% in inbred mice) (Junhee et al., 2013; Nauseef,
the National Heart, Lung, and Blood Institute (NHLBI) website 2014, 2019). But again, until we can probe more deeply into a
(https://www.nhlbi.nih.gov/) collates a very extensive collection human assay system, it remains a mystery how significant these
of species data on all known genes, and with this resource we are.
have been able to eliminate many genes that did have identified Lastly, the number of basophil genes is interesting and poten-
homologs in mice and other species. We also used the UniProt tially explains the difficulties in modeling human allergic reac-
blastp program, using the protein sequence to search the tions in mice.
mouse genome to make sure there were not any mouse Our main hope in presenting these results is not to resolve
sequence homologies of significance (Bateman et al., 2021). questions about their importance but to tantalize readers with
The resulting summary of this work is presented in Table 1 the possibility that some of these genes might mediate novel im-
together with some of the information about cell-type distribu- mune mechanisms. We also caution about thinking about indi-
tion and other species that had this particular gene. Many of vidual genes in isolation as they always work in pathways where
these genes have rat homologs, suggesting that their lack in they can be influenced by polymorphisms in other genes or by
mice was not due to very different functionality. But a few different environmental factors such as pathogens or micro-
were exclusively in primates, which would fit with novel immune biome components.

Immunity 55, August 9, 2022 1347


ll
Perspective
Figure 2. Mouse and human splenic
immune cellular architecture at steady state
There are structural differences between the mu-
rine (left) and human (right) splenic immune sys-
tem, most notably the organization of T cell zone
([TC], turquoise; also known as pediatric advanced
life support [PALS]) and B cell zone (BCZ) follicles
(gray and shades of blue, shown with light zone
[LZ] and dark zone [DZ], organization in mouse
spleen) within the WP and the border between the
WP and RP, the marginal zone (MZ) in mouse, or
perifollicular zone (PFZ) in human (dark blue outer
ring). Because applications of advanced imaging
techniques to the human spleen have been
limited, the extent to which the mouse MZ and
human PFZ are analogous remains unknown. For
example, the precise layering and composition of
macrophage subsets in the MZ is known for mice
(see bottom left box). CD169+ MMMs (dark blue)
form a concentric ring around the WP with MZMs
(light blue) and MB cells (darker blue), but not for
humans. In humans, MZB cells surround activated
B cells, containing a GC (light blue in the human
spleen on the right) and corona (gray, "Cor"). The
homeostatic location of DC subsets in mice is
shown (with cDC2s in the bridging channel [BC]
and cDC1s in the TCZ, MZ, and red pulp [RP]).
Release of blood into the MZ of the WP from a
central arteriole (CA) is shown. From Science
Immunology (Lewis et al., 2019). Adapted with
permission from AAAS.

But recent work has shown even


greater subtlety, where work by Picker
and colleagues has shown that the vac-
cine vector that they made was remark-
Another way we are likely to discover additional mechanisms ably effective against both simian immunodeficiency virus (SIV)
in immunology is by taking advantage of the vast number of hu- and tuberculosis (TB) in monkeys (Hansen et al., 2010, 2016; Ma-
man pathogens and learning what their strategies are for avoid- louli et al., 2021; Verweij et al., 2021), and this was accompanied
ing or distracting the immune system. These mechanisms may by a novel HLA-E-restricted T cell response and class II MHC-
not be unique to humans, but the sheer number of human path- restricted CD8+ T cells. Just last year, it was reported that the
ogens compared with the few commonly used in mouse class II-restricted CD8+ T cells were not important for these
models suggests that there is a wealth of insight to be gained remarkable vaccine responses, indicating that the HLA-E ones
and, enabled with recently developed technologies, important were. Also telling was the finding that no fewer than seven
discoveries to be expected. Indeed, Rolf Zinkernagel (Zinkerna- CMV genes seemed to be engaged in diverting T cell responses
gel, 1996) entitled a famous review (‘‘Immunology taught by vi- away from HLA-E (Yang et al., 2021). Thus, while T cell re-
ruses’’) to make this point years ago, but only now do we have sponses against CMV antigens are quite active, accounting for
the ability to analyze human responses to pathogens in depth. an estimated 10%–20% of all CD8+ T cells in carriers, there
Some pathogen strategies are well known, like HIV for are no known cases of the virus being cleared from anyone.
example, which uses an error-prone polymerase to produce Another interesting fact is that in 16 monozygotic twins discor-
an extreme degree of genomic diversity, producing thousands dant for CMV seropositivity, there have been massive changes
of quasispecies in each infected individual, eventually wearing in immune system variables in the positives versus the negatives
down the immune system’s ability to keep up (Bbosa (Brodin et al., 2015). The implications of all this are not very clear
et al., 2019). other than that the virus has things under control, and we have a
T. brucei, a parasite that causes sleeping sickness, is well lot to learn about what it is up to. The same is probably true for
known for employing thousands of possible surface proteins many of our other pathogens as well.
where immune pressure against one type leads to another one
becoming dominant (Cross, 1990). But these strategies are The way forward
very simple compared with the complexities of other pathogens. Almost 14 years ago, one of us (M.M.D.) wrote in this journal a
Cytomegalovirus (CMV) is remarkable in both its lifelong staying plea for the immunology community to not just pay lip service
power and its ability to enhance immune responses against other to human immunity, but to put serious effort into welcoming it
viruses in both mice and humans (Furman et al., 2015). Addition- to the fold as a distinct, but vitally important, complement to
ally, it has a number of mechanisms to subvert the immune sys- the juggernaut that is mouse immunology (Davis, 2008). There
tem, as reviewed by Goodrun et al. (2021). is still a long way to go to catch up to that standard, but human

1348 Immunity 55, August 9, 2022


Perspective
Table 1. Shows genes in human immune cells that are not found in inbred mice and thus may mediate novel mechanisms. But note that many are shared with rats and thus are
likely not related to longevity, but nonetheless may mediate important functions in human beings
The human protein atlas tissue
Gene Immune population specificity Group Top blastp against mouse
GLYATL1B B naive breast, lymphoid tissue glycine-N-acyltransferase-like 1B 0.412 primates – – –
ZNF860 B naive + memory lymphoid tissue zinc fingers C2H2-type 0.438 primates – – –
PLEKHG7 B naive + memory cervix, epididymis, fallopian tube Pleckstrin homology domain 0.258 mammals rats sea urchins –
containing/Dbl family Rho GEFs
PLAAT2 plasmablasts intestine phospholipase A and primates – – –
acyltransferase family
ZNF683 T CD8 TE testis zinc fingers C2H2-type 0.56 mammals rats – –
TRABD2A T naive intestine, lymphoid tissue, ovary TraB domain containing 2A 0.648 broad fish – –
FAAH2 Tfh + Th multiple tissues including lymphoid fatty acid amide hydrolase 2 0.304 broad fish Drosph xeno
GNLY T, NK bone marrow, lymphoid tissue granulysin, cytotoxic T cells, 0.292 broad – – –
NK cells
KIR2DP1 NK lymphoid tissue killer cell immunoglobulin like 0.401 mammals – – –
receptors
KLRF1 NK bone marrow, lymphoid tissue killer cell lectin like receptors/C-type 0.303 mammals – – –
lectin domain containing
LGALS9B NK esophagus, intestine, stomach 1 galectins 0.685 – – – –
LGALS9C NK esophagus, intestine, stomach 1 galectins 0.688 – – – –
CD1B mDCs lymphoid tissue CD molecules/C1-set domain 0.489 mammals – – –
containing
CD1E mDCs lymphoid tissue CD molecules/C1-set domain 0.521 mammals bats chinchilla –
containing
CLIC2 mDCs multiple tissues including lymphoid chloride intracellular channels 0.662 broad rats – –
KCNK17 pDCs lung, thyroid gland potassium two pore domain channel 0.42 broad Drosophila – –
subfamily K
NLRP7 pDCs testis NLR family/pyrin domain containing 0.408 mammals – – –
CTSV pDCs lymphoid tissue cathepsins 0.792 mammals mole rat – –
PROC pDCs liver receptor ligands/Gla domain 0.695 mammals rats – –
containing
Immunity 55, August 9, 2022 1349

SLC9C2 pDCs brain, choroid plexus, testis solute carriers 0.292 mammals rats – –
ADGRE3 granulocytes LD bone marrow, lymphoid tissue adhesion G protein-coupled 0.459 mammals – – –
receptors, subfamily E
NT5DC4 monocytes I testis 50 -nucleotidase domain containing 4 0.654 broad – – –
CASP5 monocytes NC brain, intestine, lymphoid tissue caspases|caspase recruitment 0.636 broad – – –
domain containing
LILRA1 monocytes NC + I lymphoid tissue CD molecules/activating leukocyte 0.507 primates – – –

ll
immunoglobulin like receptors
(Continued on next page)
1350 Immunity 55, August 9, 2022

Table 1. Continued
The human protein atlas tissue
Gene Immune population specificity Group Top blastp against mouse

ll
LILRB2 monocytes NC + I bone marrow, lung, lymphoid tissue CD molecules/inhibitory leukocyte 0.452 mammals rats – –
immunoglobulin-like receptors/Ig-
like cell adhesion molecule family
LILRA2 myeloid bone marrow, lymphoid tissue CD molecules/activating leukocyte 0.513 primates – – –
immunoglobulin like receptors
SIGLEC14 myeloid bone marrow, lymphoid tissue V-set domain containing/sialic acid 0.498 mammals
binding Ig-like lectins
– – lung, lymphoid tissue – – – – – –
OR14L1P basophils LD brain olfactory receptors, family 14 0.53 mammals reptiles – –
OR6K3 basophils LD bone marrow olfactory receptors, family 6 0.784 mammals rats – –
OXER1 basophils LD liver leukotriene receptors 0.418 mammals – – –
TRIM49D1 basophils LD undetected ring finger proteins/tripartite motif 0.364 primates – – –
containing
TRIM49D2 basophils LD undetected ring finger proteins/tripartite motif 0.364 primates – – –
containing
TRIM51 basophils LD undetected ring finger proteins/tripartite motif 0.367 – – – –
containing
TRIM64 basophils LD placenta ring finger proteins/tripartite motif 0.348 – – – –
containing
TRIM64B basophils LD placenta ring finger proteins/tripartite motif 0.348 – – – –
containing
FCAR neutrophils LD bone marrow, lung, lymphoid tissue CD molecules/immunoglobulin-like 0.327 – – – –
domain containing/Fc receptors
H2AC19 neutrophils LD multiple tissues including lymphoid H2A histones 1 – – – –
CSNK1A1L neutrophils LD testis casein kinase 1 family 0.91 broad Drosophila – –
LINC02218 neutrophils LD undetected long intergenic non-protein 0.441 – – – –
coding RNAs
PEAK3 neutrophils LD bone marrow, lymphoid tissue PEAK family member 3 0.268 broad fish – –
PI3 neutrophils LD esophagus, lymphoid tissue, vagina WAP four-disulfide core domain 0.941 broad chkns – –
containing
S100A12 neutrophils LD bone marrow S100 calcium binding proteins/EF- 0.388 mammals rats – –
hand domain containing
SIRPB2 neutrophils LD lymphoid tissue V-set domain containing/signal 0.348 mammals rats – –

Perspective
regulatory proteins
SIRPD neutrophils LD testis V-set domain containing/signal 0.48 mammals rats – –
regulatory proteins
ST20 neutrophils LD multiple tissues including lymphoid suppressor of tumorigenicity 20 0.724 ? – – –
TMEM272 neutrophils LD brain transmembrane protein 272 0.368 mammals – – –
ll
Perspective

immunology is definitely getting there. And it isn’t just conceptual novel gene featuring two PHD-type zinc-finger domains. Nat. Genet. 17,
399–403. https://doi.org/10.1038/ng1297-399.
or abstract anymore, because in the last 2+ years, we have all
been affected by this horrendous pandemic and it has had Allison, J.P., and Havran, W.L. (1991). The immunobiology of T cells with
such a wide range of health effects, from negligible symptoms invariant gd antigen receptors. Annu. Rev. Immunol.
to painful death. It has shown that serious, life-threatening infec- Bastard, P., Rosen, L.B., Zhang, Q., Michailidis, E., Hoffmann, H.H., Zhang, Y.,
tions are not just something that happen to unfortunate countries Dorgham, K., Philippot, Q., Rosain, J., Béziat, V., et al. (2020). Autoantibodies
far away from most of us; it can also be close and very real. And against type I IFNs in patients with life-threatening COVID-19. Science 370,
eabd4585. https://doi.org/10.1126/science.abd4585.
as immunologists, this has to hit close to home and make clear
the urgency of why understanding the human side of immu- UniProt Consortium, Martin, M.J., Orchard, S., Magrane, M., Agivetova, R.,
Ahmad, S., Alpi, E., Bowler-Barnett, E.H., Britto, R., Bursteinas, B., et al.
nology, with its complexity and wide variations, is critical. And (2021). UniProt: the universal protein knowledgebase in 2021. Nucleic Acids
even as this pandemic fades, as it might, another is brewing Res. 49, D480–D489. https://doi.org/10.1093/nar/gkaa1100.
somewhere.
Bbosa, N., Kaleebu, P., and Ssemwanga, D. (2019). HIV subtype diversity
Clearly, inbred mice and the amazing technology built up worldwide. Curr. Opin. HIV AIDS 14, 153–160. https://doi.org/10.1097/COH.
over the past 60+ years are such a powerful and useful system 0000000000000534.
that it will continue to be a mainstay of the field for the fore-
Bennett, C.L., Christie, J., Ramsdell, F., Brunkow, M.E., Ferguson, P.J., White-
seeable future. Even when investigating new human phenom- sell, L., Kelly, T.E., Saulsbury, F.T., Chance, P.F., and Ochs, H.D. (2001). The
ena, being able to go to mice to investigate mechanisms and/ immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome
(IPEX) is caused by mutations of FOXP3. Nat. Genet. 27, 20–21. https://doi.
or confirm hypotheses has been invaluable to my group and org/10.1038/83713.
many others. But it is also important not to ignore the many
variances, whether genetic, anatomical, or in gene expres- Beura, L.K., Hamilton, S.E., Bi, K., Schenkel, J.M., Odumade, O.A., Casey,
K.A., Thompson, E.A., Fraser, K.A., Rosato, P.C., Filali-Mouhim, A., et al.
sion. It is unlikely that all of these represent minor tweaks in (2016). Normalizing the environment recapitulates adult human immune traits
the human immune system, and so we could be missing in laboratory mice. Nature 532, 512–516. https://doi.org/10.1038/
important aspects of human immunity. While humanized nature17655.

mice might represent part of the answer in exploring these Béziat, V., Rapaport, F., Hu, J., Titeux, M., Bonnet des Claustres, M., Bourgey,
anomalies, there will always be questions about how faithfully M., Griffin, H., Bandet, É., Ma, C.S., Sherkat, R., et al. (2021). Humans with in-
herited T cell CD28 deficiency are susceptible to skin papillomaviruses but are
these hybrid animals can represent human immunity, espe- otherwise healthy. Cell 184, 3812–3828.e30. https://doi.org/10.1016/j.cell.
cially given the constraints of mouse husbandry. Work on 2021.06.004.
monkeys is one solution, which has been used to great effect
Bogunovic, D., Byun, M., Durfee, L.A., Abhyankar, A., Sanal, O., Mansouri, D.,
in many studies, but they are extremely expensive and in short Salem, S., Radovanovic, I., Grant, A.V., Adimi, P., et al. (2012). Mycobacterial
supply. Thus, the advent of human immune organoids using disease and impaired IFN-g immunity in humans with inherited ISG15 defi-
discarded tonsils or spleens may be the key to being to be ciency. Science 337, 1684–1688. https://doi.org/10.1126/science.1224026.

able to do mechanistic and hypothesis-driven experiments Brodin, P., Jojic, V., Gao, T., Bhattacharya, S., Angel, C.J.L., Furman, D.,
that will be needed (Wagar et al., 2021). The advent of gene Shen-Orr, S., Dekker, C.L., Swan, G.E., Butte, A.J., et al. (2015). Variation in
the human immune system is largely driven by non-heritable influences. Cell
modifications using CRIPR-Cas9 is also introducing powerful 160, 37–47. https://doi.org/10.1016/j.cell.2014.12.020.
ways to screen for important genes such as the genome-
wide screen of CD8 T cells by Marson and colleagues or early Brunkow, M.E., Jeffery, E.W., Hjerrild, K.A., Paeper, B., Clark, L.B., Yasayko,
S.A., Wilkinson, J.E., Galas, D., Ziegler, S.F., and Ramsdell, F. (2001). Disrup-
work on immune and intestinal organoids (Shifrut et al., 2018; tion of a new forkhead/winged-helix protein, scurfin, results in the fatal lym-
Fujii et al., 2015; Wu et al., 2018). phoproliferative disorder of the scurfy mouse. Nat. Genet. 27, 68–73. https://
doi.org/10.1038/83784.
In conclusion, while human immunology is still very much a
work in progress, there is so much potential and uncharted ter- Burnet, F.M. (1959). The Clonal Selection Theory of Acquired Immunity (Van-
ritory to explore, and with such a strong foundation from mouse derbilt University Press).
immunology and continued interactivity between the two sys- Casanova, J.L., and Abel, L. (2005). Inborn errors of immunity to infection: the
tems, one can’t help but to be optimistic about what we will learn rule rather than the exception. J. Exp. Med. 202, 197–201. https://doi.org/10.
in the coming decades. 1084/jem.20050854.

Chang, S.E., Feng, A., Meng, W., Apostolidis, S.A., Mack, E., Artandi, M.,
ACKNOWLEDGMENTS Barman, L., Bennett, K., Chakraborty, S., Chang, I., et al. (2021). New-onset
IgG autoantibodies in hospitalized patients with COVID-19. Nat. Commun.
12, 5417. https://doi.org/10.1038/s41467-021-25509-3.
We thank J.L. Casanova, Bali Pulendran, Paul Kubes, Stephanie Eisenbarth,
Karan Kathuria, Juliana Idoyaga, Guangbo Chen, Chen Wang, Xin Chen, Ed Cross, G.A. (1990). Glycolipid anchoring of plasma membrane proteins. Annu.
Mocarski, and John Boothroyd. We thank the Howard Hughes Medical Insti- Rev. Cell Biol. 6, 1–39. https://doi.org/10.1146/annurev.cb.06.110190.
tute, the Open Philantropy Foundation, and NIAID AI057229 for support. 000245.

DECLARATION OF INTERESTS Davis, M.M. (2008). A prescription for human immunology. Immunity 29,
835–838. https://doi.org/10.1016/j.immuni.2008.12.003.

The authors declare no competing interests. Day, C.L., Seth, N.P., Lucas, M., Appel, H., Gauthier, L., Lauer, G.M., Robbins,
G.K., Szczepiorkowski, Z.M., Casson, D.R., Chung, R.T., et al. (2003). Ex vivo
analysis of human memory CD4 T cells specific for hepatitis C virus using MHC
REFERENCES
class II tetramers. J. Clin. Invest. 112, 831–842. https://doi.org/10.1172/
JCI200318509.
Finnish-German APECED Consortium, Björses, P., Perheentupa, J., Horelli-
Kuitunen, N., Palotie, A., Peltonen, L., Lee Su, Y., Francis, F., Hennig, S., Thiel, Dobzhansky, T. (1973). Nothing in biology makes sense except in the light of
C., et al. (1997). An autoimmune disease, APECED, caused by mutations in a evolution. Am. Biol. Teach. 35, 125–129. https://doi.org/10.2307/4444260.

Immunity 55, August 9, 2022 1351


ll
Perspective
Fujii, M., Matano, M., Nanki, K., and Sato, T. (2015). Efficient genetic engineer- Collaborative Cross. Cell Rep. 31, 107508. https://doi.org/10.1016/j.celrep.
ing of human intestinal organoids using electroporation. Nat. Protoc. 10, 1474– 2020.03.072.
1485. https://doi.org/10.1038/nprot.2015.088.
Mathis, D., and Benoist, C. (2007). A decade of AIRE. Nat. Rev. Immunol. 7,
Furman, D., Jojic, V., Sharma, S., Shen-Orr, S.S., Angel, C.J.L., Onengut-Gu- 645–650. https://doi.org/10.1038/nri2136.
muscu, S., Kidd, B.A., Maecker, H.T., Concannon, P., Dekker, C.L., et al.
(2015). Cytomegalovirus infection enhances the immune response to influ- McCune, J.M. (1996). Development and applications of the SCID-hu mouse
enza. Sci. Transl. Med. 7, 281ra43. https://doi.org/10.1126/scitranslmed. model. Semin. Immunol. 8, 187–196. https://doi.org/10.1006/smim.
aaa2293. 1996.0024.

Goodnow, C.C., Crosbie, J., Adelstein, S., Lavoie, T.B., Smith-Gill, S.J., Brink, McCune, J.M., Namikawa, R., Kaneshima, H., Shultz, L.D., Lieberman, M., and
R.A., Pritchard-Briscoe, H., Wotherspoon, J.S., Loblay, R.H., Raphael, K., Weissman, I.L. (1988). The SCID-hu mouse: Murine model for the analysis of
et al. (1988). Altered immunoglobulin expression and functional silencing of human hematolymphoid differentiation and function. Science 241, 1632–
self-reactive B lymphocytes in transgenic mice. Nature 334, 676–682. 1639. https://doi.org/10.1126/science.2971269.
https://doi.org/10.1038/334676a0.
Miller, J.F. (1993). Self-nonself discrimination and tolerance in T and B lympho-
Goodnow, C.C., Crosbie, J., Jorgensen, H., Brink, R.A., and Basten, A. (1989). cytes. Immunol. Res. 12, 115–130. https://doi.org/10.1007/BF02918299.
Induction of self-tolerance in mature peripheral B lymphocytes. Nature 342,
385–391. https://doi.org/10.1038/342385a0. Miller, J.F.A.P., and Morahan, G. (1992). Peripheral T cell tolerance. Annu. Rev.
Immunol.
Goodrun, F., Britt, W., and Mocarski, E.S. (2021). Cytomagalovirus. In Fields
Virology. Monaco, G., Lee, B., Xu, W., Mustafah, S., Hwang, Y.Y., Carré, C., Burdin, N.,
Visan, L., Ceccarelli, M., Poidinger, M., et al. (2019). RNA-Seq Signatures
Gorer, P.A., Lyman, S., Snell, G.D., and Haldane, J.B.S. (1948). Studies on the Normalized by mRNA Abundance Allow Absolute Deconvolution of Human Im-
genetic and antigenic basis of tumour transplantation Linkage between a his- mune Cell Types. Cell Rep. 26, 1627–1640.e7. https://doi.org/10.1016/j.cel-
tocompatibility gene and ’fused’ in mice. Proceedings of the Royal Society. rep.2019.01.041.
Proc. R. Soc. B. 135, 499–505.
Moon, J.J., Chu, H.H., Pepper, M., McSorely, S.J., Jameson, S.C., Kedl, R.M.,
Hansen, S.G., Powers, C.J., Richards, R., Ventura, A.B., Ford, J.C., Siess, D., and Jenkins, M.K. (2007). Naive CD4(+) T cell frequency varies for different epi-
Axthelm, M.K., Nelson, J.A., Jarvis, M.A., Picker, L.J., and Fru€h, K. (2010). topes and predicts repertoire diversity and response magnitude. Immunity. 27,
Evasion of CD8+ T cells is critical for superinfection by cytomegalovirus. Sci- 203–213.
ence 328, 102–106. https://doi.org/10.1126/science.1185350.
Nagamine, K., Peterson, P., Scott, H.S., Kudoh, J., Minoshima, S., Heino, M.,
Hansen, S.G., Wu, H.L., Burwitz, B.J., Hughes, C.M., Hammond, K.B., Ven- Krohn, K.J., Lalioti, M.D., Mullis, P.E., Antonarakis, S.E., et al. (1997). Positional
tura, A.B., Reed, J.S., Gilbride, R.M., Ainslie, E., Morrow, D.W., et al. (2016). cloning of the APECED gene. Nat. Genet. 17, 393–398. https://doi.org/10.
Broadly targeted CD8+ T cell responses restricted by major histocompatibility 1038/ng1297-393.
complex E. Science 351, 714–720. https://doi.org/10.1126/science.aac9475.
Nauseef, W.M. (2014). Proteases, neutrophils, and periodontitis: The NET ef-
Herman, A., Kappler, J.W., Marrack, P., and Pullen, A.M. (1991). Superanti- fect. J. Clin. Invest. 124, 4237–4239. https://doi.org/10.1172/JCI77985.
gens: mechanism of T-cell stimulation and role in immune responses. Annu.
Rev. Immunol. Nauseef, W.M. (2019). The phagocyte NOX2 NADPH oxidase in microbial
killing and cell signaling. Curr. Opin. Immunol. 60, 130–140. https://doi.org/
Iraqi, F.A., Churchill, G., and Mott, R. (2008). The collaborative cross, devel- 10.1016/j.coi.2019.05.006.
oping a resource for mammalian systems genetics: a status report of the well-
come trust cohort. Mamm. Genome 19, 379–381. https://doi.org/10.1007/ Noll, K.E., Whitmore, A.C., West, A., McCarthy, M.K., Morrison, C.R., Plante,
s00335-008-9113-1. K.S., Hampton, B.K., Kollmus, H., Pilzner, C., Leist, S.R., et al. (2020). Complex
genetic architecture underlies regulation of influenza-A-virus-specific antibody
Johnson, M.D., Witherden, D.A., and Havran, W.L. (2020). The Role of Tissue- responses in the collaborative cross. Cell Rep. 31, 107587. https://doi.org/10.
resident T cells in stress surveillance and tissue maintenance. Cells 9. https:// 1016/j.celrep.2020.107587.
doi.org/10.3390/cells9030686.
Normand, R., Du, W., Briller, M., Gaujoux, R., Starosvetsky, E., Ziv-Kenet, A.,
Judson, H.F. (1996). The Eighth Day of Creation; Makers of the Revolution in Shalev-Malul, G., Tibshirani, R.J., and Shen-Orr, S.S. (2018). Found In Trans-
Biology (Cold Spring Harbor Laboratory Press). lation: a machine learning model for mouse-to-human inference. Nat Methods.
15, 1067–1073.
Seok, J., Warren, H.S., Cuenca, A.G., Mindrinos, M.N., Baker, H.V., Xu, W., Ri-
chards, D.R., McDonald-Smith, G.P., Gao, H., Hennessy, L., et al. (2013). Ohashi, P.S., Oehen, S., Buerki, K., Pircher, H., Ohashi, C.T., Odermatt, B.,
Genomic responses in mouse models poorly mimic human inflammatory dis- Malissen, B., Zinkernagel, R.M., and Hengartner, H. (1991). Ablation of "toler-
eases. Proc. Natl. Acad. Sci. USA. 110, 3507–3512. https://doi.org/10.1073/ ance" and induction of diabetes by virus infection in viral antigen transgenic
pnas.1222878110. mice. Cell 65, 305–317. https://doi.org/10.1016/0092-8674(91)90164-T.

€ ha
Kinsella, R.J., Ka €ri, A., Haider, S., Zamora, J., Proctor, G., Spudich, G., Al- Pasparakis, M., Haase, I., and Nestle, F.O. (2014). Mechanisms regulating skin
meida-King, J., Staines, D., Derwent, P., Kerhornou, A., et al. (2011). Ensembl immunity and inflammation. Nat. Rev. Immunol. 14, 289–301. https://doi.org/
BioMarts: A Hub for Data Retrieval across Taxonomic Space. Database 2011. 10.1038/nri3646.
bar030. https://doi.org/10.1093/database/bar030.
Payne, K.J., and Crooks, G.M. (2007). Immune-cell lineage commitment:
Legoux, F.P., Lim, J.B., Cauley, A.W., Dikiy, S., Ertelt, J., Mariani, T.J., Spar- translation from mice to humans. Immunity 26, 674–677. https://doi.org/10.
wasser, T., Way, S.S., and Moon, J.J. (2015). CD4+ T cell tolerance to tis- 1016/j.immuni.2007.05.011.
sue-restricted self antigens is mediated by antigen-specific regulatory
T cells rather than deletion. Immunity 43, 896–908. https://doi.org/10.1016/j. Pike, B.L., Boyd, A.W., and Nossal, G.J. (1982). Clonal anergy: the universally
immuni.2015.10.011. anergic B lymphocyte. Proc. Natl. Acad. Sci. USA. 79, 2013–2017. https://doi.
org/10.1073/pnas.79.6.2013.
Lewis, S.M., Williams, A., and Eisenbarth, S.C. (2019). Structure and function
of the immune system in the spleen. Sci. Immunol. 4, eaau6085. https://doi. Pulendran, B., and Davis, M.M. (2020). The science and medicine of human
org/10.1126/sciimmunol.aau6085. immunology. Science 369, eaay4014. https://doi.org/10.1126/science.
aay4014.
Malouli, D., Hansen, S.G., Hancock, M.H., Hughes, C.M., Ford, J.C., Gilbride,
R.M., Ventura, A.B., Morrow, D., Randall, K.T., Taher, H., et al. (2021). Cytome- Querec, T.D., Akondy, R.S., Lee, E.K., Cao, W., Nakaya, H.I., Teuwen, D., Pi-
galoviral determinants of CD8+T cell programming and RhCMV/SIV vaccine rani, A., Gernert, K., Deng, J., Marzolf, B., et al. (2009). Systems biology
efficacy. Sci. Immunol. 6, eabg5413. https://doi.org/10.1126/SCIIMMUNOL. approach predicts immunogenicity of the yellow fever vaccine in humans.
ABG5413. Nat. Immunol. 10, 116–125. https://doi.org/10.1038/ni.1688.

Martin, M.D., Sompallae, R., Winborn, C.S., Harty, J.T., and Badovinac, V.P. Ravindran, R., Loebbermann, J., Nakaya, H.I., Khan, N., Ma, H., Gama, L., Ma-
(2020). Diverse CD8 T Cell Responses to Viral Infection Revealed by the chiah, D.K., Lawson, B., Hakimpour, P., Wang, Y.C., et al. (2016). The amino

1352 Immunity 55, August 9, 2022


ll
Perspective
acid sensor GCN2 controls gut inflammation by inhibiting inflammasome acti- Von Boehmer, H. (1990). Developmental biology of T cells in T cell-receptor
vation. Nature 531, 523–527. https://doi.org/10.1038/nature17186. transgenic mice. Annu. Rev. Immunol.

Röcken, M., Urban, J.F., and Shevach, E.M. (1992). Infection breaks T-cell von Herrath, M.G., and Nepom, G.T. (2005). Lost in translation: barriers to im-
tolerance. Nature 359, 79–82. https://doi.org/10.1038/359079a0. plementing clinical immunotherapeutics for autoimmunity. J. Exp. Med. 202,
1159–1162. https://doi.org/10.1084/jem.20051224.
Rongvaux, A., Takizawa, H., Strowig, T., Willinger, T., Eynon, E.E., Flavell, R.A.,
and Manz, M.G. (2013). Human hemato-lymphoid system mice: Current use Wagar, L.E., Salahudeen, A., Constantz, C.M., Wendel, B.S., Lyons, M.M.,
and future potential for medicine. Annu. Rev. Immunol. Mallajosyula, V., Jatt, L.P., Adamska, J.Z., Blum, L.K., Gupta, N., et al.
(2021). Modeling human adaptive immune responses with tonsil organoids.
Rudensky, A.Y. (2011). Regulatory T cells and Foxp3. Immunol. Rev. 241,
Nat. Med. 27, 125–135. https://doi.org/10.1038/s41591-020-01145-0.
260–268. https://doi.org/10.1111/j.1600-065X.2011.01018.x.

Shay, T., Jojic, V., Zuk, O., Rothamel, K., Puyraimond-Zemmour, D., Feng, T., Welsh, C.E., Miller, D.R., Manly, K.F., Wang, J., McMillan, L., Morahan, G.,
Wakamatsu, E., Benoist, C., Koller, D., and Regev, A.; ImmGen Consortium Mott, R., Iraqi, F.A., Threadgill, D.W., and De Villena, F.P.M. (2012). Status
(2013). Conservation and divergence in the transcriptional programs of the hu- and access to the collaborative cross population. Mamm. Genome 23,
man and mouse immune systems. Proc Natl Acad Sci USA. 110, 2946–2951. 706–712. https://doi.org/10.1007/s00335-012-9410-6.

Shifrut, E., Carnevale, J., Tobin, V., Roth, T.L., Woo, J.M., Bui, C.T., Li, P.J., Di- Wu, C.A.M., Roth, T.L., Baglaenko, Y., Ferri, D.M., Brauer, P., Zuniga-Pflucker,
olaiti, M.E., Ashworth, A., and Marson, A. (2018). Genome-wide CRISPR J.C., Rosbe, K.W., Wither, J.E., Marson, A., and Allen, C.D.C. (2018). Genetic
Screens in Primary Human T Cells Reveal Key Regulators of Immune Function. engineering in primary human B cells with CRISPR-Cas9 ribonucleoproteins.
Cell 175, 1958–1971.e15. https://doi.org/10.1016/j.cell.2018.10.024. J. Immunol. Methods 457, 33–40. https://doi.org/10.1016/j.jim.2018.03.009.

Shorter, J.R., Najarian, M.L., Bell, T.A., Blanchard, M., Ferris, M.T., Hock, P., Yang, H., Rei, M., Brackenridge, S., Brenna, E., Sun, H., Abdulhaqq, S., Liu,
Kashfeen, A., Kirchoff, K.E., Linnertz, C.L., Sigmon, J.S., et al. (2019). Whole M.K.P., Ma, W., Kurupati, P., Xu, X., et al. (2021). HLA-E-restricted, Gag-spe-
genome sequencing and progress toward full inbreeding of the mouse collab- cific CD8+T cells can suppress HIV-1 infection, offering vaccine opportunities.
orative cross population. G3: Genes, Genomes, Genetics 9, 1303–1311. Sci. Immunol. 6, eabg1703. https://doi.org/10.1126/SCIIMMUNOL.ABG1703.
https://doi.org/10.1534/g3.119.400039.
Yong, K.S.M., Her, Z., and Chen, Q. (2018). Humanized mice as unique tools
Steiniger, B.S. (2015). Human spleen microanatomy: why mice do not suffice. for human-specific studies. Arch. Immunol. Ther. Exp. 66, 245–266. https://
Immunology 145, 334–346. https://doi.org/10.1111/imm.12469. doi.org/10.1007/s00005-018-0506-x.
Steinman, R.M., and Mellman, I. (2004). Immunotherapy: bewitched, bothered, €ller, S., Lund, P.J., Juang, J.,
Yu, W., Jiang, N., Ebert, P.J.R., Kidd, B.A., Mu
and bewildered no more. Science 305, 197–200. https://doi.org/10.1126/sci- Adachi, K., Tse, T., Birnbaum, M.E., et al. (2015). Clonal deletion prunes but
ence.1099688. does not eliminate self-specific ab CD8+ T lymphocytes. Immunity 42,
929–941. https://doi.org/10.1016/j.immuni.2015.05.001.
Su, L.F., Kidd, B.A., Han, A., Kotzin, J.J., and Davis, M.M. (2013). Virus-specific
CD4+ memory-phenotype T cells are abundant in unexposed adults. Immunity
Zeiss, C.J., Gatti, D.M., Toro-Salazar, O., Davis, C., Lutz, C.M., Spinale, F.,
38, 373–383. https://doi.org/10.1016/j.immuni.2012.10.021.
Stearns, T., Furtado, M.B., and Churchill, G.A. (2019). Doxorubicin-induced
Tangye, S.G., Al-Herz, W., Bousfiha, A., Cunningham-Rundles, C., Franco, cardiotoxicity in Collaborative Cross (CC) mice recapitulates individual cardi-
J.L., Holland, S.M., Klein, C., Morio, T., Oksenhendler, E., Picard, C., et al. otoxicity in humans. G3 (Bethesda) 9, 2637–2646. https://doi.org/10.1534/
(2022). Human inborn errors of immunity: 2022 update on the classification g3.119.400232.
from the international union of immunological societies expert committee.
J. Clin. Immunol. https://doi.org/10.1007/s10875-022-01289-3. Zhang, X., Bogunovic, D., Payelle-Brogard, B., Francois-Newton, V., Speer,
S.D., Yuan, C., Volpi, S., Li, Z., Sanal, O., Mansouri, D., et al. (2015). Human
van Hoeven, V., Drylewicz, J., Westera, L., den Braber, I., Mugwagwa, T., Tes- intracellular ISG15 prevents interferon-a/b over-amplification and auto-inflam-
selaar, K., Borghans, J.A.M., and de Boer, R.J. (2017). Dynamics of recent mation. Nature 517, 89–93. https://doi.org/10.1038/nature13801.
thymic emigrants in young adult mice. Front. Immunol. 8, 933. https://doi.
org/10.3389/fimmu.2017.00933. Zinkernagel, R.M. (1996). Immunology taught by viruses. Science 271,
173–178.
Verweij, M.C., Hansen, S.G., Iyer, R., John, N., Malouli, D., Morrow, D., Scholz,
I., Womack, J., Abdulhaqq, S., Gilbride, R.M., et al. (2021). Modulation of Zomer, H.D., and Trentin, A.G. (2018). Skin wound healing in humans and
MHC-E transport by viral decoy ligands is required for RhCMV/SIV vaccine ef- mice: Challenges in translational research. J. Dermatol. Sci. 90, 3–12.
ficacy. Science 372, eabe9233. https://doi.org/10.1126/science.abe9233. https://doi.org/10.1016/j.jdermsci.2017.12.009.

Immunity 55, August 9, 2022 1353

You might also like