Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Cytotherapy 26 (2024) 334 339

Contents lists available at ScienceDirect

CYTOTHERAPY journal homepage: www.isct-cytotherapy.org

FULL-LENGTH ARTICLE
Immunotherapy

Source of hematopoietic progenitor cells determines their capacity to


generate innate lymphoid cells ex vivo
Said Z. Omar1,2, Vera van Hoeven1,2, Nienke J.E. Haverkate1,2, Jolien M.R. Van der Meer2,3,
Carlijn Voermans2,3, Bianca Blom1,2, Mette D. Hazenberg1,2,3,4,5,*
1
Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
2
Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
3
Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
4
Cancer Center Amsterdam, Amsterdam, The Netherlands
5
Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands

A R T I C L E I N F O A B S T R A C T

Article History: Background aims: The success of allogeneic hematopoietic cell transplantation (HCT) as therapy for hemato-
Received 17 July 2023 logic conditions is negatively impacted by the occurrence of graft-versus-host disease (GVHD). Tissue dam-
Accepted 30 January 2024 age, caused, for example, by chemotherapy and radiotherapy, is a key factor in GVHD pathogenesis. Innate
lymphoid cells (ILCs) are important mediators of tissue repair and homeostasis. The presence of ILCs before,
Key Words: and enhanced ILC reconstitution after, allogeneic HCT is associated with a reduced risk to develop mucositis
allogeneic HCT and GVHD. However, ILC reconstitution after allogeneic HCT is slow and often incomplete. A way to replenish
GVHD the pool of ILC relies on the differentiation of hematopoietic progenitor cells (HPCs) into ILC.
immune reconstitution
Methods: We developed an ex vivo stromal cell containing culture system to study the capacity of HPCs to
ILC
differentiate into all mature helper ILC subsets.
ILC development
Results: ILC development depended on the source of HPCs. ILCs developed at high frequencies from umbilical
cord blood and fetal liver derived HPC and at low frequencies when HPCs were obtained from allogeneic
or autologous adult HCT grafts or healthy adult bone marrow. Although all helper ILC subsets could be gener-
ated from adult HPC sources, development of tissue protective ILC2 and NKp44+ ILC3 was notoriously
difficult.
Conclusions: Our data suggest that slow ILC recovery after allogeneic HCT may be related to an intrinsic inca-
pability of adult HPC to develop into ILC.
© 2024 International Society for Cell & Gene Therapy. Published by Elsevier Inc. This is an open access article
under the CC BY license (http://creativecommons.org/licenses/by/4.0/)

Introduction lymphocytes that additionally have immune-regulating properties.


ILCs do not express antigen-specific receptors like T cells or B cells
Graft-versus-host disease (GVHD) is a common complication of and respond to environmental factors such as aryl hydrocarbon
allogeneic hematopoietic cell transplantation (HCT) that is associated receptor ligands, short chain fatty acids and cytokines [5,6]. In the
with high morbidity and mortality [1,2]. Tissue damage, induced, for human intestine, which is one of the organs most affected in acute
example, by conditioning chemotherapy and/or irradiation, plays an GVHD, IL-22 producing ILC3 mediate tissue homeostasis and tissue
important role in the development of mucositis and the pathophysi- repair [7,8]. In addition, ILCs can have immunosuppressive properties
ology of GVHD. Tissue damage leads to activation and expansion of when expressing the ecto-enzymes CD39+ and CD73+. Ecto+ ILC3
donor alloreactive lymphocytes, resulting in a cascade of inflamma- metabolize adenosine triphosphate, released by damaged cells, into
tion and more tissue destruction [3,4]. Tissue repair can be mediated adenosine that suppresses activation of co-cultured T cells [9]. Other
by innate lymphoid cells (ILCs), which are a family of tissue-resident ILC subsets include interferon-g producing ILC1, and ILC2 that pro-
duce “type 2” cytokines like interleukin (IL)-5, IL-9, IL-13 and the tis-
sue protective factor amphiregulin [10 12].
* Correspondence: Prof. dr. Mette D. Hazenberg, Department of Hematology, The tissue healing and immune modulating capacities of ILCs have
Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands. led to studies investigating their role in the context of GVHD. In a
E-mail address: m.d.hazenberg@amsterdamumc.nl (M.D. Hazenberg).

https://doi.org/10.1016/j.jcyt.2024.01.013
1465-3249/© 2024 International Society for Cell & Gene Therapy. Published by Elsevier Inc. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/)
S.Z. Omar et al. / Cytotherapy 26 (2024) 334 339 335

previous study, we observed that patients with relatively high fre- and lin CD3 CD34+CD45RA HPCs were sorted, and 5.000 HPCs
quencies of circulating activated ILCs before and/or after allogeneic were seeded on to 5.000 OP9-JAG1 cells in Minimum Essential
HCT had a lower risk to develop mucositis and acute GVHD [13,14]. Medium a supplemented with 20% fetal calf serum, p/s, 10 Units/mL
Moreover, patients who developed acute or chronic GVHD had lower recombinant human (rh)IL-2 (BioLegend, San Diego, CA, USA),
numbers of circulating ILCs, and biopsies of GVHD-affected tissues 10 ng/mL rhIL-7 (Tebubio, Le Perray-en-Yvelines, France), 10 ng/mL
demonstrated a significant reduction of ILC in these tissues [9]. rhSCF (Invitrogen) and 10 ng/mL rhFlt3L (Tebubio; Figure 1A).
Rapid reconstitution of ILC after chemotherapy-induced depletion Cells were transferred to freshly plated OP9-JAG1 cells once a week.
and after allogeneic HCT thus seems favorable. However, ILC recovery Cytokines were replenished twice a week.
after allogeneic HCT, in fact, was slow, and ILC numbers did not reach
normal levels until 6 months’ post-transplantation [13]. Flow cytometry and cytokines
The requirements and mechanisms determining ILC reconstitu-
tion remain largely unknown. We hypothesized that, on the one Intracellular cytokine production was measured by stimulating
hand, expansion of mature ILCs present in the graft may contribute the cells with 10 ng/mL phorbol myristate acetate (Sigma) and
to post-transplantation ILC reconstitution. By investigating the com- 500 nmol/L Ionomycin (Merck, Rahway, NJ, USA) for 6 hours at 37°C,
position of adult HCT grafts, we showed that the presence of mature with GolgiPlug (BD Biosciences, San Diego, CA, USA) added for the
ILCs in the graft was associated with ILC recovery after transplanta- final 4 hours. To measure intracellular transcription factors or cyto-
tion. Patients with greater-than-average proportions of ILCs in the kines, cells were incubated with Zombie UV Dye (BioLegend) and cell
graft had a reduced risk to develop acute GVHD after transplantation surface antigen monoclonal antibodies, incubated with Fixation
[14]. On the other hand, or in addition, ILC reconstitution may rely on Buffer and Intracellular staining Perm Wash Buffer (Thermo Fisher
the development of ILCs from graft-derived multipotent hematopoi- Scientific, Waltham, MA, USA) and stained with transcription factor/
etic progenitor cells (HPCs). It is known that ILCs develop from com- cytokine antibodies. Data were acquired with a LSRII flow cytometer
mon lymphoid progenitors in the bone marrow. Differentiation of (Becton Dickinson) and analyzed using FlowJo software.
common lymphoid progenitors into mature ILC requires the tran-
scription factors DNA-binding protein inhibitor ID2, nuclear factor Quantitative real-time polymerase chain reaction (PCR)
interleukin-3 regulated protein, the cytokines IL-7 and stem cell fac-
tor (SCF) and cell surface expression of Notch ligands [15 17]. Quantitative PCR was performed on RNA of CD45+Lin CD127+
In the present study, we investigated the potential of human HPCs cells that were sorted after 4 weeks of culture. RNA was isolated
obtained from fetal, neonatal and adult sources to develop into using the NucleoSpin RNA XS kit (Macherey-Nagel, Du € ren, Nordr-
mature ILC subsets. We established an ex vivo differentiation model hein-Westfalen, Germany) and reversed transcribed into comple-
allowing for simultaneous differentiation of HPCs into all helper ILC mentary DNA (cDNA) using the High Capacity cDNA Archive Kit
subsets. We found that independent of the source, HPCs were able (Applied Biosystems, Waltham, MA, USA). Quantitative PCR was per-
to give rise to all helper ILC subsets ex vivo, although the relative formed to measure expression of the Tbx21, Gata3 and Rorc genes in
frequencies of ILC that developed in this model differed between HPC a CFX Connect Real-Time System using IQ SYBR Green Supermix
sources. (both Bio-Rad, Hercules, CA, USA). Primer sets are listed in supple-
mentary Table 2. Expression was analyzed using Bio-Rad CFX Man-
Methods ager 3.1 software and normalized to the expression of
glyceraldehyde 3-phosphate dehydrogenase.
Human cells
Statistical analysis
Study protocols were approved by the Medical Ethical Committee
of the Amsterdam University Medical Centers location AMC and San- Results are presented as median plus interquartile range. To cal-
quin Blood Supply (Amsterdam, The Netherlands). Human fetal liver culate statistical significance, the Wilcoxon signed-rank test or Man-
was obtained from elective abortions (14 20 weeks of gestational n Whitney U test was used (GraphPad Prism software). *P < 0.05
age) and umbilical cord blood from term deliveries. Adult bone mar- and **P < 0.01 were considered statistically significant.
row cells were obtained from patients who underwent sternotomy
for thoracic surgery. Aliquots of granulocyte colony-stimulating fac- Results
tor mobilized peripheral blood allogeneic and autologous HCT grafts
were collected from apheresis materials [14]. Fetal liver tissues were ILC can be generated from CD34+ HPCs
mechanically disrupted using the Stomacher 80 Biomaster (Seward,
West Sussex, UK). Mononuclear cells were isolated by Ficoll-Paque First, we set up a protocol to differentiate CD34+ HPCs into mature
density gradient centrifugation (Lymphoprep; GE Healthcare, Chi- ILC ex vivo. We sorted CD34+ HPC populations from healthy human
cago, IL, USA), CD34+ cells purified (CD34 MicroBead kit; Miltenyi fetal liver by fluorescence-activated cell sorting based on the differ-
Biotec, San Diego, CA, USA), frozen in 20% FCS containing 10% DMSO ential expression of CD45RA into common CD34+CD45RA HPCs and
and kept in liquid nitrogen. early lymphoid progenitor CD34+CD45RA+ HPCs (Figure 1A) [18,19].
Each HPC subset was co-cultured with OP9 mouse stromal cells that
HPC culture constitutively expressed the Notch ligand Jagged-1 (OP9-JAG1), as
Notch signaling has been shown to contribute to ILC development
OP9 murine stromal cells transfected with Notch ligand Jagged 1 [20]. Cultures were supplemented with a differentiation and expan-
(OP9-JAG1) were maintained in Minimum Essential Medium a sion cytokine cocktail that included IL-7, SCF, FLT3L and/or IL-2, up to
medium (Invitrogen, Waltham, MA, USA) supplemented with 8% fetal 28 days (Figure 1B). Flow cytometric analysis was performed on days
calf serum and 1% penicillin/streptomycin (Sigma-Aldrich, St. Louis, 14, 21 and 28. Mature ILCs were defined as live, lineage negative
MO, USA). Aliquots of CD34-positive hematopoietic stem cells were (lin ) CD45+CD3 CD127+ lymphocytes. Using this protocol, both HPC
thawed, stained with fluorochrome-conjugated antibodies (supple- subsets were able to differentiate into ILC, but the percentage of ILCs
mentary Table 1) and LIVE/DEAD Fixable Green Dead Cell Stain (Invi- obtained from CD34+CD45RA+ HPC was greater than that derived
trogen), and sorted with a FACSAria (Becton Dickinson). Flow from CD34+CD45RA HPC (Figure 1C). For follow-up experiments, we
cytometry purified human live, lineage (lin) CD3 CD34+CD45RA+ therefore focused on CD34+CD45RA+HPC.
336 S.Z. Omar et al. / Cytotherapy 26 (2024) 334 339

Figure 1. Generation of ILC and ILC subsets from CD34+ HPCs. (A) Representative flow cytometry dot plot of CD34+CD45RA and CD34+CD45RA+ HPC derived from human fetal liver.
The lineage (lin) antibody cocktail included antibodies directed against CD1a, CD4, CD5, CD14, CD16, CD19, CD94, CD123, BDCA2, TCRab, TCRgd and FcƐR1a. (B) Experimental setup
to culture purified CD34+CD45RA and CD34+CD45RA+ HPC on OP9-JAG1 expressing stromal cells for 28 days in the presence of differentiation and expansion recombinant human
cytokine cocktails as indicated. (C) ILC development from CD34+CD45RA or CD34+CD45RA+ HPC after 14 (n = 5), 21 (n = 8) and 28 (n = 8) days of culture. (D) ILC subsets derived
from fetal liver HPC (n = 8) at day 28. ILC subsets are defined as CRTH2 CD117 NKp44 ILC1, CRTH2+ ILC2 and CRTH2 CD117+ NKp44 or NKp44+ ILC3. (E) Frequency of transcrip-
tion factors analyzed by flow cytometry (n = 5) or (F) qPCR (n = 5). (G) Flow cytometry analysis of intracellular cytokine expression (n = 4). Data represent mean plus standard devia-
tion of n independent experiments. *P < 0.05; **P < 0.01 by Wilcoxon signed-rank test. (Color version of figure is available online.)

To evaluate which helper ILC subsets were generated ex vivo, expression of RORg t. We found that most of the CD127+ ILC
we analyzed the phenotype of ILC. Lin CD45+CD127+ helper expressed RORg t, consistent with the fact that most cells expressed
ILC subsets were defined by distinct cell-surface markers as an ILC3 phenotype (supplementary Figure 1B and Figure 1E). Expres-
CRTH2 CD117 NKp44 ILC1, CRTH2+ ILC2 and NKp44 or NKp44+ sion of GATA3 was detected in 25% of the ILC in line with the percent-
CRTH2 CD117+ ILC3. All ILC subsets differentiated from age of ILC2 generated in the cultures. Expression of T-bet, which is
CD34+CD45RA+ fetal liver HPC in culture, with the lowest frequency associated with ILC1, and EOMES, associated with natural killer cells,
being observed for ILC1 (Figure 1D). To confirm that cells generated was lower compared with RORg t and GATA3 (Figure 1E), which is
were bona fide ILC, we evaluated signature transcription factor also consistent with the ILC phenotype distribution (Figure 1D).
expression by flow cytometry (Figure 1E and supplementary Figure Finally, to evaluate the functional capacity of ex vivo generated
1A) and quantitative PCR (Figure 1F). ILC1s are defined by the expres- ILC subsets, we analyzed signature cytokine production in
sion of Tbet, ILC2 express GATA3 and ILC3 are characterized by the lin CD127+EOMES helper ILC after restimulation with phorbol
S.Z. Omar et al. / Cytotherapy 26 (2024) 334 339 337

myristate acetate and ionomycin (Figure 1G). We observed intracel- that are used for allogeneic HCT are also able to differentiate into
lular expression of interferon-g , IL-13, IL-17 and IL-22, which is asso- mature ILC subsets in this model. Bone marrow aspirates, umbilical
ciated with the presence of ILC1, ILC2 and ILC3, respectively cord blood samples, peripheral blood granulocyte colony-stimulating
(Figure 1G). Taken together, these data demonstrate that HPCs iso- factor mobilized autologous and allogeneic HCT grafts all contained
lated from human fetal liver are able to differentiate into all mature CD34+CD45RA and CD34+CD45RA+ HPC, albeit at different ratios
helper ILC subsets ex vivo and express signature transcription factors (Figure 2A). In all sources, the frequency of CD34+CD45RA HPC was
and effector cytokines. greater than the frequency of CD34+CD45RA+ HPC. We sorted
CD34+CD45RA+ HPC from each source and cultured the cells for
Generation of ILCs from HPC differs between sources 28 days ex vivo as described for fetal liver derived HPCs (Figure 1B).
We observed that HPCs of all sources had the capacity to differentiate
Having established an ex vivo model for development of all ILC into mature ILCs (Figure 2B). However, significantly greater propor-
subsets, we next investigated whether HPCs derived from sources tions of ILCs were generated from fetal liver and umbilical cord blood

Figure 2. Generation of ILCs from HPC differs between stem cell sources. (A) Frequency of CD34+CD45RA and CD34+CD45RA+ HPCs present in adult G-CSF mobilized peripheral
blood allogeneic HCT grafts (ALLO) (n = 4), autologous HCT grafts (AUTO) (n = 4), adult bone marrow (BM) (n = 3), umbilical cord blood (UCB) (n = 3) and fetal liver (FL) (n = 7). (B)
Frequency of ILC and (C) frequency of ILC subsets obtained from fetal, neonatal and adult (n = 4) CD34+CD45RA+ HPC sources after 28 days of culture. Data represent mean and stan-
dard deviation of n independent experiments. *P < 0.05; **P < 0.01 by non-parametric paired Wilcoxon t-test (A) or Mann Whitney U test (B-C). G-CSF, granulocyte colony stimu-
lating factor. (Color version of figure is available online.)
338 S.Z. Omar et al. / Cytotherapy 26 (2024) 334 339

as compared with adult bone marrow and mobilized autologous ILC subsets have not been evaluated to date, these data raise the
or allogeneic HCT grafts. A similar trend was observed when we hypothesis that human ILC development may also rely on the thy-
cultured CD34+CD45RA HPCs (supplementary Figure 2). All helper mus, similar as naive T-cell development. Since thymic activity at
ILC subsets were generated independent of the source of HPC, but adult age is notorious low, this may provide an additional explana-
the relative frequencies of ex vivo generated ILC subsets did tion for the slow recovery of ILC2 after allogeneic HCT.
differ depending on the source of HPC (Figure 2C and supplementary ILC3s were generated from HPCs at greater frequencies than ILC2,
Figure 2B). Umbilical cord blood derived HPC differentiated mostly irrespective of the HPC source. This is in line with the observation
into NKp44+ ILC3 and NKp44 ILC3, whereas adult bone mar- that in adults who received allogeneic HCT with adult HPC ILC3
row derived HPC mostly gave rise to ILC1 and NKp44 ILC3. HPCs reconstitution was relatively fast, when compared with ILC2, with
from autologous HCT grafts predominantly differentiated into relatively high numbers of circulating ILC3 compared with numbers
NKp44 ILC3. Unfortunately, the number of ILCs that differentiated obtained from healthy individuals [13]. Altogether, these findings
from HPCs sorted from allogeneic HCT grafts was too low to allow for support the hypothesis that early ILC reconstitution after allogeneic
reliable ILC subset analysis. We conclude that ILCs can be generated HCT with adult HPC may depend more on expansion of mature ILCs
ex vivo from all HPC sources but that the relative frequencies of ILC present in the graft rather than de novo generation of ILCs from HPCs,
subsets generated from these HPC do depend on the source of HPCs. in analogy with post-transplantation T-cell reconstitution.
In conclusion, stem cell grafts contain HPCs that are able to differenti-
Discussion ate into mature ILCs. These results are important because better under-
standing of ILC reconstitution may pave the way for novel approaches to
Donor-derived ILCs likely play an important role in protecting tis- improve ILC reconstitution, such as adoptive transfer of mature ILC,
sues of allogeneic HCT recipients against the development of tissue which may help to prevent GVHD in allogeneic HCT recipients.
damage and acute GVHD [9,13,14,21 23]. Similar to T cells, however,
ILCs have slow reconstitution dynamics after allogeneic HCT Declaration of Competing Interest
[13,14,21,24 26]. T-cell reconstitution after transplantation is a
biphasic process that initially, in the first few months after transplan- The authors have no commercial, proprietary or financial interest
tation, is dominated by expansion of mature T cells that were present in the products or companies described in this article.
in the graft. Later, naive T cells that are produced de novo from trans-
planted donor HPC in the thymus enter the circulation and contribute Funding
to a more diverse T-cell population [27 29]. It can be envisioned that
ILC reconstitution follows a similar dynamic. We previously demon- This study was supported by a VIDI grant (NWO ZonMW
strated that mature ILCs are present in allogeneic HCT grafts and that #91715362; M.D.H.) and a LSBR Fellowship (1438F; M.D.H.).
the proportion of ILC in the grafts tended to correlate with ILC recon-
stitution after transplantation [14]. To what extend de novo develop- Author Contributions
ment of ILC from HPC contributes to ILC reconstitution in allografted
recipients remains to be determined. Conception and design of the study: BB and MDH. Acquisition of
Here, we investigated the capacity of human HPCs derived from data: SZO, NJEH and VvH. Analysis and interpretation of data: SZO,
sources used for transplantation, such as mobilized peripheral blood, NJEH and VvH. Drafting or revising the manuscript: SZO, NJEH, VvH,
bone marrow and umbilical cord blood, to differentiate into mature JMRvdM, CV, BB and MDH. All authors have approved the final
ILCs. For this, we set up an ex vivo differentiation model using fetal article.
liver HPC that allows for simultaneous development of all helper ILC
subsets, using a Notch ligand expressing mouse stromal cell line.
Acknowledgments
Both CD34+CD45RA and CD34+CD45RA+ fetal liver HPCs were able
to differentiate into ILC, although CD34+CD45RA+ HPC were more
The authors thank Esther Siteur van Rijnstra and Cynthia van der
potent, which may be related to the fact that CD34+CD45RA+ HPC are
Linden of the Mice Facility for providing the OP9 stromal cells, and
more committed to the lymphoid lineage than CD34+CD45RA HPC
fetal liver CD34+ HPC. The authors also thank the Flow Cytometry
[18,19]. We found that under these culture conditions HPC isolated
Facility of the Amsterdam UMC, location AMC, department of EXIM
from pre- and neonatal sources, i.e., fetal liver and umbilical cord
for their excellent technical support.
blood, have a better capacity to differentiate toward ILCs than HPC
from adult sources, which yielded significantly fewer ILCs. This is in
Data Availability
line with previous findings, where HPCs were cultured under
stroma-free conditions [30]. Together, these data suggest that if ILC
Data and protocols will be made available upon written request to
development is dependent on the age of the HPCs, and we assume
the corresponding author.
that de novo generation of ILC from transplanted HPCs is an important
contributor to ILC reconstitution after allogeneic HCT, ILC recovery
may be better after transplantation with CB-derived HPCs compared Supplementary materials
with transplantation of adult donor derived HPCs. In a small study
performed among a diverse cohort of pediatric and adult allogeneic Supplementary material associated with this article can be found
HCT recipients, however, no clear differences in ILC reconstitution in the online version at doi:10.1016/j.jcyt.2024.01.013.
were observed between pediatric patients who received cord blood
or adult HPCs [21]. The observation in this study that ILC2 reconstitu- References
tion was significantly slower in adults receiving adult HPCs compared
[1] Blazar BR, Murphy WJ, Abedi M. Advances in graft-versus-host disease biology
with pediatric allogeneic HCT recipients who received cord blood
and therapy. Nat Rev Immunol 2012;12(6):443–58.
HPC [21] does suggest that age-related factors play a role in ILC2 [2] Ferrara JL, Levine JE, Reddy P, Holler E. Graft-versus-host disease. Lancet 2009;373
reconstitution. In mice, ILC2 were reported to harbor abortive T-cell (9674):1550–61.
receptor (TCR)g rearrangements, but no other TCR transcripts, and [3] Ara T, Hashimoto D. Novel insights into the mechanism of GVHD-induced tissue
damage. Front Immunol 2021;12:713631.
intracellular CD3 [31,32]. In human fetal thymus, CD5+ ILC progeni- [4] Gyurkocza B, Sandmaier BM. Conditioning regimens for hematopoietic cell trans-
tors have been identified [33]. Although genomic TCR loci in human plantation: one size does not fit all. Blood 2014;124(3):344–53.
S.Z. Omar et al. / Cytotherapy 26 (2024) 334 339 339

[5] Sonnenberg GF, Artis D. Innate lymphoid cells in the initiation, regulation and res- [20] Gentek R, Munneke JM, Helbig C, Blom B, Hazenberg MD, Spits H, Amsen D. Mod-
olution of inflammation. Nat Med 2015;21(7):698–708. ulation of signal strength switches notch from an inducer of T cells to an inducer
[6] Montaldo E, Juelke K, Romagnani C. Group 3 innate lymphoid cells (ILC3s): origin, of ILC2. Front Immunol 2013;4:334.
differentiation, and plasticity in humans and mice. Eur J Immunol 2015;45 [21] Piperoglou C, Larid G, Vallentin B, Balligand L, Crinier A, Banzet N, et al. Innate
(8):2171–82. lymphoid cell recovery and occurrence of GVHD after hematopoietic stem cell
[7] Hepworth MR, Fung TC, Masur SH, Kelsen JR, McConnell FM, Dubrot J, et al. transplantation. J Leukoc Biol 2022;111(1):161–72.
Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of [22] Shao L, Pan S, Zhang QP, Jamal M, Chen LH, Yin Q, et al. An essential role of innate
commensal bacteria-specific CD4(+) T cells. Science 2015;348(6238):1031–5. lymphoid cells in the pathophysiology of graft-vs.-host disease. Front Immunol
[8] Qiu J, Guo X, Chen ZM, He L, Sonnenberg GF, Artis D, et al. Group 3 innate lymphoid 2019;10:1233.
cells inhibit T-cell mediated intestinal inflammation through aryl hydrocarbon [23] Bruce DW, Stefanski HE, Vincent BG, Dant TA, Reisdorf S, Bommiasamy H, et al.
receptor signaling and regulation of microflora. Immunity 2013;39(2):386–99. Type 2 innate lymphoid cells treat and prevent acute gastrointestinal graft-ver-
[9] Hazenberg MD, Haverkate NJE, van Lier YF, Spits H, Krabbendam L, Bemelman sus-host disease. J Clin Invest 2017;127(5):1813–25.
WA, et al. Human ectoenzyme-expressing ILC3: immunosuppressive innate cells [24] Williams KM, Hakim FT, Gress RE. T cell immune reconstitution following lym-
that are depleted in graft-versus-host disease. Blood Adv 2019;3(22):3650–60. phodepletion. Semin Immunol 2007;19(5):318–30.
[10] Bernink JH, Peters CP, Munneke M, te Velde AA, Meijer SL, Weijer K, et al. Human [25] Chang YJ, Zhao XY, Huang XJ. Immune reconstitution after haploidentical
type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immu- hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2014;20
nol 2013;14(3):221–9. (4):440–9.
[11] Spits H, Mjosberg J. Heterogeneity of type 2 innate lymphoid cells. Nat Rev Immu- [26] Ogonek J, Kralj Juric M, Ghimire S, Varanasi PR, Holler E, Greinix H, Weissinger E.
nol 2022;22(11):701–12. Immune reconstitution after allogeneic hematopoietic stem cell transplantation.
[12] Golebski K, Ros XR, Nagasawa M, van Tol S, Heesters BA, Aglmous H, et al. IL- Front Immunol 2016;7:507.
1beta, IL-23, and TGF-beta drive plasticity of human ILC2s towards IL- [27] Dekker L, Sanders E, Lindemans CA, de Koning C, Nierkens S. Naive T cells in graft
17 producing ILCs in nasal inflammation. Nat Commun 2019;10(1):2162. versus host disease and graft versus leukemia: innocent or guilty? Front Immunol
[13] Munneke JM, Bjorklund AT, Mjosberg JM, Garming-Legert K, Bernink JH, Blom B, 2022;13:893545.
et al. Activated innate lymphoid cells are associated with a reduced susceptibility [28] Hiwarkar P, Hubank M, Qasim W, Chiesa R, Gilmour KC, Saudemont A, et al.
to graft-versus-host disease. Blood 2014;124(5):812–21. Cord blood transplantation recapitulates fetal ontogeny with a distinct molecular
[14] Kroeze A, van Hoeven V, Verheij MW, Turksma AW, Weterings N, van Gassen S, signature that supports CD4(+) T-cell reconstitution. Blood Adv 2017;1
et al. Presence of innate lymphoid cells in allogeneic hematopoietic grafts corre- (24):2206–16.
lates with reduced graft-versus-host disease. Cytotherapy 2022;24(3):302–10. [29] Roberto A, Castagna L, Zanon V, Bramanti S, Crocchiolo R, McLaren JE, et al. Role of
[15] Cherrier M, Sawa S, Eberl G. Notch, Id2, and RORgammat sequentially orchestrate naive-derived T memory stem cells in T-cell reconstitution following allogeneic
the fetal development of lymphoid tissue inducer cells. J Exp Med 2012;209 transplantation. Blood 2015;125(18):2855–64.
(4):729–40. [30] Moretta F, Petronelli F, Lucarelli B, Pitisci A, Bertaina A, Locatelli F, et al. The gen-
[16] Constantinides MG, McDonald BD, Verhoef PA, Bendelac A. A committed precur- eration of human innate lymphoid cells is influenced by the source of hematopoi-
sor to innate lymphoid cells. Nature 2014;508(7496):397–401. etic stem cells and by the use of G-CSF. Eur J Immunol 2016;46(5):1271–8.
[17] Hernandez DC, Juelke K, Muller NC, Durek P, Ugursu B, Mashreghi MF, et al. An in [31] Shin SB, Lo BC, Ghaedi M, Scott RW, Li Y, Messing M, et al. Abortive gammadel-
vitro platform supports generation of human innate lymphoid cells from CD34(+) taTCR rearrangements suggest ILC2s are derived from T-cell precursors. Blood
hematopoietic progenitors that recapitulate ex vivo identity. Immunity 2021;54 Adv 2020;4(21):5362–72.
(10). 2417-32 e5. [32] Bajana S, Pankow A, Liu K, Michniowska M, Urban Jr. JF, Chen WR, Sun XH. Corre-
[18] Scoville SD, Freud AG, Caligiuri MA. Cellular pathways in the development of lation between circulating innate lymphoid cell precursors and thymic function.
human and murine innate lymphoid cells. Curr Opin Immunol 2019;56:100–6. iScience 2022;25(2):103732.
[19] Galy AH, Cen D, Travis M, Chen S, Chen BP. Delineation of T-progenitor cell activ- [33] Nagasawa M, Germar K, Blom B, Spits H. Human CD5(+) innate lymphoid cells are
ity within the CD34+ compartment of adult bone marrow. Blood 1995;85 functionally immature and their development from CD34(+) progenitor cells is
(10):2770–8. regulated by Id2. Front Immunol 2017;8:1047.

You might also like