ArticlePDF AvailableLiterature Review

Against all odds: The road to success in the development of human immune reconstitution mice

Wiley
Animal Models and Experimental Medicine
Authors:

Abstract

The mouse genome has a high degree of homology with the human genome, and its physiological, biochemical, and developmental regulation mechanisms are similar to those of humans; therefore, mice are widely used as experimental animals. However, it is undeniable that interspecies differences between humans and mice can lead to experimental errors. The differences in the immune system have become an important factor limiting current immunological research. The application of immunodeficient mice provides a possible solution to these problems. By transplanting human immune cells or tissues, such as peripheral blood mononuclear cells or hematopoietic stem cells, into immunodeficient mice, a human immune system can be reconstituted in the mouse body, and the engrafted immune cells can elicit human‐specific immune responses. Researchers have been actively exploring the development and differentiation conditions of host recipient animals and grafts in order to achieve better immune reconstitution. Through genetic engineering methods, immunodeficient mice can be further modified to provide a favorable developmental and differentiation microenvironment for the grafts. From initially only being able to reconstruct single T lymphocyte lineages, it is now possible to reconstruct lymphoid and myeloid cells, providing important research tools for immunology‐related studies. In this review, we compare the differences in immune systems of humans and mice, describe the development history of human immune reconstitution from the perspectives of immunodeficient mice and grafts, and discuss the latest advances in enhancing the efficiency of human immune cell reconstitution, aiming to provide important references for immunological related researches.
Anim Models Exp Med. 2024;00:1–11.
|
1wileyonlinelibrary.com/journal/ame2
Received: 1 Februa ry 2024 
|
Accepted: 17 March 2024
DOI: 10.1002 /ame2.12407
REVIEW
Against all odds: The road to success in the development of
human immune reconstitution mice
Yixiao Bin1,2,3| Jing Ren1,2,3| Haowei Zhang4| Tianjiao Zhang2,3| Peijuan Liu2,3|
Zhiqian Xin2,3| Haijiao Yang2,3| Zhuan Feng2,3| Zhinan Chen2,3| Hai Zhang2,3
This is an op en access arti cle under the ter ms of the Creative Commons Attribution L icense, which pe rmits use, dis tribu tion and reprod uction in any med ium,
provide d the original wor k is properly cited.
© 2024 The Aut hors. Animal Models and Experimental Medicine publish ed by John W iley & Sons Australia, Ltd on behalf of The Chinese A ssociation for
Labor atory Animal Sciences.
1School of B asic Medical Sciences , Shaanxi
University of Ch inese Medicine, Xianyang,
China
2Depar tment of Cell Biology, National
Translational Science Cente r for Molecular
Medicine, Four th Military Medical
University, Xi'an , China
3State Key Laboratory of New Targets
Discover y and Drug Development for
Major Dis eases , Fourt h Milit ary Me dical
University, Xi'an , China
4Depar tment of Occupat ional &
Environmental H ealth a nd the Ministr y of
Education Key Lab of Hazard A ssessment
and Control in Special Op erational
Environment, School of Public He alth,
Fourth Milita ry Medical University, Xi'an,
China
Correspondence
Zhinan Chen and Hai Zhang, Depar tment
of Cell Biology, National Translational
Science C enter for Molecular Medicine,
Fourth Milita ry Medical University, Xi'an,
China.
Email: znchen@fmmu.edu.cn and hzhang@
fmmu.eud.cn
Funding information
Scientific and Technological Resources
Coordination Project of Shaanxi Province,
Grant /Award Number: 2020 PT- 002,
2022PT- 43 and CX- PT- 18; Special
Fund for Militar y Laborator y Animals,
Grant /Award Number: SYDW_KY
(2021)13; State Key Laborator y of
Holistic Integr ative Ma nageme nt of
Gastrointestinal Cancers , Grant/Award
Number: CBSKL2022ZZ28
Abstract
The mouse genome has a high degree of homology with the human genome, and its
physiological, biochemical, and developmental regulation mechanisms are similar to
those of humans; therefore, mice are widely used as experimental animals. However,
it is undeniable that interspecies differences between humans and mice can lead to
experimental errors. The differences in the immune system have become an impor-
tant factor limiting current immunological research. The application of immunodefi-
cient mice provides a possible solution to these problems. By transplanting human
immune cells or tissues, such as peripheral blood mononuclear cells or hematopoietic
stem cells, into immunodeficient mice, a human immune system can be reconstituted
in the mouse body, and the engrafted immune cells can elicit human- specific immune
responses. Researchers have been actively exploring the development and differen-
tiation conditions of host recipient animals and grafts in order to achieve better im-
mune reconstitution. Through genetic engineering methods, immunodeficient mice
can be further modified to provide a favorable developmental and differentiation
microenvironment for the grafts. From initially only being able to reconstruct single
T lymphocyte lineages, it is now possible to reconstruct lymphoid and myeloid cells,
providing important research tools for immunology- related studies. In this review, we
compare the differences in immune systems of humans and mice, describe the devel-
opment history of human immune reconstitution from the perspectives of immuno-
deficient mice and grafts, and discuss the latest advances in enhancing the efficiency
of human immune cell reconstitution, aiming to provide important references for im-
munological related researches.
KEYWORDS
hematopoietic stem cell, human immune reconstitution, immune response, immunodeficient
mice, peripheral blood mononuclear cell, transplantation
2 
|
    BIN et a l.
1 | INTRODUC TION
The mouse has become the most widely used animal in biomedical
research due to its small size, fast reproduction, well- defined genetic
background, and ease of manipulation. Over the last 100 years, thou-
sands of different strains of inbred, outbred, hybrid, and mutant mice
have been artificially bred for biomedical research1; however, due to
the interspecies differences between humans and mice, the exper-
imental results obtained in mice cannot be directly generalized to
humans. Thus, it is necessary to develop a model organism that can
closely simulate human pathological and physiological responses.
The immune response is an important mechanism for maintaining
homeostasis in the body; however, for ethical reasons, the majority of
experiments can only be conducted in mice, and the observations of
immune responses in mice provide evidence for human immunological
reactions. There are differences in immune cell composition, immune
cell distribution, and immune molecule expression between humans
and mice, thus making it difficult to deduce the overall picture of human
immune responses based on results obtained in mice.2 For example, the
first therapeutic antibody drug approved by the U.S. Food and Drug
Administration, OKT3, did not induce cytokine storms in mice and mon-
keys during preclinical studies, but it caused typical cytokine storms
in humans.3 Therefore, the ar tificial breeding of mice that fully exhibit
human immune responses for biomedical research is difficult, and if mice
are to be used for biomedical research, the experiment al errors caused
by interspecies differences must be overcome. Decades ago, researchers
attempted to overcome these differences by introducing human genes
into mice through transgenic methods or knocking out genes closely re-
lated to immune responses in mice; however, it has been shown that the
knockout or introduction of single genes cannot completely eliminate
these effects.4–6 Xenotransplantation is an effective method to address
the experimental errors caused by interspecies differences.7 By selecting
appropriate human immune tissues or cells and transplanting them into
suitable mice, human immune tissues can be engrafted and functionally
rebuilt in mice, thus overcoming the differences in immune responses be-
tween humans and mice.8 Researchers have made unremitting efforts
over the las t few decades, going through multiple stages of development
and actively exploring the biological charac teristics of donor grafts and
host animals. They have successfully transplanted human peripheral
blood mononuclear cells (PBMCs) and hematopoietic stem cells (HSCs)
into different strains of immunodeficient mice, reconstructing the human
immune system in mice. Starting from the initial reconstruction of the
human lymphoid immune cells in the microenvironment of mice, it is now
possible to reconstruct both lymphoid and myeloid immune cells, solving
the current challenges in biomedical research and providing an ideal ani-
mal model for immunological research.
2 | IMMUNE SYSTEM DIFFERENCES
BETWEEN HUMANS AND MICE
Natural selection and artificial breeding are the mechanisms behind
the differences in the immune systems of humans and mice. The last
common ancestor of humans and mice lived approximately 65–75
million years ago.9 Comparative genomics studies have found that
humans and mice share 8 0% of their genes, but only 40% of these
genes are complete matches, with approximately 30 0 genes, includ-
ing 169 immune- related genes, showing species specificity and dif-
ferences between the two species.10,11
Humans and mice are mammals, and although there are no
significant anatomical differences in the overall structure of their
immune organs, there are differences in immune cell composi-
tion, immune cell distribution, and immune molecule expression.2
There is a significant dif ference in the percentage of lymphocy tes
and neutrophils in human and mouse peripheral blood. Human
peripheral blood has a higher percentage of neutrophils and a
lower percentage of lymphocytes, while the opposite is the case
in mice.12 The immunog lo bulin ty pe s in human and mouse sera a re
also different. In mice, IgG is divided into four subclasses, namely,
IgG1, IgG2a, IgG2b, and IgG3, while in humans, IgG is divided into
different subclasses, namely, IgG1, IgG2, IgG3, and IgG4. This
difference leads to different binding abilities between different
subclasses of IgG and Fc receptors.13 Although the anatomical
structure of the spleen is similar in humans and mice, the distri-
bution of T cells in the white pulp of the spleen dif fers. T cells
in mice are clustered, while in humans, they are scattered in the
germinal centers.14 There are also differences in the composition
of immune cells in the epidermis of the skin between humans and
mice. Dendritic epidermal γδ T cells (DETCs) are only found in the
epidermis of mice, while humans have αβ T ce ll s but no DET Cs.15 ,16
The interspecies differences between humans and mice also de-
termine differences in immune molecule expression and func-
tion. Human neutrophils can express defense molecules with
antimicrobial effects, while mouse neutrophils do not possess this
function. In human neutrophils, FcαRI can mediate immune kill-
ing after binding with antibodies, but mouse neutrophils do not
express FcαRI; instead, they use Fcα/μR to bind with IgM.17 Ly49
is an inhibitory receptor for NK and NKT cells in mice, while the
inh ibito ry re cepto r fo r huma n NK cells is KIR , no t Ly49.18 Similarly,
there are differences in ligand binding between human and mouse
NK cells. Human NKG2D can bind to its ligands MHC- I and UL16,
while mouse NKG2D binds to H- 60 and Rae1β to exert biologi-
cal effects.19, 20 These interspecies differences in immune- related
genes also result in different functions. Mutation of the X- linked
interleukin 2 receptor subunit gamma chain gene (IL2rg , CD132 or
γc) affec ts the development of human T cells and NK cells, but not
B cells; however, mutation of the IL 2rg gene severely decreases
the number of B cells in mice.21 Similarly, mutation of the IL7R
gene inhibits the development of human T cells, but in mice, it
affects both T and B cell development.22, 23 Interspecies differ-
ences between humans and mice also result in different immune
response processes. In humans, activation of Th2 cells results in
the recruitment of eosinophils and the production of IgE by B
cells, which is the main defense mechanism against schistosomia-
sis; however, mi ce use Th1 ce ll- produced IFN- γ to combat parasite
infection.24 ,25 IFN- αβ or IL- 4 combined with anti- CD23 antibodies
   
|
 3
BIN et al.
can induce human macrophages to produce inducible nitric oxide
synthase (iNOS) to resist pathogenic microbial infections, while
mouse macrophages can only produce significant levels of iNOS
when induced by LPS and IFN- γ.26, 27
The currently available laboratory mice are inbred or transgenic
mice that have been artificially bred for a long time and are main-
tained in an SPF environment. They have lost genetic diversity and
are sensitive to the external environment; moreover, the immune
differences caused by interspecies differences further limit the
range of application of these mice. However, the reconstruction of
the human immune system in mice through xenotransplantation can
effectively solve the problems encountered in mouse experiments.
3 | CHOICE OF RECIPIENT HOST ANIMAL
AND DONOR GRAFT
Reconstructing the human immune system in mice through
xenotransplantation involves both a donor graft and a recipient
host animal. The donor graft from humans should include cells
or tissues that are immunologically relevant, while the recipient
host animal should be able to accept the donor graft and allow
it to proliferate or dif ferentiate in the body. The discovery and
application of immunodeficient mice are important milestones in
the process of human immune reconstruction. Donor grafts can
survive in immunocompromised mice due to their immunodefi-
ciency. The development of immunodeficient mice has progressed
from t he sim pl est t ype s, such as nu de mice (w it h T cell de fi ci en cy),
to the currently used severe combined immunodeficient (SCID)
mice, which have severe combined deficiencies in T, B, and NK cell
functions. This development has gone through different stages
(Figure 1). SCID mice now serve as a suitable host for human im-
mune transplantation.
3.1  |prkdc−/− mice
The discovery of hairless mice with a single T cell deficiency due
to a Fo xn1 mutation paved the way for studying immunodeficient
mice and provided favorable conditions for human xenotransplan-
tation models.28,29 The DNA- dependent protein kinase (DNA- PK),
encoded by the Prkdc gene, is a critical component of the non-
homologous end joining pathway for repairing double- strand
breaks in DNA during V(D)J recombination. In addition to DNA- PK,
Artemis, Rag1/2, XRCC4 and Ligase IV are also important regulatory
genes in the V(D)J recombination process. The complexes consisting
of DNA- PK / Artemis, Rag1/2, and Ligase IV/XRCC4 complete the
repair of double- strand gaps during V(D)J recombination, generat-
ing diverse T and B cell receptors that mediate adaptive immune
responses.30–32 Mutations in any of these genes in the complex
result in abnormal V(D)J recombination, leading to impaired T and
B cell development; this is also known as SCID.33 Introducing the
mutated Prkdc gene in C.B- 17, C3H, and Beige mice can generate
typical SCID animals.3 4–36 SCID mice exhibit more severe immune
deficiencies than nude mice, making them a preferred model for im-
mune reconstitution studies. Although SCID mice have combined
T and B cell deficiencies, their NK cells, macrophages, and comple-
ment sys tem function normally. Non- ob ese dia betic (NOD) mice, on
the other hand, have lower NK cell function, macrophage defects,
and complement deficiency.37 NOD- SCID mice were generated
FIGURE 1 Diagram of immunodeficient
mouse development.
4 
|
    BIN et a l.
by crossing NOD mice with C.B- 17/SCID mice, which retained the
characteristics of both strains with combined T and B cell defi-
ciencies, as well as low NK cell function, macrophage defects, and
complement deficiency38 (Figure 1). Human immune grafts develop
and proliferate better in NOD- SCID mice compared to SCID mice.
The introduction of mutations in key genes involved in the repair of
double- strand gaps in V(D)J recombination, such as Prkdc, Rag1/2,
Dcl re1c, and Ligase IV, using embryonic stem cell targeting tech-
niques, can also result in severe combined immunodeficiency SCID
in mice with impaired T and B cell function.39– 42
3.2  |IL2rg−/− mice
IL2rg encodes a glycoprotein mainly expressed on the surface of
me mor y T ce lls and NK ce lls , se r vin g as a co m m o n rece pto r su b unit
for various important immune factors such as IL- 2, IL- 4, IL- 7, IL- 9,
IL- 15, and IL- 21. Janus kinase 3 (JAK3) is one of the crucial kinases
th at me di ate IL- 2 re cep to r sig nalin g, the int era c ti on bet wee n IL2 rg
and JAK3 regulates the activit y of immune cells, either a mutation
in the IL2rg or JAK3 gene can cause immune deficiency, character-
ized by NK ce ll deficie ncy, downregula ti on of cyt ok in e ex pressi on ,
and inhibition of immune cell proliferation and differentiation.43–46
IL2rg and JAK3 mutation s were introd uced into NOD - S CID mice in
order to fur ther enh an ce immune def ic ie ncy in the mice (Figure 1).
The resulting NSG (NOD/LtSz- Prkdcscid Il2rgtm1W jl/J), NOG (NOD/
Shi- Prkdcscid Il2rgtm1 Sug /Jic), and NOD/SCID- JAK3null mice not
onl y retain ed the T an d B ce ll def ic ienc y, ma crophag e de fe ct s, and
complement deficiency of NOD- SCID mice, but also exhibited NK
cell deficiency and downregulated expression of cytokines IL- 2,
IL- 4, IL- 7, IL- 9, IL- 15, and IL- 21 due to the IL2rg mutation.47, 4 8 These
mice are more immunodeficient than NOD- SCID mice, Rag1 or
Rag2 mutant mice, thus making them more suitable for human im-
mune system engraftment. Using gene editing technologies such
as ZFN, TALEN, and CRISPR/Cas9, severe combined immunodefi-
cient mice with T, B, and NK cell defects, like NCG and BRG mice,
can be generated by mutating Prkdc, Rag1/2, Dclre1c, and IL2rg.49, 50
Many different strains of SCID mice are currently used for human
immune reconstitution, every strain has different biological char-
acteristics, and the efficiency in engraf ting human immune cells
varies among str ains. In ge neral, NOD- SCID mice with T an d B ce ll
defect s have lower reconstitution efficienc y than NSG, NOG, and
BRG mice wit h combine d T, B, an d NK ce ll deficien ci es , and the re -
constitution effects in BRG mice are less pronounced than in NSG
and NOG mice51, 52; therefore, NSG and NOG mice are currently
the most widely used animal models for human immune reconsti-
tution and immune system transplantation research.
3.3  | Donor grafts
The thymus, bone marrow, lymph nodes, spleen, and mucosal tissues
are the immune organs, where immune cells develop, differentiate,
mature, and reside. Immune cells that have matured in the immune
organs circulate through the bloodstream and play a part in the im-
mune response; therefore, immune organs and peripheral blood have
become the main choice for transplantation in the process of human
immune reconstitution. In the early stages of human immune recon-
stitution, fetal thymus, bone marrow, lymph nodes, and other tissues
were often transplanted,53, 54 but obtaining immune organs was dif-
ficult and the transplantation technique was demanding. Later, adult
peripheral blood became a common choice for transplantation. PBMCs
are a mixed population of mononuclear cells with a single nucleus found
in peripheral blood, which includes T lymphocytes, B lymphocytes, NK
cells, monocytes, phagocytes, and dendritic cells that are produced in
the thymus, bone marrow, spleen, and other immune organs.55 The im-
mune cells in PBMCs are mostly mature cells, and following transplan-
tation, some of them can directly proliferate in the immunodeficient
mice, thus par tially reconstructing the human immune system.
CD34+ HSCs obt ained from umbilical cord blood, the embryonic
liver, mobilized peripheral blood, or bone marrow are precursors for
all immune cells and possess the ability to self- renew and differ-
entiate into immune cells.56 Following transplantation, pluripotent
CD34+ cells show different differentiation abilities, depending on
the host animal, and can differentiate into different types of human
immune cells for immune reconstitution.
4 | HUMAN IMMUNE RECONSTITUTION
MICE
Humanized mice for immune reconstitution can be classified into
three categories, namely, hu- PBL, hu- HSC, and hu- BLT, based on the
source of the graft and the host animal (Figure 2).
4.1  | hu- PBL immune reconstitution mice
Human PBMCs or human peripheral blood lymphocytes (hu- PBLs)
are easy to obtain and manipulate. They have already differen-
tiated and matured in the human body, so they can be directly
transplanted into immunodeficient mice for rapid reconstruction
of human immune cells57 (Figure 2). This is a relatively simple and
economical model, providing humanized mice with a human im-
mune system. In the early stages, the PBMCs or lymphocytes were
often transplanted into SCID mice, but with the discovery and ap-
plication of NSG or NOG mice, which have higher degrees of im-
munodeficiency, these two types of mice are now commonly used
for hu- PBL reconstitution. The presence of human CD45+ T cells
can be detected as early as 2 weeks after hu- PBL transplantation.
After 3 weeks, human CD45+CD3+ cells, including a large num-
ber of CD 4+ and CD8
+ T cell subsets, proliferate rapidly. Further
analysis revealed an increase in the number of CD45RO+CD27 ef-
fector T cells in the CD4+ and CD8+ T cell subsets, while the ra-
tios of initial CD45ROCD27+ T cells/CD45RO+CD27+ memory
T cells and central CD45RO+CD27+CD62L+ memory T cells/
   
|
 5
BIN et al.
CD45RO+CD27+CD62L effector memory T cells were both de-
creased. This indicates that in hu- PBL- SCID mice, the human T cell
subset mainly comprises central memory T cells, while other T cell
subsets such as FOXP3+CD25+CD127low regulatory T cells are not
found to proliferate.58–60 Human T cell subsets often colonize and
proliferate in the peripheral blood, thymus, spleen, liver, and lymph
nodes of mice, and the presence of different human T cell subsets
can be detected in these tissues.60 ,61 In hu- PBL mice, only the pro-
liferation of different types of human T cells can be detected, while
CD19+ B cells can only be maintained at very low levels for a short
time in the bon e mar row and spleen of the se mice.62 Hu man NK ce lls
and other myeloid immune cells, such as macrophages, dendritic
cells, and granulocytes, cannot proliferate in the body of hu- PBL
mice.63 Due to the differences in human and mouse MHC antigens,
the transplantation of human PBMCs often causes graft- versus- host
disease (GVHD) in hu- PBL mice in the short term, leading to weight
loss, a hunching posture, hair loss, and decreased activity; GVHD can
even result in death, thus shortening the experimental window.64–66
Th er efo re , hu- PB L mic e ar e onl y use d to obse r ve T cel l- m ediated im-
mune responses and conduct short- term experimental studies.
4.2  | hu- HSC immune reconstitution mice
CD34+ HSCs have the ability to differentiate into multiple lin-
eages, and transplantation into immunocompromised mice with
the help of the animal's microenvironment has become an impor-
tant method for human immune reconstruction (Figure 2). HSCs
us ed f or tr a n s p lantati on a re ty p i c a l l y so u r c e d fr o m fe t a l li v e r, um -
bilical cord blood, bone marrow, or G- CSF- mobilized peripheral
blood.67,68 Notably, the source of HSCs can influence the func-
ti on of hum an T cell s that de vel o p in th e tr ans p l ante d mi c e . T ce lls
derived from CD34+ HSCs from fetal sources have higher immu-
notolerance compared to those from adult sources.69 Following
transpla nt at io n into NS G or NO G mice, HSC s c an pa rtially recon -
stitute the human immune system within 10–12 weeks. Human
innate immune response cells, adaptive immune response cells,
and a small number of red blood cells and platelets can be de-
tected in the mice.7 0–7 2 Unlike PB MC t ransplantation, w hich only
allows the reconstitution of human T cells, in hu- HSC mice, vari-
ous types of human immune cells can be detected. In addition,
HSCs from CD34+ HSC transplants can differentiate into dif-
ferent lineages of immune cells in SCID mice, leading to a lower
rejection response from the mouse immune system and delayed
development of GVHD71; however, hu- HSC mice are not an ideal
animal model for immune reconstruction, as human myeloid cells,
such as m ac rop ha ge s, dendr it ic ce lls, and NK cel ls do not deve lop
fully in the mice. This results in decreased production of antigen-
specific IgGs and reduced HLA restriction. In light of these char-
acteristics, hu- HSC mice are suitable for conducting experiments
that require long research periods and observation of various im-
mune responses.
FIGURE 2 Diagram of human immune reconstitution mouse classification.
6 
|
    BIN et a l.
4.3  | hu- BLT immune reconstitution mice
T cell development depends on the thymic microenvironment, but
immunodeficient mice have severe thymus degeneration, which is
one of the reasons for the low efficiency of immune reconstruction.
Transplantation of fetal thymic and liver tissues into immunodefi-
cient mice under the renal capsule creates an artificial thymic mi-
croenvironment, which overcomes the problem of impaired T cell
development due to thymus degeneration in mice73 (Figure 2). Based
on this principle, researchers have created BLT mice to improve im-
mune reconstruction efficiency. BLT mice are created by simultane-
ous transplantation of fetal thymic and liver tissues into NSG or NOG
mice under the renal capsule, followed by injection of CD34+ HSCs
to reconstitute the human immune system. The humanized thymus
in BLT mice provides an ideal environment for T cell development,
education, and selection, resulting in HLA- restricted T cells. Further,
this mouse model can reconstitute human myeloid cells, including
monocytes, granulocytes, and the mucosal immune system.74 ,75
Transplanted human tissues in recipient mice establish a
complete human hematopoietic microenvironment, enhancing
the multi- lineage reconstitution of human hematopoietic cells.
Notably, a functional and complete human immune system, includ-
ing T cells, B cells, and dendritic cells, is developed. These mice
also produce high levels of human IgM and IgG antibodies. The
development of T cells relies on selection in the human thymus.
BLT mice, which contain diverse HLA- restricted T cells, can estab-
lish effective adaptive immune responses; therefore, BLT mice are
often used in re se ar ch rel ated to ad ap ti ve imm un e re sp on se s, such
as HIV infection.76 The use of BLT mouse models is limited by the
availability of human embryonic tissues, and due to the positive
sel ection in the thy mu s be in g sp ec if ic to huma n T cells, T cells with
mouse MHC specificity cannot be eliminated. As a result, hu- BLT-
SCID mic e are mor e pro ne to devel op ing GVH D t ha n other human-
ized mouse models.77
5 | OPTIMIZATION OF HUMAN IMMUNE
RECONSTITUTION EFFECT
5.1  | Improvements in immunodeficient mice
5.1.1 |  Promoting multidirectional
differentiation of HSCs
HSCs are a type of progenitor cell that can self- renew, differenti-
ate, and regenerate into various types of blood cells. The regula-
tion, proliferation, and dif ferentiation of HSCs involve a series of
hematopoietic growth factors. Positive regulators such as stem cell
factor (SCF), FLT3, colony- stimulating factors (CSFs), IL- 2, IL- 3, IL-
6, IL- 15, and thrombopoietin (TPO) promote HSC differentiation,
while negative regulators such as TGF- β, TNF- α, β, and chemokines
inhibit HSC differentiation.78–80 Positive regulators bind to recep-
tors on HSCs, activating intracellular signaling pathways such as the
ERK, JNK, and TKR pathways, inducing the division and proliferation
of hematopoietic progenitor cells, and promoting the transition of
HSCs from the G0 phase to the G1 phase, accelerating HSC prolif-
eration and differentiation.81,82 Negative regulators such as TGF- β
induce HSCs to enter the G0 phase, inhibiting HSC proliferation and
differentiation.83 Under the influence of positive regulators, HSCs
can differentiate into various blood cell lineages, including myeloid
cells (monocytes, macrophages, granulocytes, red blood cells, mega-
karyocytes/platelets, and dendritic cells) and lymphoid cells (T, B,
and NK cells).
As mentioned above, following the transplantation of human
HSCs into immunodeficient mice, the mice lack the microenviron-
ment necessary for the proliferation and dif ferentiation of human
HSCs. Consequently, HSCs are unable to differentiate into lymphoid
NK cells and myeloid cells such as granulocytes and macrophages;
however, researchers have used genetic engineering methods to
introduce positive regulatory factors that regulate HSC differenti-
ation into the mice (Figure 1), thereby improving the developmental
microenvironment for human HSCs and enabling their differentia-
tion into various lineages of immune cells. IL- 2 is an important cy-
tokine that induces the proliferation of NK cells; it activates various
kinase pathways, increases NK cell activit y, and promotes NK cell
proliferation. IL- 15 upregulates the expression of sur face receptors
on NK cells and also promotes the expansion of CD56+ cells.84 By
expressing human IL- 2 and IL- 15 in NOG or NSG mice, researchers
have overcome the limitations of HSC differentiation into NK cells
observed in prior studies. The differentiation and proliferation of
CD56+ NK cells in the peripheral blood and spleen of these mice
are more than 10 times higher than that in NOG mice. Further, these
NK cells express various receptors, such as NKp30, NKp44, NKp46,
NKG2D, and CD94, and produce cytotoxic factors such as perforin
and granzyme upon stimulation.84–86
Granulocyte- macrophage colony- stimulating factor (GM- CSF),
SCF, IL- 3, and TPO are cytokines that are essential for the differen-
tiation of CD34+ HSCs into granulocytes. MISTRG, NSG- SGM3, or
NOG- EXL mice expressing these cy tokines can generate not only
lymphoid T and B cells but also myeloid cells such as granulocytes,
mac ro ph ages, and dendrit ic cells (Figure 1). In sum, the development
of human immune reconstitution mouse models has undergone a
complex process, from the simple transplantation of PBMCs to the
development of CD34+ HSC- based models capable of reconstituting
both innate and adaptive immune cells.87– 89 These models involved
continuous modifications to provide the necessary microenviron-
ment for HSC differentiation. Ultimately, the transplanted CD34+
HSCs can reconstitute multi- lineage immune cells, providing valu-
able tools for immunological research.
HLA plays a pivotal part in the education of T progenitor cells.
The low efficiency of human T cell reconstitution in immunodefi-
cie nt mice is believed to be rel ated to the lack of human HL A ex pres-
sion in the se mice90; however, by expressing HLA- DR4, HLA- A2, and
other HLA molecules in immunodeficient mice, transplanted human
HSCs can differentiate into CD4+ helper T cells, antigen- specific
CD8+ T cells, and functional B cells.91–9 4
   
|
 7
BIN et al.
5.1.2  |  Increasing the degree of immunodeficiency
Currently, commonly used human immune- reconstituted mice are
con st ructed based on mice wit h co mb in ed T, B, and NK cell def icie n-
cies; however, the deficiency of T, B, and NK cells alone is not suf-
ficient to ensure the sur vival of human grafts in mice, as other types
of immune cells or immune molecules are still key factors influenc-
ing immune reconstitution. FLT3L, a hematopoietic growth factor,
can promote the development of hematopoietic precursor cells into
dendritic cells and enhance the survival and proliferation of den-
dritic cells. However, normal FLT3L expression in the bone marrow
of immunodeficient mice can affect the effectiveness of immune
reconstitution. Immunodeficient mice with the Flk2/Flt3 molecule
knocked out, also known as BRGF mice, not only have impaired de-
velo p me nt of thei r own dend rit ic ce lls but al so pr om ote the re con sti -
tution of human dendritic cells following transplantation of CD34+
HSCs, thereby improving the efficiency of human T cell and NK cell
reconstitution.95 The binding of the macrophage- expressed Sirpα
receptor with its ligand CD47 mediates the generation of inhibi-
tory ‘don't eat me’ signals, which inhibits the phagocytic activity of
macrophages against target cells.96,97 The Sirpα in NOD background
mice exhibits structural similarity to the human form and is able to
bind human CD47. As a result, introducing NOD mouse Sirpα into
BRGF mice can reduce the phagocytic activit y of macrophages and
enhance transplantation efficiency (Figure 1). CD34+ HSCs trans-
planted into these mice can differentiate into functional immune
cells without developing GVHD.98 –10 0
5.1.3  |  Reducing the occurrence of GVHD
Alt ho ugh GVHD af te r CD34+ HSC transplantation appears later and
with milder reactions compared to PBMC transplantation, GVHD
resulting from PBMC transplantation is the most common issue in
immune reconstitution, severely limiting the application of such
models. Dif ferent factors contribute to the occurrence of GVHD,
including animal pre- treatment methods (such as irradiation), the
PBMC inoculation dose, and host immune responses. The Prkdc is
a radiosensitive gene, and immunodeficient mice based on Prkdc
gene mutations are commonly irradiated with low doses of γ- r a y s
to reduce GVHD and improve the engraftment rate of human im-
mune cells.101 However, loss of c- kit function impairs endogenous
mouse hematopoietic stem cells, allowing engraftment of human
HSC without irradiation. Thus, NSG or NOG mice with a c- kit muta-
tion (NSGW41 mice) can achieve a higher reconstitution efficiency
of human immune cells after HSC transplantation free from the need
of γ- ray irradiation.102 In PBMC transplantation, the occurrence of
GVHD resulting from the recipient mouse's MHC antigen recogni-
tion of donor T cells leads to a significantly shorter experimental
window, thus impacting experimental results. Immunodeficient mice
with mutations in MHC- I and/or MHC- II subunits (β2- microglobulin)
and H2- Ab1 show a significant reduction in GVHD occurrence, thus
extending the experimental window59,103,104 (Figure 1). Further, in
TKO mice with CD47 knockout, the onset of GVHD after human
PBMC transplantation is delayed.98
5.2  | Improvement of the microenvironment for
HSC development
5.2.1  |  Lymph nodes
Lymph nodes are the primary sites for the residence of mature T
and B cells; however, immunodeficient mice with IL2rg knockout
have underdeveloped lymph nodes, which affects the development
of transplanted human T and B cells. By specifically re- expressing
the IL2rg gene or thymic stromal cell- derived lymphopoietin (TSLP)
in lymphoid tissues of immunodeficient mice, the development of
lymph nodes can be promoted, providing suitable developmental
niches for immune cells. As a result, human T cells can reside in the
lymph nodes of the mice following immune reconstitution, with a
predominance of residence in the intestinal lymph nodes. In addi-
tion, they can generate antigen- specific IgG antibodies.105,10 6
5.2.2  |  Hematopoietic niche
The bone marrow microenvironment is composed of mesenchymal
stem cells (MSCs) and differentiated cells such as osteoblasts, os-
teoclasts, vascular endothelial cells, stromal cells, and hematopoi-
etic cells.107 MSCs play a pivotal part in providing essential suppor t
for self- renewal and differentiation of HSCs. MSCs can direc tly
interact with HSCs through adhesion molecules on their cell mem-
brane or regulate HSC differentiation and maturation through the
secretion of various cy tokines such as IL- 6, IL- 11, LIF, G- CSF, SCF,
a n d G M - C S F . 108,109 In vitro co- culture of MSCs and HSCs effec-
tively enhances HSC proliferation, while in vivo co- transplantation
enhances HSC engraftment and differentiation, with a higher en-
graftment of CD13+ myeloid lineage cells and earlier appearance
of CD19+ B cells.110–112 MSCs overexpressing PDGFB have a more
pronounced regulator y effect on HSCs, promoting HSC engraftment
and maintaining their self- renewal capacity, as compared to wild-
type MSCs.113,114 In addition, co- infusion of MSCs and allogeneic
HSCs can prevent and alleviate GVHD, facilitate HSC engraftment,
and accelerate hematopoietic reconstitution.114,11 5 The antioxidant
N- acetyl- L- cysteine (NAC) can reduce ROS levels in the bone mar-
row of NOD- SCID mice, improve the bone marrow hematopoietic
microenvironment, and enhance HSC engraftment efficiency. NAC-
treated NOD- SCID mice exhibit a 10.8- fold higher efficiency in im-
mune reconstitution compared to untreated mice.116
5.2.3  |  Organoids
Although thymus and bone marrow transplantation provide good
strategies for immune reconstitution in humans, ethical issues limit
8 
|
    BIN et a l.
the widespread application of bone marrow–liver–thymus (BLT)
mice. Researchers have developed thymic organoid as a substitute
for the human thymus in order to address this. In vitro cultured in-
duced pluripotent stem cells (iPSC) can be directed to differentiate
into functional thymic epithelial progenitor cells, supporting de novo
construction of a T cell compartment in humanized mice engrafted
with iPSC- derived thymus organoids after CD34+ HSCs implanta-
tion.117–119 The transplantation of a human bone marrow MSC-
derived ossicle- like organoid under the skin of immunodeficient
mice can establish a humanized bone marrow microenvironment.
The humanized bone marrow microenvironment provides a suitable
homing and engraftment site for human HSCs compared to mouse
bone marrow, thus promoting their differentiation and proliferation
and significantly improving the engraftment efficiency of HSCs.120
6 | PROSPECT S
After decades of development, researchers have successfully recon-
st itu t e d the hu m a n imm une sy s tem in im m unod efi c i ent mi c e by tr ans -
planting PBMCs or HSCs. These humanized immune- reconstituted
mice overcome interspecies differences and provide important tools
for immunolog y- related research; however, it is undeniable that cur-
rent humanized immune- reconstituted mice still have limitations,
such as a single- lineage immune cell repertoire, a high risk of GVHD
after PBMC transplantation, and a long HSC differentiation period.
Regarding the host animal, SCID mice are currently the most suitable
host animal. Developing genetically engineered mice that differ from
the commonly used SCID mice may be an important approach to im-
prove the efficiency of immune reconstitution. Regarding the graft,
MSC co- transplantation with HSCs can effectively enhance the HSC
differentiation capacity, and the use of organoids provides a supe-
rior microenvironment for HSC differentiation. Therefore, MSC co-
transplantation with HSCs or simultaneous transplantation of HSCs
and organoids in immunodeficient mice may be important strategies
for improving immune reconstitution ef ficiency in the future.
AUTHOR CONTRIBUTIONS
Yixiao Bin conceived and wrote the original draft of this manuscript,
Hai Zhang and Zhinan Chen revised it. All authors critically read and
contributed to the manuscript, and approved its final version.
ACKNOWLEDGMENTS
None.
FUNDING INFORMATION
This work was supported by Scientific and Technological Resources
Coordination Project of Shaanxi Province (2020PT- 002, 2022PT-
43, 2023- CX- PT- 18), Special Fund for Military Laborator y Animals
(S YD W_K Y (2021 )13 ) a n d St a t e Ke y La bor a t o r y of Ho l i s t i c In tegr a t i v e
Management of Gastrointestinal Cancers (CBSKL2022ZZ28).
CONFLICT OF INTEREST STATEMENT
Hai Zhang is an editorial board member of AMEM and co- author of
this article. To minimize bias, he was excluded from all editorial deci-
sion making related to the acceptance of this article for publication.
The authors declare that there is no conflict of interest regarding the
publication of this article.
ETHICS STATEMENT
None.
ORCID
Hai Zhang https://orcid.org/0000-0002-7999-0208
REFERENCES
1. Casellas J. Inbred mouse strains and genetic stability : a review.
Animal. 2011;5(1):1-7.
2. Masopust D, Sivula CP, Jameson SC. Of mice, dirt y mice, and
men: using mice to understand human immunolog y. J Immunol.
2017;199(2):383-388 .
3. Bugelski PJ, Achuthanandam R, Capocasale RJ, Treacy G, Bouman-
Thio E. Monoclonal antibody- induced cytokine- release syndrome.
Exper t Rev Clin Immunol. 2009;5(5):499-521.
4. Vitiello A, Marchesini D, Furze J, Sherman LA, Chesnut RW.
Analysis of the HLA- res tricted influenza- spe cific cytotoxic T
lymphocyte response in transgenic mice carrying a chimeric
human- mouse class I major histocompatibility complex. J Exp Med.
1991;173(4):10 07-1015.
5. Ito K , Bian HJ, Molina M, et al. HL A- DR4- IE chimeric class
II transgenic, murine class II- deficient mice are suscepti-
ble to experimental allergic encephalomyelitis. J Exp Med.
1996;183(6):26 35-264 4.
6. Zhai Y, Chen L, Zhao Q, et al. Cysteine c arbox yethylation gener-
ates neoantigens to induce HL A- restricted autoimmunity. Science.
2023;379(6637):eabg2482.
7. Saito Y, Mat sumoto N, Yamanaka S, Yokoo T, Kobayashi E .
Beneficial impact of interspecies chimeric renal organoids against
a xenogeneic immune response. Front Immunol. 2022;13:848433.
8. Thomsen M, Galvani S, Canivet C, Kamar N, Böhler T. Reconstitution
of immunodeficient SCID/beige mice with human cells: applica-
tions in preclinical studies. Toxicology. 2008;246(1):18-23.
9. Emes RD, Goods tadt L , Winter EE, Ponting CP. Comparison of the
genomes of human and mouse lays the foundation of genome zo-
olog y. Hum Mol Genet. 2003;12(7):701-709.
10. Waterston RH, Birney E, Rogers J, et al. Initial sequencing and
comparative analysis of the mouse genome. Nature (London).
2002;420(6915):520-562.
11. Medetgul- Ernar K , Davis MM. Standing on the shoulders of mice.
Immunity. 2022;55(8):1343-1353.
12. Doeing DC, Borowicz JL, Crockett ET. Gender dimorphism in dif-
ferential peripher al blood leukocy te counts in mice using cardiac,
tail, foot, and saphenous vein puncture methods. BMC Clin Pathol.
2003;3 (1):3.
13. Martin RM, Lew AM. Is IgG2a a good Th1 marker in mice? Immunol
Tod ay. 1998;19(1):49.
14. Lewis SM, Williams A, Eisenbarth SC. Structure and function of the
immune system in the spleen. Sci Immunol. 2019;4(33):eaau6085.
15. Johnson MD, Witherden DA, Havran WL . The role of tissue-
resident T cells in stress surveillance and tissue maintenance. Cells.
2020;9(3):686.
16. Elbe A, Foster CA, Stingl G. T- cell receptor alpha beta and gamma
delta T cells in rat and human skin–are they equivalent? Semin
Immunol. 1996;8(6):341-349.
   
|
 9
BIN et al.
17. Monteiro RC, Van De Winkel JG. IgA Fc receptor s. Annu Rev
Immunol. 2003;21:177-204.
18. Lanier LL. NK cell receptors. Annu Rev Immunol. 1998;16:359-393.
19. Lodoen M, Ogasawara K, Hamerman JA, et al. NKG2D- mediated
natural killer cell protection against cytomegalovirus is impaired
by viral gp4 0 modulation of retinoic acid early inducible 1 gene
molecules. J Exp Med. 20 03;197(10):1245-1253.
20. Kegasawa T, Tatsumi T, Yoshioka T, et al. Soluble UL16- binding
protein 2 is associated with a poor prognosis in pancreatic c ancer
patients. Biochem Biophys Res Commun. 2019;517(1):84-88 .
21. Fischer A, Cavazzana- Calvo M, De Saint BG, et al. Naturally oc-
curring primary deficiencies of the immune s ystem. Annu Rev
Immunol. 1997;15:93-124.
22. Peschon JJ, Morrissey PJ, Grabs tein KH, et al. Early lymphocy te
expansion is severely impaired in interleukin 7 receptor- deficient
mice. J Exp Med. 19 94;180 (5):1955 -1960.
23. Roifman CM, Zhang J, Chitayat D, Shar fe N. A partial deficiency
of interleukin- 7R alpha is sufficient to abrogate T- cell devel-
opment and cause severe combined immunodeficiency. Blood.
2000;96(8):2803-2807.
24. Hagan P. IgE and protective immunity to helminth infections.
Parasite Immunol. 1993;15(1):1-4.
25. Pearce EJ, Sher A. Functional dichotomy in the CD4+ T cell re-
sponse to Schistosoma mansoni. Exp Parasitol. 1991;73(1):110 -116.
26. Bogdan C. Nitric oxide and the immune response. Nat Immunol.
2001;2(10) :907-916.
27. Weinberg JB. Nitric oxide production and nitric oxide synthase
type 2 expression by human mononuclear phagocytes: a review.
Mol Med. 1998;4(9):557-591.
28. Flanagan SP. ‘Nude’, a new hairless gene with pleiotropic effect s in
the mouse. Genet Res. 1966;8(3):295-309.
29. Pantelouris EM. Absence of thymus in a mouse mutant. Nature.
1968;217(5126):370-371.
30. Bassing CH, Swat W, Alt FW. The mechanism and regulation of
chromosomal V(D)J recombination. Cell. 2002;109(Suppl):S45-S55.
31. Chen X, Xu X, Chen Y, et al. Structure of an activated DNA- PK and
its implications for NHE J. Mol Cell. 2021;81(4):801-810.
32. Niewolik D, Schwarz K. Physical ARTEMIS:DNA- PKcs inter-
action is necessary for V(D)J recombination. Nucleic Acids Res.
2022;50(4):2096-2110.
33. de Villartay JP. Congenital defects in V(D)J recombination. Br Med
Bull. 2015;114(1):157-167.
34. Bosma G C, Custer RP, Bosma MJ. A severe combined immunode-
ficiency mutation in the mouse. Nature. 1983;301(59 00 ):527-530.
35. Mcelwee KJ, Boggess D, King LJ, Sundberg JP. Experiment al in-
duction of alopecia areata- like hair loss in C3H/HeJ mice using
full- thickness skin gr afts. J Invest Dermatol. 1998;111(5 ):797-80 3.
36. Kenney LL, S hultz LD, Greiner DL , Brehm MA. Humanized
mouse models for transplant immunology. Am J Transplant.
2016;16( 2):389-397.
37. Kataoka S, Satoh J, Fujiya H, et al. Immunologic aspects of the non-
obese diabetic (NOD) mouse. Abnormalities of cellular immunity.
Diabetes. 1983;32(3):247-253.
38. Prochazka M, Gask ins HR, Shultz LD, Leiter EH. The nonobese
diabetic scid mouse: model for spontaneous thymomagene-
sis associated with immunodeficiency. Proc Natl Acad Sci USA.
1992;89(8):3290-3294.
39. Pannicke U, Ma Y, Hopfner KP, Niewolik D, Lieb er MR, Schwar z
K. Functional and biochemical dissection of the structure- specific
nuclease ARTEMIS. EMBO J. 2 004;23 (9):1987-1997.
40. Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S,
Papaioannou VE. RAG- 1- deficient mice have no mature B and T
lymphocytes. Cell. 1992;68(5):869-877.
41. Shinkai Y, Rathbun G, Lam KP, et al. RAG- 2- deficient mice lack
mature lymphocytes owing to inability to initiate V(D)J rearrange-
ment. Cell. 1992;68(5):855-867.
42. Goldman JP, Blundell MP, Lopes L, Kinnon C, di Santo JP, Thrasher
AJ. Enhanced human cell engr aftment in mice deficient in RAG2
and the common cytokine receptor gamma chain. Br J Haematol.
1998;103(2):335-3 42.
43. O'Shea JJ, Pesu M, Borie DC, Changelian P S. A new modality for
immunosuppression: targeting the JAK/STAT pathway. Nat Rev
Drug Discov. 200 4;3(7):555-564.
44. Giliani S, Mella P, Savoldi G, Mazzolari E. Cy tokine- mediated
signalling and early defects in lymphoid development . Curr O pin
Allergy Clin Immunol. 2005;5(6):519-524.
45. Liao W, Lin JX, Leonard WJ. IL- 2 family cytokines: new insights
into the complex roles of IL- 2 as a broad regulator of T helper cell
differentiation. Curr Opin Immunol. 2011;23(5):598-604.
46. Goldberg L, Simon A J, Lev A , et al. Atypical immune phenotype in
severe combined immunodeficiency patients with novel mutations
in IL2RG and JAK3. Genes Immun. 2020;21(5):326-334.
47. Shultz LD, Lyons BL, Burzenski LM, et al. Human lymphoid and
myeloid cell development in NOD/LtSz- scid IL2R gamma null
mice engrafted with mobilized human hemopoietic stem cells. J
Immunol. 2005;174(10):647 7-6489.
48. Koyanagi Y, Tanaka Y, Kira J, et al. Primary human immunodefi-
ciency virus t ype 1 viremia and centr al ner vous system inva-
sion in a novel hu- PBL- immunodeficient mouse strain. J Virol.
1997;71(3) :2417-2424.
49. Zhao Y, Liu P, Xin Z, et al. Biological charac teristics of severe com-
bined immunodeficient mice produced by CRISPR/Cas9- mediated
Rag2 and IL2rg mutation. Front Genet. 2019;10:401.
50. Bin Y, Wei S, Chen R, et al. Dclre1c- mutation- induced immuno-
compromised mice are a novel model for human xenograft re-
search. Biomol Ther. 2024;14(2):180-194.
51. Brehm MA, Cuthber t A, Yang C, et al. Parameters for establishing
humanized mouse models to study human immunity: analysis of
human hematopoietic stem cell engraftment in three immunode-
ficient strains of mice bearing the IL2rgamma(null) mutation. Clin
Immunol. 2010;135(1):84-98.
52. Ito R, Takahashi T, Katano I, Ito M. Current advances in humanized
mouse models. Cell Mol Immunol. 2012;9(3):208-214.
53. Kyoizumi S, Baum CM, Kaneshima H, McCune J, Yee EJ, Namikawa
R. Implantation and maintenance of functional human bone mar-
row in SCID- hu mice. Blood. 1992;79( 7):1704-1711.
54. Frey JR, Erns t B, Surh CD, Sprent J. Thymus- grafted SCID mice
show transient thymopoiesis and limited deplet io n of V bet a 11+ T
cells. J Exp Med. 1992;175(4):1067-1071.
55. Diaz I. Rules of thumb to obtain, isolate, and preserve porcine
peripheral blood mononuclear cells. Vet Immunol Immunopathol.
2022;251:110461.
56. Hughes MR, Canals HD, Cait J, et al. A stick y wicket : defining mo-
lecular functions for CD34 in hematopoietic cells. Exp Hematol.
2020;86:1-14.
57. Pearson T, Greiner DL, Shultz LD. Creation of “humanized” mice
to study human immunity. Curr Protoc Immunol. 2008;CHAPTER:
Unit–15:21.
58. King MA, Covassin L, Brehm MA, et al. Human peripheral blood
leucocyte non- obese diabetic- severe combined immunodefi-
ciency interleukin- 2 receptor gamma chain gene mouse model
of xenogeneic graft- versus- host- like disease and the role
of host major histocompatibility complex. Clin Exp Immunol.
2009;157(1):104-118.
59. Brehm MA, Kenney LL , Wiles MV, et al. Lack of acute xenogeneic
graft- versus- host disease, but retention of T- cell function follow-
ing engraftment of human peripheral blood mononuclear cells
in NSG mice deficient in MHC class I and II expression. FASEB J.
2019;33(3):3137-3151.
60. Ali N , Flutter B, Sanchez RR , et al. Xenogeneic graft- versus- host-
disease in NOD- scid IL- 2Rgammanull mice display a T- effector
memory phenotype. PLoS One. 2012;7(8):e44219.
10 
|
    BIN et a l.
61. Holguin L , Echavarria L, Burnett JC. Novel humanized periph-
eral blood mononuclear cell mouse model with delayed onset
of graf t- versus- host disease for preclinical HIV research. J Virol.
2022;96(3):e0139421.
62. Yanagawa S, Tahara H, Shirouzu T, et al. Development of a human-
ized mouse model to analyze antibodies specific for human leuko-
cyte antigen (HL A). PLoS One. 2021;16(2):e0236614.
63. Shultz LD, Brehm MA, Garcia- Martinez JV, Greiner DL. Humanized
mice for immune system investigation: progress, promise and chal-
lenges. Nat Rev Immunol. 2012;12(11):786-798.
64. Tary- Lehmann M, Lehmann PV, Schols D, Roncarolo MG,
Saxon A. A nti- SCID mouse reactivity shapes the human
CD4+ T cell repertoire in hu- PBL- SCID chimeras. J Exp Med.
1994;180(5):1817-1827.
65. Mosier DE . Adoptive transfer of human lymphoid cells to severely
immunodeficient mice: models for normal human immune func-
tion, autoimmunity, lymphomagenesis, and AIDS. Adv Immunol.
1991;50:303-325.
66. Pino S, Brehm MA, Covassin- Barberis L, et al. Development
of novel major histocompatibility complex class I and class II-
deficient NOD- SCID IL2R gamma chain knockout mice for model-
ing human xenogeneic graft- versus- host disease. Methods Mol Biol.
2010; 602 :105-117.
67. Beksac M , Yurdakul P. How to improve cord blood engraftment?
Front Med (Lausanne). 2016;3:7.
68. Fomin ME, Beyer AI, Muench MO. Human fetal liver cultures sup-
port multiple cell lineages that can engraft immunodeficient mice.
Open Biol. 2017;7(12):17010 8-170114.
69. Mold JE, Venkatasubr ahmanyam S, Burt TD, et al. Fetal and adult
hematopoietic stem cells give rise to distinct T cell lineages in hu-
mans. Science. 2010;330 (6011):1695-1699.
70. Cheng H, Zheng Z, Cheng T. New paradigms on hematopoietic
stem cell differentiation. Protein Cell. 2020;11(1):34-44.
71. Kim SS, Kumar P, Ye C, Shankar P. Humanized mice for study-
ing human leukocyte integrins in vivo. Methods Mol Biol.
2012;757:5 09-521.
72. Rongvaux A, Takizawa H, Strowig T, et al. Human hemato- lymphoid
system mice: current use and future potential for medicine. Annu
Rev Immunol. 2013;31:635-674.
73. Lan P, Tonomura N, Shimizu A, Wang S, Yang YG. Reconstitution
of a functional human immune system in immunodeficient mice
through combined human fetal thymus/liver and CD34+ cell
transplantation. Blood. 2006;108 (2):4 87-492.
74. Stoddar t CA, Maidji E, Galkina SA , et al. Superior human leukocyte
reconstitution and susceptibility to vaginal HIV transmission in
humanized NOD- scid IL- 2Rgamma(−/−) (NSG) BLT mice. Virology.
2011;417(1):154-160.
75. Roy CN, Shu S T, Kline C , R igatti L , Smithgall TE, Ambrose Z . Use
of pediatric thymus to humanize mice for HIV- 1 mucosal transmis-
sion. Sci Rep. 2023;13 (1):17067.
76. Garcia- Beltran WF, Claiborne DT, Maldini CR, et al. Innate immune
reconstitution in humanized bone marrow- liver- thymus (HuBLT)
mice governs adaptive cellular immune func tion and responses to
HIV- 1 infection. Front Immunol. 2021;12:667393.
77. Greenblatt MB, Vrbanac V, Tivey T, Tsang K, Tager AM, Aliprantis
AO. Graft versus host disease in the bone marrow, liver and thy-
mus humanized mouse model. PLoS One. 2012;7(9):e44664.
78. Ivanovic Z. Interleukin- 3 and ex vivo maintenance of hemato-
poietic stem cells: facts and controversies. Eur Cytokine Netw.
20 0 4;15 ( 1) : 6 -13 .
79. Drexler HG. Expression of FLT3 receptor and response to FLT3
ligand by leukemic cells. Leukemia. 1996;10(4):588-599.
80. Richter R, Forssmann W, Henschler R . Current developments in
mobilization of hematopoietic stem and progenitor cells and their
interac tion with niches in bone marrow. Transfus Med Hemother.
2017;44(3):151-164.
81. Wang W, Zhang Y, Dettinger P, et al. Cytokine combinations for
human blood stem cell expansion induce cell- t ype- and cytokine-
specific signaling dynamics. Blood. 2021;138 (10) :8 47-857.
82. Rausch O, Marshall CJ. Cooperation of p38 and extracellu-
lar signal- regulated kinase mitogen- activated protein ki-
nase pathways during granulocyte colony- stimulating
factor- induced hemopoietic cell proliferation. J Biol Chem.
1999;274(7):4096-4105.
83. Wang X, Dong F, Zhang S, et al. TGF- beta1 negatively regulates
the number and function of hematopoietic stem cells. Stem Cell
Reports. 2018;11(1):274-287.
84. Pillet AH, Theze J, Rose T. Interleukin (IL)- 2 and IL- 15 have dif-
ferent effect s on human natural killer lymphocytes. Hum Immunol.
2011;72(11):1013-1017.
85. Katano I, Takahashi T, Ito R, et al. Predominant development of ma-
ture and functional human NK cells in a novel human IL- 2- producing
transgenic NOG mouse. J Immunol. 2015;194(7):3513-3525.
86. Aryee KE, Burzenski LM, Yao LC , et al. Enhanced development of
functional human NK cells in NOD- scid- IL2rg(null) mice express-
ing human IL15. FAS EB J. 2022;36(9):e22476.
87. Rongvaux A, Willinger T, Martinek J, et al. Development and func-
tion of human innate immune cells in a humanized mouse model.
Nat Biotechnol. 2014;32(4):364-372.
88. Wunderlich M, Chou FS, Link KA, et al. AML xenograft effi-
ciency is significantly improved in NOD/SCID- IL 2RG mice con-
stitutively expressing human SCF, GM- CSF and IL- 3. Leukemia.
2010 ;24(10):17 85-1788.
89. Ito R, Takahashi T, Katano I, et al. Establishment of a human al-
lergy model using human IL- 3/GM- CSF- transgenic NOG mice. J
Immunol. 2013;191(6):2890-2899.
90. Watanabe Y, Takahashi T, Okajima A, et al. The analysis of the
functions of human B and T cells in humanized NOD/shi- scid/
gammac(null) (NOG) mice (hu- HSC NOG mice). Int Immunol.
2009;21(7):843-858.
91. Majji S, Wijayalath W, Shashikumar S, et al. Differential effect of
HL A class- I ver sus cla ss- I I tr ansge ne s on huma n T and B cel l rec on-
stitution and function in NRG mice. Sci Rep. 2016;6:28093.
92. Majji S, Wijayalath W, Shashikumar S, Brumeanu TD, Casares S.
Humanized DR AGA mice immunized with plasmodium falciparum
sporozoites and chloroquine elicit protective pre- erythrocytic im-
munity. Malar J. 2018;17(1):114.
93. Labar the L, Henriquez S, Lambotte O, et al. Frontline science:
exhaustion and senescence marker profiles on human T cells in
BRGSF- A2 humanized mice resemble those in human samples. J
Leukoc Biol. 2020;107(1):27-42.
94. Billerbeck E, Horwit z JA, Labitt RN, et al. Characterization of
human antiviral adaptive immune responses during hepatotropic
virus infection in HLA- transgenic human immune system mice. J
Immunol. 2013;191(4):1753-1764.
95. Li Y, Mention JJ, Court N, et al. A novel Flt3- deficient HIS mouse
model with selective enhancement of human DC development.
Eur J Immunol. 2016;46(5):1291-1299.
96. Takenaka K , Prasolava TK, Wang JC, et al. Polymorphism in Sirpa
modulates engraftment of human hematopoietic stem cells. Nat
Immunol. 2007;8 (12):1313-1323.
97. Legrand N, Huntington ND, Nagasawa M, et al. Functional CD47/
signal regulatory protein alpha (SIRP(alpha)) interaction is required
for optimal human T- and natural killer- (NK) cell homeostasis
in vivo. Proc Natl Acad Sci USA. 2011;108(32):13224-13229.
98. Lavender KJ, Pang WW, Messer RJ, et al. BLT- humanized
C57BL/6 Rag2−/−gammac−/- CD47−/ mice are resistant to
GVHD and develop B- and T- cell immunity to HIV infection. Blood.
2013;122(25):4013-4020.
99. Lopez- Lastra S, Masse- Ranson G, Fiquet O, et al. A functional DC
cross talk promotes human ILC homeostasis in humanized mice.
Blood Adv. 2017;1(10) :601-614.
   
|
11
BIN et al.
100. Jinnouchi F, Yamauchi T, Yurino A, et al. A human SIRPA knock- in
xenograft mouse model to study human hematopoietic and cancer
stem cells. Blood. 2020;135(19):1661-1672.
101. Cowan MJ, Gennery AR. Radiation- sensitive severe combined im-
munodeficiency: the arguments for and against conditioning be-
fore hematopoietic cell transplantation–what to do? J Allergy Clin
Immunol. 2015;136(5):1178-1185.
102. Cosgun K N, Rahmig S , Mende N, et al. Kit regulates HSC engraft-
ment across the human- mouse species barrier. Cell Stem Cell.
2014;15(2):227-238.
103. Chen SS, Wang H, Xing J, et al. NCG- MHC- dKO mice—an excellent
model for PBMC reconstitution and pharmacodynamic evalua-
tion in the absence of GvHD. J Immunol. 2023;89(1_Supplement):
(210) :2 2-23.
104. Yaguchi T, Kobayashi A, Inozume T, et al. Human PBMC-
transferred murine MHC class I/II- deficient NOG mice enable
long- term evaluation of human immune responses. Cell Mol
Immunol. 2018;15(11):953-962.
105. Takahashi T, Katano I, Ito R, et al. Enhanced antib ody responses in
a novel NOG transge nic mouse with re stored ly mph node or gano-
genesis. Front Immunol. 2017;8:2017.
106. Li Y, Masse- Ranson G, Garcia Z, et al. A human immune system
mouse model with robust lymph node development. Nat Methods.
2018;15(8):623-630.
107. Mendelson A, Frenette PS. Hematopoietic stem cell niche
maintenance during homeostasis and regeneration. Nat Med.
2014;20(8):833-846.
108. Majumdar MK, Thiede MA, Haynesworth SE, Bruder SP, Gerson
SL. Human marrow- derived mesenchymal stem cells (MSCs) ex-
press hematopoietic cytokines and support long- term hematopoi-
esis when differentiated toward stromal and osteogenic lineages.
J Hematother Stem Cell Res. 2000;9(6):841-848.
109. Ran Y, Dong Y, Li Y, et al. Mesenchymal stem cell aggregation me-
diated by integrin alpha4/VCAM- 1 after intrathecal transplanta-
tion in MC AO rats. Stem Cell Res Ther. 2022;13(1):507.
110. Zhang Y, Chai C , Jiang XS, Teoh SH, Leong KW. Co- culture of um-
bilical cord blood CD34+ cells with human mesenchymal stem
cells. Tissue Eng. 2006;12(8):2161-2170.
111. Carrancio S, Romo C, Ramos T, et al. Effects of MSC coadminis tra-
tion and route of delivery on cord blood hematopoietic stem cell
engraftment. Cell Transplant. 2013;22( 7) :1171-1183.
112. Garrigos MM, de Oliveira FA, Nucci MP, et al. How mesenchy-
mal stem cell cotransplantation with hematopoietic s tem cells
can improve engraftment in animal models. World J Stem Cells.
2022;14(8):658-679.
113. Y in X, Hu L, Zhang Y, et al. PDG FB- expressing mesenchy mal stem
ce ll s im p r o ve human he m atopoietic stem cell engraf t m e n t in im m u -
nodeficient mice. Bone Marrow Transplant. 2020;55(6):1029-1040.
114. Hansen M, Stahl L, Heider A, et al. Reduction of graft- versus- host-
disease in NOD.Cg- Prkdc(scid) Il2rg (tm1Wjl)/SzJ (NSG) mice by
Cotransplantation of syngeneic human umbilical cord- derived mes-
enchymal stromal cells. Transplant Cell Ther. 2021;27(8):651-658.
115. Le B lanc K, Rasmusson I, Sundberg B, et al. Treatment of severe
acute graft- versus- host disease with third party haploidentical
mesenchymal stem cells. Lancet. 200 4;363 (9419):14 39-14 41.
116. Hu L, Cheng H , Gao Y, et al. Antioxidant N- acetyl- L- cysteine
increases engraftment of human hematopoietic stem cells in
immune- deficient mice. Blood. 2014;124(20):e45-e48.
117. Zeleniak A, Wiegand C, Liu W, et al. De novo constr uction of T
cell compartment in humanized mice engr afted with iPSC- derived
thymus organoids. Nat Methods. 20 22;19(10 ):13 06-1319.
118. Ramos SA, Armitage LH, Morton JJ, et al. Generation of functional
thymic organoids from human pluripotent stem cells. Stem Cell
Reports. 2023;18(4):829-840.
119. Provin N, Giraud M . Differentiation of pluripotent stem cells into
thymic epithelial cells and generation of thymic organoids: appli-
cations for therapeutic strategies against APECED. Front Immunol.
2022;13:930963.
120. Reinisch A, Thomas D, Corces MR, et al. A humanized bone mar-
row ossicle xenotransplantation model enables improved engraft-
ment of healthy and leukemic human hematopoietic cells. Nat Med.
2016;22(7):812-821.
How to cite this article: Bin Y, Ren J, Zhang H, et al. Against
all odds: The road to success in the development of human
immune reconstitution mice. Anim Models Exp Med.
2024;00:1-11. doi:10.1002/ame2.124 07
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Severe combined immunodeficient (SCID) mice serve as a critical model for human xenotransplantation studies, yet they often suffer from low engraftment rates and susceptibility to graft-versus-host disease (GVHD). Moreover, certain SCID strains demonstrate ‘immune leakage’, underscoring the need for novel model development. Here, we introduce an SCID mouse model with a targeted disruption of the dclre1c gene, encoding Artemis, which is essential for V(D)J recombination and DNA repair during T cell receptor (TCR) and B cell receptor (BCR) assembly. Artemis deficiency precipitates a profound immunodeficiency syndrome, marked by radiosensitivity and compromised T and B lymphocyte functionality. Utilizing CRISPR/Cas9-mediated gene editing, we generated dclre1c-deficient mice with an NOD genetic background. These mice exhibited a radiosensitive SCID phenotype, with pronounced DNA damage and defective thymic, splenic and lymph node development, culminating in reduced T and B lymphocyte populations. Notably, both cell lines and patient-derived tumor xenografts were successfully engrafted into these mice. Furthermore, the human immune system was effectively rebuilt following peripheral blood mononuclear cells (PBMCs) transplantation. The dclre1c-knockout NOD mice described herein represent a promising addition to the armamentarium of models for xenotransplantation, offering a valuable platform for advancing human immunobiological research.
Article
Full-text available
Humanized mice have been used to study human immunodeficiency virus type 1 (HIV-1) transmission, pathogenesis, and treatment. The ability of pediatric thymus tissue implanted either in the leg (Leg PedThy) or under the renal capsule (Renal PedThy) with allogeneic CD34+ hematopoietic cells (HSCs) in NSG mice was evaluated for reconstitution of human immune cells and for rectal transmission of HIV-1. These mice were compared to traditional BLT mice implanted with fetal liver and thymus under the renal capsule and mice injected only with HSCs. Renal PedThy mice had similar immune reconstitution in the blood, spleen and intestine as BLT mice, while Leg PedThy mice had transient detection of immune cells, particularly CD4+ T cells and macrophages, the target cells for HIV-1 infection. Rectal transmission and replication of HIV-1 was efficient in BLT mice but lower and more variable in Renal PedThy mice. HIV-1 was poorly transmitted in HSC mice and not transmitted in Leg PedThy mice, which correlated with the frequencies of target cells in the spleen and intestine. Humanization of NSG mice with pediatric thymus was successful when implanted under the kidney capsule, but led to less efficient HIV-1 rectal transmission and replication compared to BLT mice.
Article
Full-text available
The thymus is critical for the establishment of a functional and self-tolerant adaptive immune system but involutes with age, resulting in reduced naive T cell output. Generation of a functional human thymus from human pluripotent stem cells (hPSCs) is an attractive regenerative strategy. Direct differentiation of thymic epithelial progenitors (TEPs) from hPSCs has been demonstrated in vitro, but functional thymic epithelial cells (TECs) only form months after transplantation of TEPs in vivo. We show the generation of TECs in vitro in isogenic stem cell-derived thymic organoids (sTOs) consisting of TEPs, hematopoietic progenitor cells, and mesenchymal cells, differentiated from the same hPSC line. sTOs support T cell development, express key markers of negative selection, including the autoimmune regulator (AIRE) protein, and facilitate regulatory T cell development. sTOs provide the basis for functional patient-specific thymic organoid models, allowing for the study of human thymus function, T cell development, and transplant immunity.
Article
Full-text available
Background Mesenchymal stem cells (MSCs) have shown immense therapeutic potential for various brain diseases. Intrathecal administration of MSCs may enhance their recruitment to lesions in the central nervous system, but any impact on cerebrospinal fluid (CSF) flow remains unclear. Methods Rats with or without middle cerebral artery occlusion (MCAO) received intrathecal injections of 2D cultured MSCs, 3D cultured MSCs or an equal volume of artificial cerebrospinal fluid (ACSF). Ventricle volume was assessed by MRI on Days 2 and 14 post-MCAO surgery. A beam walking test was used to assess fine motor coordination and balance. Aggregation of MSCs was evaluated in CSF and frozen brain tissue. Differential expression of cell adhesion molecules was evaluated by RNA-Seq, flow cytometry and immunofluorescence analyses. The influence of VCAM-1 blockade in mediating the aggregation of 2D MSCs was investigated in vitro by counting cells that passed through a strainer and in vivo by evaluating ventricular dilation. Results MSC expanded in 2D culture formed aggregates in the CSF and caused ventricular enlargement in both MCAO and normal rats. Aggregates were associated with impaired motor function. 2D MSCs expressed higher levels of integrin α4 and VCAM-1 than 3D MSCs. Blockade of VCAM-1 in 2D MSCs reduced their aggregation in vitro and reduced lateral ventricular enlargement after intrathecal infusion. 3D MSCs exhibited lower cell aggregation and reduced cerebral ventricular dilation after intrathecal transplantation Conclusions The aggregation of 2D MSCs, mediated by the interaction of integrin α4 and VCAM-1, is a potential risk for obstruction of CSF flow after intrathecal transplantation.
Article
Full-text available
Hematopoietic humanized (hu) mice are powerful tools for modeling the action of human immune system and are widely used for preclinical studies and drug discovery. However, generating a functional human T cell compartment in hu mice remains challenging, primarily due to the species-related differences between human and mouse thymus. While engrafting human fetal thymic tissues can support robust T cell development in hu mice, tissue scarcity and ethical concerns limit their wide use. Here, we describe the tissue engineering of human thymus organoids from inducible pluripotent stem cells (iPSC-thymus) that can support the de novo generation of a diverse population of functional human T cells. T cells of iPSC-thymus-engrafted hu mice could mediate both cellular and humoral immune responses, including mounting robust proinflammatory responses on T cell receptor engagement, inhibiting allogeneic tumor graft growth and facilitating efficient Ig class switching. Our findings indicate that hu mice engrafted with iPSC-thymus can serve as a new animal model to study human T cell-mediated immunity and accelerate the translation of findings from animal studies into the clinic. Engraftment of human thymic organoids supports de novo development of a functional human T cell compartment in a humanized mouse model.
Article
Full-text available
Background: Bone marrow transplantation (BMT) can be applied to both hematopoietic and nonhematopoietic diseases; nonetheless, it still comes with a number of challenges and limitations that contribute to treatment failure. Bearing this in mind, a possible way to increase the success rate of BMT would be cotransplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) to improve the bone marrow niche and secrete molecules that enhance the hematopoietic engraftment. Aim: To analyze HSC and MSC characteristics and their interactions through cotransplantation in murine models. Methods: We searched for original articles indexed in PubMed and Scopus during the last decade that used HSC and MSC cotransplantation and in vivo BMT in animal models while evaluating cell engraftment. We excluded in vitro studies or studies that involved graft versus host disease or other hematological diseases and publications in languages other than English. In PubMed, we initially identified 555 articles and after selection, only 12 were chosen. In Scopus, 2010 were identified, and six were left after the screening and eligibility process. Results: Of the 2565 articles found in the databases, only 18 original studies met the eligibility criteria. HSC distribution by source showed similar ratios, with human umbilical cord blood or animal bone marrow being administered mainly with a dose of 1 × 107 cells by intravenous or intrabone routes. However, MSCs had a high prevalence of human donors with a variety of sources (umbilical cord blood, bone marrow, tonsil, adipose tissue or fetal lung), using a lower dose, mainly 106 cells and ranging 104 to 1.5 × 107 cells, utilizing the same routes. MSCs were characterized prior to administration in almost every experiment. The recipient used was mostly immunodeficient mice submitted to low-dose irradiation or chemotherapy. The main technique of engraftment for HSC and MSC cotransplantation evaluation was chimerism, followed by hematopoietic reconstitution and survival analysis. Besides the engraftment, homing and cellularity were also evaluated in some studies. Conclusion: The preclinical findings validate the potential of MSCs to enable HSC engraftment in vivo in both xenogeneic and allogeneic hematopoietic cell transplantation animal models, in the absence of toxicity.
Article
Full-text available
Human innate immunity plays a critical role in tumor surveillance and in immunoregulation within the tumor microenvironment. Natural killer (NK) cells are innate lymphoid cells that have opposing roles in the tumor microenvironment, including NK cell subsets that mediate tumor cell cytotoxicity and subsets with regulatory function that contribute to the tumor immune suppressive environment. The balance between effector and regulatory NK cell subsets has been studied extensively in murine models of cancer, but there is a paucity of models to study human NK cell function in tumorigenesis. Humanized mice are a powerful alternative to syngeneic mouse tumor models for the study of human immuno‐oncology and have proven effective tools to test immunotherapies targeting T cells. However, human NK cell development and survival in humanized NOD‐scid‐IL2rgnull (NSG) mice are severely limited. To enhance NK cell development, we have developed NSG mice that constitutively expresses human Interleukin 15 (IL15), NSG‐Tg(Hu‐IL15). Following hematopoietic stem cell engraftment of NSG‐Tg(Hu‐IL15) mice, significantly higher levels of functional human CD56+ NK cells are detectable in blood and spleen, as compared to NSG mice. Hematopoietic stem cell (HSC)‐engrafted NSG‐Tg(Hu‐IL15) mice also supported the development of human CD3+ T cells, CD20+ B cells, and CD33+ myeloid cells. Moreover, the growth kinetics of a patient‐derived xenograft (PDX) melanoma were significantly delayed in HSC‐engrafted NSG‐Tg(Hu‐IL15) mice as compared to HSC‐engrafted NSG mice demonstrating that human NK cells have a key role in limiting the tumor growth. Together, these data demonstrate that HSC‐engrafted NSG‐Tg(Hu‐IL15) mice support enhanced development of functional human NK cells, which limit the growth of PDX tumors.
Article
Human immune cell reconstitution (Peripheral Blood Mononuclear Cells; PBMC or CD34 +hematopoietic stem cell; HSC) is commonly performed in immunodeficient mouse strains, such as NCG, NSG, or NOG. Human immune reconstitution utilizing PBMCs is a much more rapid model, allowing for T-cell reconstitution within 2 weeks. Due to the occurrence of xenogeneic graft-versus-host disease (xGvHD) around 4 weeks post-engrafment, the application of PBMC-reconstituted NCG mice is relatively limited. xGvHD is mainly induced by T cells derived from donor PBMC engraftment recognizing the murine host MHC class I and class II as foreign and attacking the host cells and organs. Deleting the β2m (NCG-β2m-KO) could reduce the occurrence of xGvHD; however, β2m is not only in the MHC class I subunit but also in the FcRn subunit, of which deletion shortens the half-life of IgG, making it unsuitable for IgG antibody agent evaluation. In addition, the deletion of MHC class I or class II alone results in an imbalanced CD4/CD8 ratio. To solve these problems, we developed the NCG-MHC-dKO mouse model by knocking out the H2K1, H2D1, and H2Ab1genes. Compared with the existing NCG-B2m-KO mouse model, the NCG-MHC-dKO significantly prolonged survival, reduced xGvHD occurrence, and did not affect the CD4/CD8 ratio when reconstituted with PBMC. Furthermore, treatment with anti-PD-L1 antibodies significantly inhibited MDA-MB-231 tumor cell growth in PBMC-reconstituted NCG-MHC-dKO mice, similar to NCG mice. Based on our preliminary data, the NCG-MHC-dKO mouse is a promising model for evaluating antibody and cell therapy agents, assessing treatment-related cytokine release syndrome, and evaluating long-term toxicities of cell therapies.
Article
Autoimmune diseases such as ankylosing spondylitis (AS) can be driven by emerging neoantigens that disrupt immune tolerance. Here, we developed a workflow to profile posttranslational modifications involved in neoantigen formation. Using mass spectrometry, we identified a panel of cysteine residues differentially modified by carboxyethylation that required 3-hydroxypropionic acid to generate neoantigens in patients with AS. The lysosomal degradation of integrin αIIb [ITGA2B (CD41)] carboxyethylated at Cys96 (ITGA2B-ceC96) generated carboxyethylated peptides that were presented by HLA-DRB1*04 to stimulate CD4 ⁺ T cell responses and induce autoantibody production. Immunization of HLA-DR4 transgenic mice with the ITGA2B-ceC96 peptide promoted colitis and vertebral bone erosion. Thus, metabolite-induced cysteine carboxyethylation can give rise to pathogenic neoantigens that lead to autoreactive CD4 ⁺ T cell responses and autoantibody production in autoimmune diseases.
Article
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 microbial 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 information 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.