Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2017

Open Access 01.12.2017 | Research

Interleukin-21 promotes thymopoiesis recovery following hematopoietic stem cell transplantation

verfasst von: Aurélie Tormo, Fatemeh Khodayarian, Yun Cui, Edouard Al-Chami, Reem Kanjarawi, Beatriz Noé, Huijie Wang, Moutih Rafei

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2017

Abstract

Background

Impaired T cell reconstitution remains a major deterrent in the field of bone marrow (BM) transplantation (BMT) due to pre-conditioning-induced damages inflicted to the thymi of recipient hosts. Given the previously reported thymo-stimulatory property of interleukin (IL)-21, we reasoned that its use post-BMT could have a profound effect on de novo T cell development.

Methods

To evaluate the effect of IL-21 on de novo T cell development in vivo, BM derived from RAG2p-GFP mice was transplanted into LP/J mice. Lymphocyte reconstitution was first assessed using a hematological analyzer and a flow cytometer on collected blood samples. Detailed flow cytometry analysis was then performed on the BM, thymus, and spleen of transplanted animals. Finally, the effect of human IL-21 on thymopoiesis was validated in humanized mice.

Results

Using a major histocompatibility complex (MHC)-matched allogeneic BMT model, we found that IL-21 administration improves immune reconstitution by triggering the proliferation of BM LinSca1+c-kit+ (LSK) subsets. The pharmacological effect of IL-21 also culminates in the recovery of both hematopoietic (thymocytes) and non-hematopoietic (stromal) cells within the thymi of IL-21-treated recipient animals. Although T cells derived from all transplanted groups proliferate, secrete various cytokines, and express granzyme B similarly in response to T cell receptor (TCR) stimulation, full regeneration of peripheral naïve CD4+ and CD8+ T cells and normal TCRvβ distribution could only be detected in IL-21-treated recipient mice. Astonishingly, none of the recipient mice who underwent IL-21 treatment developed graft-versus-host disease (GVHD) in the MHC-matched allogeneic setting while the graft-versus-tumor (GVT) effect was strongly retained. Inhibition of GVHD onset could also be attributed to the enhanced generation of regulatory B cells (B10) observed in the IL-21, but not PBS, recipient mice. We also tested the thymopoiesis-stimulating property of human IL-21 in NSG mice transplanted with cord blood (CB) and found significant improvement in de novo human CD3+ T cell development.

Conclusions

In sum, our study indicates that IL-21 represents a new class of unforeseen thymopoietin capable of restoring thymic function following BMT.
Begleitmaterial
Additional file 1: Figure S1. Gating strategies for LSKs. BM cells collected from WT or IL-21R−/− C57BL/6 mice were treated in vitro for proliferation, then stained with the Lin−Sca1+c-kit+ antibody cocktail prior to Ki-67+ incorporation analysis. All described experiments were conducted at least three times with n = 5/group. Figure S2. Functional characterization of T cells. (A) Representative cell trace dilution analysis on CD4+ or CD8+ T cells derived from ctl (unirradiated), PBS-treated, or IL-21-treated LP/J recipient mice. (B) Cytokine quantification by ELISA from T cells derived from the same groups described in panel (A). (C) Representative flow cytometry analysis of granzyme B expression. (D) Quantification of T cells expressing granzyme B. For all presented studies, T cells were stimulated with CD3-CD28 dynabeads for 48 h prior to analyses. All described experiments were conducted at least three times with an n = 5/group. Figure S3. Molecular characterization of T cells. T cells sorted from ctl, PBS-treated, or IL-21-treated LP/J recipient mice were analyzed for their expression of various transcription factors involved in T cell differentiation. All described experiments were conducted at least three times with n = 5/group. Figure S4. Gating strategies for Breg analysis. For detection of IL-10-producing Bregs, CD19+ B cells were first isolated from spleens of treated mice (isotype shown by the filled gray histogram), then stained after in vitro treatment with CD1d and CD5 antibodies. The B cell subset CD1dhiCD5+ was gated prior to IL-10 assessment by intracellular staining. All described experiments were conducted at least three times with n = 5/group. (PDF 2111 kb)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13045-017-0490-3) contains supplementary material, which is available to authorized users.
Abkürzungen
B10
IL-10 producing regulatory B cells
BM
Bone marrow
BMT
Bone marrow transplantation
CB
Cord blood
DLI
Donor lymphocyte infusion
DN
Double-negative
DP
Double-positive
GVHD
Graft-versus-host-disease
GVT
Graft-versus-tumor
HSC
Hematopoietic stem cells
IL
Interleukin
IL-21R
IL-21 receptor
LSK
LinSca1+c-kit+
LT-HSCs
Long-term hematopoietic stem cells
MHC
Major histocompatibility complex
MPP
Multipotent progenitors
RTE
Recent thymic emigrant
SP
Single-positive
ST-HSCs
Short-term hematopoietic stem cells
TCR
T cell receptor
TEC
Thymic epithelial cell
Tregs
Regulatory T cells
WT
Wild-type

Background

BMT is commonly used in the clinic as a curative therapy for a variety of life-threatening diseases of lympho-hematopoietic origin [17]. Prior to BMT, patients undergo myeloablative or non-myeloablative conditioning regimens consisting of radiation, chemotherapy, and/or administration of immunosuppressive drugs [17]. Although innate immunity mediated by myeloid and natural killer cells recovers relatively quickly in patients of all ages, T cell reconstitution requires years and even then rarely re-establishes a fully competent T cell repertoire [810]. This prolonged post-transplant lymphoid deficiency is associated with increased risks of (i) cancer relapse or development of secondary malignancies [1117], (ii) infection [1821], and (iii) reduced responses to immunotherapies such as vaccination [5, 22]. In line with this notion, a clinical study based on 42 mantle cell lymphoma patients who underwent BMT revealed that absolute lymphocyte count recovery is a powerful prognostic factor for clinical outcome [23]. Furthermore, accelerated lymphocyte recovery culminates in improved responses to immune-based treatments as previously achieved with the combined use of rituximab and BMT [2326]. Thus, identifying factors capable of inducing de novo intrathymic T cell development (thymopoiesis) in patients undergoing BMT remains a central goal in the field.
IL-21 is the most recently identified member of the common γ-chain family of cytokines [27]. Some of its pleiotropic functions include (i) promoting Th17 differentiation in the presence of transforming growth factor-β [28], (ii) activating NK and CD8 lymphocytes [29, 30], (iii) desensitizing responder cells to the inhibitory effects of regulatory T cells (Tregs) [31], and (iv) acting as a switch for IgG production in B cells [32]. Although not required for hematopoiesis, in vivo expansion of bone marrow (BM) progenitors was observed in response to IL-21 overexpression [33]. Consistently, Simard et al. showed that IL-21 signaling in BM-derived B cell progenitors accelerates the development of immature/mature B cells by regulating the expression of Blimp1, Aid, and the germline transcript gamma 2b [34]. For the T cell lineage, however, IL-21 is not necessary for thymocyte differentiation due to normal thymopoiesis in mice exhibiting an IL-21 receptor (IL-21R) deficiency [27, 32]. Paradoxically, we previously reported that TCR engagement on double-positive (DP) thymocytes triggers the de novo expression of IL-21R [35]. Although DP thymocytes undergo a three- to fourfold expansion in response to IL-21 treatment in vitro, no differentiation towards CD8+ single-positive (SP) T cells could be detected [35]. As no mitogen has ever been described for DP thymocytes, we then tested the thymopoiesis-stimulating ability of IL-21 in models of acute and chronic thymic atrophy. For instance, IL-21 administration to mice suffering from corticosteroid-induced thymic atrophy led to double-negative (DN) and DP thymocyte expansion, which resulted in accelerated recovery of thymic function [36]. In wild-type (WT) aged mice displaying naturally induced thymic atrophy, IL-21 enhanced the output of recent thymic emigrants (RTEs), which qualitatively changed the peripheral T cell pool [37]. On top of correcting several T cell defects commonly seen with aging such as impaired proliferation and biased cytokine production, an enhanced anti-tumoral response to a Trp2-based melanoma cancer vaccine was achieved in aged mice pre-conditioned with IL-21 [37]. The overall significance of these findings epitomizes IL-21 as a potent pharmacological agent for stimulating thymopoiesis and serves as the basis to further investigate its use in T cell reconstitution following allogeneic BMT.

Methods

Cell line and mice

The P815 cell line was purchased from ATCC (Manassas, VA, USA). The RAG2p-GFP transgenic mice were kindly provided by Dr. M. Nussenzweig (Rockefeller University, NY, USA). C57BL/6 (H2-Kb), IL-7−/− (C57BL/6 background, H2-Kb), IL-21R−/− (C57BL/6 background, H2-Kb), LP/J (H2-Kbc), BALB/c (H2-Kd), MμMT (H2-Kb), and NSG mice were purchased from the Jackson Laboratory (Bar Harbor, ME). Littermate mice were interbred and housed in a pathogen-free environment at the animal facility of the Institute for Research in Immunology and Cancer. Animal protocols were approved by the Animal Care Committee of Université de Montréal.

Antibodies, cytokines, reagents, and kits

Recombinant IL-7 (#217-17) and IL-21 (#210-21 for mouse and #200-21 for human) cytokines were purchased from PeproTech (Rocky Hill, NJ, USA). The flow cytometry antibodies CD3 (17A2), CD4 (GK1.5), CD5 (53-7.3), CD8 (53-6.7), NK1.1 (PK136), CD19 (1D3), CD25 (PC61), CD34 (RAM34), CD44 (1M7), CD45 (30-F11), CD62L (MEL-14), CD135 (A2F10.1), IL-10 (JES5-16E3), human CD3 (UCHT1), human CD4 (RPA-T4), human CD8 (SK1), human CD13 (WM15), human CD45 (H130), and Cytofix/Cytoperm Kits were purchased from BD Pharmingen (San Diego, CA, USA). FOXP3 (FJK-16S), granzyme B (NGZB), and human CD161 (HP-3G10) antibodies as well as monensin were purchased from eBioscence/Thermo Fisher (Waltham, MA, USA). Quantikines were purchased from R&D Systems (Minneapolis, MN). CD3-CD28 beads, CellTraceTM, and TRIzol were purchased from Invitrogen (Burlington, ON, CA). Lipopolysaccharide (LPS), PMA, and ionomycin were purchased from Sigma (St. Louis, MO, USA). All cell isolation kits were purchased from STEMCELL Technologies (Vancouver, BC, Canada). RNA extraction kits were purchased from Qiagen (Toronto, ON, Canada).

BMT and immune recovery assessment

Irradiation of LP/J (11 Gy), IL-7−/− (8.5 Gy), or BALB/c (8.5 Gy) recipient female was conducted prior to transplantation with T cell-depleted 5 × 106 RAG2p-GFP-derived BM cells by intravenous injection. On the following day, mice were administered IL-21, IL-7 (both at 50 μg/kg), or equivalent volume (100 μl) of sterile PBS by intraperitoneal injections every 2 days over a period of 2 weeks (total of 6 injections). Immune reconstitution was assessed weekly using both the Scil vet ABC Plus+ hematological analyzer and flow cytometry on collected blood samples. BM, thymic, and spleen analyses were conducted as previously described [37].

GVHD/GVT induction and analysis

To induce GVHD, T cell-depleted BM derived from RAG2p-GFP or MμMT mice was supplemented with 106 enriched T cells (from RAG2p-GFP), then used to transplant irradiated LP/J recipient animals followed by PBS/IL-21 treatments as previously described. For assessment of GVHD severity, mice were scored individually for five clinical parameters on a scale from 0 to 2 for weight loss, posture, activity, fur, and skin lesions. For GVT experiments, transplanted LP/J mice where challenged intravenously with 5 × 105 P815 tumor cells the day following BMT. Mice were sacrificed when weight loss reached ≥20% and/or the sum of total clinical scores was above 8.

Proliferation, cytokine measurements, and intracellular staining

T cells were isolated from splenocyte suspension by negative selection and were stimulated with CD3-CD28 dynabeads. Cells or supernatants were collected 48 h post-stimulation for further analyses. For intracellular detection of cytokines, stimulated T cells were stained for CD4/CD8 prior to intracellular staining according to the manufacturer’s instructions. FOXP3 staining on T cells and IL-10 analysis in B cells were conducted as previously described [37, 38].

Expression analysis of transcription factors

T cells from spleens of transplanted mice were sorted in 900 μl TRIzol (106 cells per tube) followed by RNA extraction. Reverse transcription was performed by qPCR. Target gene values were normalized to endogenous control Gapdh.

NSG transplantation using CB

Cord blood (CB) units were obtained from the Ste-Justine blood bank (Montreal, Qc, Canada) following ethics approval. Mononucleated cells were first isolated by density centrifugation, then depleted from human T cells prior to their transplantation (105 cells) in sublethally irradiated (3 Gy) NSG mice [3941]. Transplanted NSG mice were then intraperitoneally injected with human IL-21 (50 or 100 μg/kg) or human IL-7 (50 μg/kg) versus equivalent volumes of PBS (100 μl) every 2 days over a period of 2 weeks (total of 6 injections). Peripheral blood was collected every 2 weeks and analyzed as described above. Transplanted mice were sacrificed at the end of the experiment to analyze their thymi and spleens.

Statistical analyses

P values were calculated using the ANOVA and log-rank statistical test where applicable.

Results

IL-21 administration post-BMT correlates with accelerated lymphoid recovery

Amongst all immune subsets, T cells are the slowest to emerge (if any) post-BMT [810]. Following their development in the thymus, newly generated CD4+ and CD8+ SP thymocytes, which are referred to as RTEs, egress to the periphery where they continue their maturation in secondary lymphoid organs [42]. To detect RTEs, peripheral blood samples are usually used to quantify TCR excision circle content, which represents a direct reflection of TCR rearrangements during intrathymic T cell development [4345]. However, TCR excision circle quantification by PCR has several limitations and remains an indirect method that needs to be interpreted with caution [4345]. Alternatively, RTEs and newly generated B cells both can be monitored using RAG2p-GFP mice, where GFP expression is controlled by the Rag2 promoter activity during T and B lymphopoiesis [37, 46, 47]. Therefore, we first quantified the physiological ranges of total GFP+ (RTEs and newly developed B cells), GFP+CD19+ (newly developed B cells), or GFP+CD3+ (RTEs) cells in circulation using blood samples collected from unirradiated control RAG2p-GFP mice prior to conducting BMT experiments (Fig. 1a). According to these ranges (displayed as pink-shaded areas), IL-21 administration to LP/J mice accelerated significantly lymphoid recovery in contrast to PBS- or IL-7-treated mice (Fig. 1b). In particular, GFP+CD19+ cells (Fig. 1c) reached physiological levels 3 weeks post-BMT whereas GFP+CD3+ levels (Fig. 1d) were normalized by the 5th week (indicated by red arrows) following transplantation. Upon further dissection, enhanced re-establishment of peripheral GFP+CD4+, GFP+CD8+, GFP+NKT+, and NK+ cells were observed at both 5th and 8th weeks post-BMT in IL-21-treated mice (Fig. 1e) with no overrepresentation of myeloid versus lymphoid cells in any of the transplanted groups (Fig. 1f).
Given the profound effect of IL-21 on T cell recovery post-BMT, we next examined whether it displays redundant function with IL-7, the major thymopoietin known to support T cell development [48]. To test this hypothesis, peripheral GFP+CD3+ T cells were quantified in IL-7−/− recipient mice transplanted with T cell-depleted RAG2p-GFP BM. Although no differences in the generation of GFP+CD3+ T cells were depicted between PBS- and IL-21-treated IL-7−/− recipient animals, the overall RTE levels were significantly lower compared to WT C57BL/6 recipient mice on the PBS regimen (Fig. 1g). These results clearly indicate that IL-21 can indeed accelerate T cell recovery but requires endogenous IL-7 signaling to exert its effects.

IL-21 triggers the expansion of BM progenitors

The thymus lacks self-renewing stem cells [49, 50]. Its function relies therefore on sustained seeding by BM-derived progenitors [49, 50]. Although the nature/phenotype of the thymus-settling population(s) remains a matter of debate, BM progenitors are still considered as the limiting substrate for immune reconstitution post-BMT [4958]. To determine whether the improved peripheral T cell reconstitution previously observed in LP/J recipient mice was due to a direct effect mediated by IL-21 on the BM compartment, hematopoietic stem cells (HSCs) were analyzed in recipient mice 4 weeks following transplantation using well-defined cell surface markers [59]. Aside from the significant increase in total BM cell number (Fig. 2a), LinSca1+c-kit+ (LSK) counts in IL-21-treated animals were highly comparable to those in unirradiated control mice (Fig. 2b). This observed increase in LSKs was not due to long-term (LT)-HSC expansion (Fig. 2c), but rather to augmented numbers of short-term (ST)-HSCs (Fig. 2d) and multipotent progenitors (MPPs) (Fig. 2e). In fact, the effect of IL-21 on LSKs is not surprising as these BM subsets express the IL-21R (Fig. 2f). To further support our in vivo observations, we supplemented freshly plated BM cells with ascending doses of IL-21 (0–1000 ng/ml) and noticed a dose-depending increase in LSK frequency with an optimal effect reached at 25–50 ng/ml (Fig. 2g, h). Further validations by intracellular Ki-67 staining conducted on WT- versus IL-21R−/−-derived BM treated with IL-21 (50 ng/ml) revealed a twofold increase in LSK expansion over basal levels (Additional file 1: Figure S1; Fig. 2i, j). The sum of these results clearly links accelerated lymphoid recovery mediated by IL-21 to the proliferation of BM-resident ST-HSCs and MPPs, which represent the hub of thymus-settling progenitors [49, 5155].

Thymopoiesis stimulation by IL-21 re-establishes a peripheral naïve T cell pool

The enhanced recovery of ST-HSCs and MPPs post-BMT is indicative of improved engraftment and certainly accounts for the increased peripheral T cell development observed in IL-21-treated animals [6062]. However, IL-21R was also reported to be expressed on thymic progenitors at the DN and DP stages [35, 36]. This suggests that IL-21 can directly mediate thymocyte proliferation in situ. To confirm this assumption, thymic tissues were resected from transplanted mice 4 weeks post-BMT and analyzed for both the hematopoietic (CD45+) and non-hematopoietic (CD45) compartments (Fig. 3a). Interestingly, thymi derived from the IL-21-treated group were comparable in size to those from unirradiated control mice as opposed to PBS-treated animals, which remained atrophied (Fig. 3b). Thymocyte analysis by flow cytometry clearly shows normal GFP frequency levels in IL-21-treated animals (Fig. 3c) with full recovery of both total and GFP+ thymocytes (Fig. 3d). All major thymic subsets (Fig. 3e) including early thymic progenitors (ETPs; LinCD8αCD25c-kit+), DN2 (linCD4CD8CD25+CD44+), and DN3 (linCD4CD8CD25+CD44) thymocytes (Fig. 3f) recovered in response to IL-21 treatment. Finally, analysis of enzymatically digested thymic tissues revealed significant improvement in total (CD45EpCam+), cortical (CD45EpCam+UEA1Ly51+), and medullary (CD45EpCam+UEA1+Ly51) thymic epithelial cells (TECs) in the IL-21-treated group when compared to PBS control mice (Fig. 3g).
The observed T cell development triggered by IL-21 is expected to qualitatively change the content of secondary lymphoid organs due to enhanced RTE output (Fig. 1d, e). Although all spleens derived from LP/J transplanted animals looked similar in size to those from unirradiated control mice (Fig. 4a), higher GFP frequency (Fig. 4b) and counts of total as well as GFP+ splenocytes were detected in the IL-21 group (Fig. 4c). Not only did IL-21 administration culminate in full reconstitution of CD4+, CD8+, and CD19+ cells (Fig. 4d), but the majority of developed CD4+ and CD8+ T cells displayed a naïve phenotype (CD62LhiCD44lo) (Fig. 4e) with a normal TCRvβ distribution (Fig. 4f). To determine whether IL-21 administration to transplanted mice affects the nature of newly developed T lymphocytes, we next examined their functionality in response to TCR stimulation. No significant changes could be detected in their proliferative capacity (Additional file 1: Figure S2A) or ability to secrete a wide range of cytokines (Additional file 1: Figure S2B). Furthermore, all stimulated T cells expressed similar levels of granzyme B (Additional file 1: Figure S2C, D) with no depicted differences in their expression of Bcl6, Nfat, T-bet, Gata3, Rorγt, and Foxp3 transcripts (Additional file 1: Figure S3). Together, our results suggest that IL-21 administration post-BMT stimulates thymopoiesis efficiently consequently triggering the emergence of peripheral naïve T cell pool displaying normal functionality.

IL-21 drives regulatory B10 cell expansion in transplanted animals

IL-21 can exert actions on multiple lymphoid and myeloid populations with a potential role in the modulation/development of immune regulatory cells [63]. For instance, IL-21−/− mice display high numbers of FOXP3-expressing Tregs, suggesting a function for IL-21 in blocking Treg development [28]. Elegant mechanistic studies further demonstrated that IL-21 impedes Treg generation and viability in vitro by inhibiting IL-2 production [64]. Besides Tregs, IL-21 supports B cell development in the BM, suggesting a possible parallel effect on regulatory B cells (B10) [38]. In support of this notion, activation and proliferation of B10 cells were reported to be dependent on both cognate interactions with CD4+ T cells and IL-21 during an inflammatory process [38]. Based on all these facts, we next scrutinized whether IL-21 administration affects the development of Tregs in mice undergoing de novo hematopoiesis and found a significant but trivial increase in CD4+CD25+FOXP3+ T cell number (Fig. 4g). However, the same IL-21 group displayed a pronounced increase in regulatory B cells based on the quantification of IL-10-competent CD1dhiCD5+ B cells (20% as opposed to 1–4% in control PBS-treated animals—Additional file 1: Figure S4; Fig. 4h). Therefore, IL-21 administration post-BMT enhances significantly the level of regulatory B10 cells along with a limited effect on Treg development.

IL-21 protects transplanted mice from GVHD while preserving the GVT effect

So far, all BMT experiments were conducted using T cell-depleted BM to assess the thymus-boosting properties of IL-21 in the absence of GVHD-induced damages. To evaluate the effect of IL-21 on GVHD, we next supplemented the BM graft with mature RAG2p-GFP-derived allogeneic T cells (donor lymphocyte infusion (DLI)) and found that PBS-treated animals became ill as of the 3rd week post-BMT with a 50% survival rate (Fig. 5a) according to weight loss assessment (Fig. 5b). In sharp contrast, none of the IL-21-treated animals developed GVHD signs (Fig. 5a, b). As the proportion of regulatory B cells was enhanced in IL-21-treated animals (Fig. 4h), we next addressed whether these Bregs were behind the GVHD protection observed in transplanted LP/J mice. To test this hypothesis, T cell-depleted BM derived from MμMT mice (deficient in B cell development) was supplemented with mature T cells from RAG2p-GFP mice, then transplanted into LP/J animals followed by PBS or IL-21 treatments. Interestingly, 40% survival was obtained in the IL-21-treated group whereas all PBS control mice died by the 3rd week post-BMT (Fig. 5c, d). As these observations clearly indicate a role for Bregs in mediating GVHD protection, we next tested whether IL-21-enhanced Breg development would lead to similar outcome in a complete MHC-mismatch BMT model (C57BL/6 → BALB/c). Under this context, all BALB/c transplanted mice died within the 3rd week post-BMT with no observed differences between PBS and IL-21 groups (Fig. 5e).
GVHD induction is usually concomitant with GVT [65]. We therefore corroborated impaired GVT effect in IL-21-treated animals due to the absence of GVHD signs. To validate this hypothesis, LP/J recipient mice were transplanted with RAG2p-GFP-derived BM with or without DLI followed by a challenge the following day using the mastocytoma cell line P815 (DBA/2-derived). As predicted, all PBS-treated mice transplanted with T cell-depleted BM died around the 4th week (Fig. 5f; green line) whereas DLI led to 60% survival up to 12 weeks post-BMT (Fig. 5f; blue line). Surprisingly, 90% of challenged IL-21-treated mice transplanted with T cell-depleted BM survived (Fig. 5f; black line), whereas complete protection was obtained if animals underwent DLI (Fig. 5f; red line). It should be noted however that IL-21 can trigger apoptosis of IL-21R-expressing lymphomas [66, 67]. To confirm that the observed GVT effect is solely dependent on the anti-tumoral effect mediated by the newly developed thymus-selected T cells as opposed to a direct apoptosis-inducing effect mediated by IL-21 itself, we assessed the expression of the IL-21Rα chain on P815 by flow cytometry and found that it was indeed negative (Fig. 5f; histogram figure). We concluded therefore that IL-21 can bypass GVHD induction, at least in the MHC-matched allogeneic graft setting, while retaining beneficial GVT effects mediated by the newly generated T cells.

IL-21 triggers T cell development in humanized mice

To test whether human IL-21 displays a thymopoiesis-stimulating effect akin to its murine ortholog, we next transplanted immunodeficient NSG mice with T cell-depleted CB units. Two doses of human IL-21 were tested (50 and 100 μg/kg) versus IL-7 (50 μg/kg) or equal volume of PBS following the same injection schedule previously used for LP/J mice. Peripheral blood analysis over 16 weeks revealed a significant increase in both human CD45+ and CD3+ cells (Fig. 6a) only in the group receiving IL-21 at a dose of 50 μg/kg. The same IL-21-treated NSG group displayed 90% chimerism with over 30% circulating human CD3+ T cells (Fig. 6b). Besides the absence of a beneficial effect for IL-7, the high IL-21 dose (100 μg/kg) failed at improving human CD3+ T cell development (Fig. 6a), which could be explained by a blockade at the DN stage in the thymi of NSG transplanted mice (Fig. 6c). Detailed thymic analysis of the 50 μg/kg group showed pronounced human thymocyte counts (total and human CD45+ cells) with increased levels of DN, DP, and SP thymocytes (Fig. 6d). Their spleens contained higher counts of total and human CD45+ cells as well as significantly increased levels of CD3+, CD4+, CD8+, and CD19+ SP lymphocytes with no apparent differences in CD13+ (granulocytes) or CD161+ (NK/NKT) counts (Fig. 6e). Altogether, these observations clearly indicate that human IL-21 is equivalent to its murine ortholog in stimulating the development of MHC-restricted human CD3+ T cells in immunodeficient NSG mice.

Discussion

The thymus is a specialized microenvironment dedicated to the generation of self-tolerant T cells with a broad TCR repertoire [6870]. This is vital to ensure the development of adaptive immune responses without leading to autoimmune diseases. The importance of the thymus, however, must be reconciled with the fact that it is exquisitely sensitive to a range of external insults including those derived from the conditioning regimen used prior to BMT [71, 72]. Although pediatric patients can recover their thymic function post-transplantation, the latter remains blunted in aged individuals leading consequently to diverse clinical complications [6]. This explains why accelerated T cell recovery correlates with improved survival post-BMT [1113, 16, 20, 24]. For de novo generation of thymus-derived naïve T cells to occur, three conditions must be fulfilled following BMT: (i) efficient HSC engraftment, (ii) active thymic seeding by BM-derived progenitor cells, and (iii) a thymic architecture conducive to progenitor migration/differentiation. Using a series of BMT experiments, we have shown that IL-21 administration to transplanted mice enhances de novo thymopoiesis by targeting both primary lymphoid tissues: the BM and the thymus (Fig. 7). More specifically, IL-21 promotes the expansion of ST-HSCs and MPPs within the BM-resident LSK population, which represents the source of building blocks required for rapid immune recovery [49, 5155]. Although not directly assessed in our studies, we presume that such increase in the pool of BM progenitors enhances thymic seeding [60, 73]. In parallel, we demonstrated that IL-21 stimulates intrathymic T cell development directly by boosting the count of thymic progenitor cells while enhancing TEC recovery. The indirect effect of IL-21 on TECs is particularly important as these stromal cells guide the migration of BM-resident progenitors via the production of chemotactic gradients and mediate positive/negative selection of developing thymocytes [74]. Consistent with previous reports, our working model also stipulates the likely possibility that newly generated thymus-selected RTEs compete with peripheral donor-derived alloreactive T cells via a process we refer to as “homeostatic pressure” diminishing therefore the recipient’s susceptibility to GVHD while ensuring proper GVT effect [75, 76]. Our observations also hint to a possible role for IL-21 in inhibiting GVHD onset through the development, activation, and/or proliferation of B10 cells.
Thymopoiesis is a complex multilayered process with various factors at play [74]. Of all cytokines involved in T cell biology, IL-7 remains the central growth factor due to its role in the development, survival, and homeostatic proliferation of T cells [48]. Based on these properties, it was extensively studied in the context of BMT where it was shown to exert powerful thymopoietic-stimulating effects [7789]. By directly comparing the pharmacological potential of IL-7 and IL-21 using the optimal IL-21 dose (50 μg/kg) previously identified, we found no significant increase in RTE output triggered by IL-7 administration to LP/J recipient mice (Fig. 1b–d). This is not surprising as IL-7 supports thymopoiesis when administered at doses ranging from 250 to 500 μg/kg (e.g., five- to tenfold higher than the dose used in our BMT experiments) [79, 84, 87]. It must be noted, however, that IL-21 cannot support the development of SP thymocytes in the absence of IL-7 signaling (Fig. 1g). This is consistent with our previous in vitro observations where IL-21 was found to be inert at driving T cell differentiation due to its inability to trigger ERK phosphorylation in TCR-engaged DP thymocytes [35]. Thus, T cell reconstitution could be enhanced/accelerated by IL-21 via the induction of BM/thymic progenitor expansion but relies exclusively on IL-7 signaling to exert its pharmacological effect.
One of the most seminal observations made in the herein study was the absence of GVHD signs in IL-21-treated animals while the GVT effect was retained. This is counterintuitive as GVT is usually associated with GVHD induction [65]. Furthermore, the IL-21/IL-21R signaling axis was previously reported to exacerbate GVHD in a CD4-dependent manner involving B cell expansion and production of autoantibodies [73, 90, 91]. Our study puts at play two important factors, which may help depict the discrepancy between our observations and those reported by others. First, GVHD and GVT are usually mediated by allogeneic T cells (e.g., DLI) transferred along or after BMT [65]. The purpose of DLI is to provide transient protection against infectious disease while ensuring eradication of post-conditioning regimen-resistant cancer cells [59]. As T cell development is severely delayed following allogeneic BMT, donor-derived alloreactive T cells can expand in lymphopenic recipients consequently increasing recipients’ susceptibility to GVHD [75, 76, 9294]. Under these circumstances, the endogenous production of any pro-inflammatory cytokine(s), including IL-21, during GVHD onset would exacerbate the magnitude of the disease [75, 76, 92, 93]. In this regard, the accelerated appearance of thymus-selected RTEs triggered by exogenous IL-21 administration would not only compete with peripheral alloreactive T cells to access secondary lymphoid organs, homeostatic cytokines, and self-MHC ligands impeding therefore alloreactive T cell expansion/activation, but it will also ensure efficient immune surveillance and tumor rejection as we have observed with the P815 challenge experiment conducted in IL-21-treated animals (Fig. 6). Second, GVHD could be alleviated through the concrete action of B10 cells, which were previously reported to be induced by IL-21 [38]. Interestingly, B10 cells are potent regulators of macrophages and dendritic cell function, which suggest that IL-21-induced B10 cells may contribute to GVHD inhibition by restraining monocyte and dendritic cell-mediated stimulation of alloreactive T cells [9599]. Although we did not directly investigate the Breg mechanism of action in our BMT models, the decreased survival rate observed in IL-21-treated LP/J mice transplanted with B cell-deficient BM supports a central role for B10 cells in alleviating GVHD (Fig. 5c, d). This prompts us to formulate the hypothesis that IL-21-mediated induction of B10 cells would be beneficial for controlling GVHD in a complete MHC-mismatch model (C57BL/6 → BALB/c). Since the complete loss of all transplanted IL-21-treated BALB/c animals occurred within the first 3 weeks post-BMT, one can argue that the strong alloreactive response mediated by allogeneic T cells overrides the B10 suppressive effect. Furthermore, potent production of interferon-gamma triggered by strong alloreactivity may counterbalance B10 cell expansion in vivo [38]. Besides, the observed alloreactivity in the complete MHC-mismatch BMT model occurred faster (less than 3 weeks) than the time required for de novo B (~3–4 weeks) and T cell development (~5 weeks) previously observed in LP/J recipient mice. As for the discrepancy between our observations and the exacerbated xeno-GVHD onset observed by Wu et al. in NSG mice, two central points must be discussed: the source of DLI and the bioavailability of human IL-21. In the study by Wu et al., peripheral blood mononuclear cells containing mature T cells were infused instead of T cell-depleted CB. In this context, mature B and T cells would certainly promote a superior and faster xenoreactivity in NSG mice leading consequently to xeno-GVHD. Second, human IL-21 was undetectable in peripheral blood of hydrodynamically transfected NSG mice beyond 5 days. As we administered human IL-21 over 2 weeks, we presume based on our LP/J observations that our approach ensures higher de novo T cell development with the potential induction of IL-21-induced human B10 cells, thus significantly improving NSG survival. Although evidence suggests that both positive and negative selection can occur in NSG mice [40, 100], we cannot preclude the possibility of xeno-GVHD development in NSG mice beyond 16 weeks post-transplantation. Additional mechanistic studies are therefore warranted to further demystify IL-21’s mode of action in NSG mice.

Conclusions

In summary, our study clearly establishes a role for IL-21 in immune recovery following BMT, where it acts as a regulator of BM progenitor expansion as well as on the earliest phases of intrathymic T cell development and recovery of the stromal compartment. Its ability to simultaneously affect the BM and thymic compartments while repressing alloreactive T cells gives it an advantageous edge over currently tested therapies. Although our studies suggest that IL-21 could be exploited as an immunotherapeutic agent for the restoration of essential immune system functioning following ablative therapy, its impact on inducing/promoting autoimmune diseases in BMT patients remains elusive. Therefore, future pre-clinical and clinical trials should foresee the use of human IL-21 to determine its safety and efficacy in facilitating immune recovery post-BMT.

Acknowledgements

We are grateful to the Institute for Research in Immunology and Cancer flow cytometry, genomics, and animal care facility staff for their assistance.

Funding

This work was supported by the Canadian Cancer Society (Innovation Grant #2013-701623; the Louisa Gale Scholar), the Cole Foundation, the Cancer Research Society (OG2016-#21012; for the human IL-21 work), and the Benoit Gagnon Research Fund. Aurélie Tormo holds a Cole Foundation Post-Doctoral Fellowship. Moutih Rafei holds a Fonds de la Recherche en Santé du Québec Junior II Award.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.

Authors’ contributions

AT designed most of the study, carried out most of the experiments, analyzed the data, and prepared the following figures: 1, 2a–e, 3, 4, 5a, b, e, and 6b–e. FK, YC, EAC, RK, BN, and HW assisted with some experiments and contributed to the data analysis and manuscript preparation. MR designed the study, discussed the results with all authors, and wrote the manuscript. All authors approved this manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Animal protocols were approved by the Animal Care Committee of Université de Montréal. Ethics approval for the use of human CB was granted from the Ste-Justine human ethical board.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Additional file

Additional file 1: Figure S1. Gating strategies for LSKs. BM cells collected from WT or IL-21R−/− C57BL/6 mice were treated in vitro for proliferation, then stained with the LinSca1+c-kit+ antibody cocktail prior to Ki-67+ incorporation analysis. All described experiments were conducted at least three times with n = 5/group. Figure S2. Functional characterization of T cells. (A) Representative cell trace dilution analysis on CD4+ or CD8+ T cells derived from ctl (unirradiated), PBS-treated, or IL-21-treated LP/J recipient mice. (B) Cytokine quantification by ELISA from T cells derived from the same groups described in panel (A). (C) Representative flow cytometry analysis of granzyme B expression. (D) Quantification of T cells expressing granzyme B. For all presented studies, T cells were stimulated with CD3-CD28 dynabeads for 48 h prior to analyses. All described experiments were conducted at least three times with an n = 5/group. Figure S3. Molecular characterization of T cells. T cells sorted from ctl, PBS-treated, or IL-21-treated LP/J recipient mice were analyzed for their expression of various transcription factors involved in T cell differentiation. All described experiments were conducted at least three times with n = 5/group. Figure S4. Gating strategies for Breg analysis. For detection of IL-10-producing Bregs, CD19+ B cells were first isolated from spleens of treated mice (isotype shown by the filled gray histogram), then stained after in vitro treatment with CD1d and CD5 antibodies. The B cell subset CD1dhiCD5+ was gated prior to IL-10 assessment by intracellular staining. All described experiments were conducted at least three times with n = 5/group. (PDF 2111 kb)
Literatur
1.
Zurück zum Zitat Wils EJ, Cornelissen JJ. Thymopoiesis following allogeneic stem cell transplantation: new possibilities for improvement. Blood Rev. 2005;19:89–98.CrossRefPubMed Wils EJ, Cornelissen JJ. Thymopoiesis following allogeneic stem cell transplantation: new possibilities for improvement. Blood Rev. 2005;19:89–98.CrossRefPubMed
2.
Zurück zum Zitat Williams KM, Gress RE. Immune reconstitution and implications for immunotherapy following haematopoietic stem cell transplantation. Best Pract Res Clin Haematol. 2008;21:579–96.CrossRefPubMedPubMedCentral Williams KM, Gress RE. Immune reconstitution and implications for immunotherapy following haematopoietic stem cell transplantation. Best Pract Res Clin Haematol. 2008;21:579–96.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Porter DL, June CH. T-cell reconstitution and expansion after hematopoietic stem cell transplantation: ‘T’ it up! Bone Marrow Transplant. 2005;35:935–42.CrossRefPubMed Porter DL, June CH. T-cell reconstitution and expansion after hematopoietic stem cell transplantation: ‘T’ it up! Bone Marrow Transplant. 2005;35:935–42.CrossRefPubMed
4.
Zurück zum Zitat Novitzky N, Davison GM. Immune reconstitution following hematopoietic stem-cell transplantation. Cytotherapy. 2001;3:211–20.CrossRefPubMed Novitzky N, Davison GM. Immune reconstitution following hematopoietic stem-cell transplantation. Cytotherapy. 2001;3:211–20.CrossRefPubMed
5.
Zurück zum Zitat Geddes M, Storek J. Immune reconstitution following hematopoietic stem-cell transplantation. Best Pract Res Clin Haematol. 2007;20:329–48.CrossRefPubMed Geddes M, Storek J. Immune reconstitution following hematopoietic stem-cell transplantation. Best Pract Res Clin Haematol. 2007;20:329–48.CrossRefPubMed
6.
Zurück zum Zitat Dudakov JA, van den Brink MR. Greater than the sum of their parts: combination strategies for immune regeneration following allogeneic hematopoietic stem cell transplantation. Best Pract Res Clin Haematol. 2011;24:467–76.CrossRefPubMedPubMedCentral Dudakov JA, van den Brink MR. Greater than the sum of their parts: combination strategies for immune regeneration following allogeneic hematopoietic stem cell transplantation. Best Pract Res Clin Haematol. 2011;24:467–76.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Chidgey AP, Seach N, Dudakov J, Hammett MV, Boyd RL. Strategies for reconstituting and boosting T cell-based immunity following haematopoietic stem cell transplantation: pre-clinical and clinical approaches. Semin Immunopathol. 2008;30:457–77.CrossRefPubMed Chidgey AP, Seach N, Dudakov J, Hammett MV, Boyd RL. Strategies for reconstituting and boosting T cell-based immunity following haematopoietic stem cell transplantation: pre-clinical and clinical approaches. Semin Immunopathol. 2008;30:457–77.CrossRefPubMed
8.
Zurück zum Zitat Petersen SL, Ryder LP, Bjork P, Madsen HO, Heilmann C, Jacobsen N, Sengelov H, Vindelov LL. A comparison of T-, B- and NK-cell reconstitution following conventional or nonmyeloablative conditioning and transplantation with bone marrow or peripheral blood stem cells from human leucocyte antigen identical sibling donors. Bone Marrow Transplant. 2003;32:65–72.CrossRefPubMed Petersen SL, Ryder LP, Bjork P, Madsen HO, Heilmann C, Jacobsen N, Sengelov H, Vindelov LL. A comparison of T-, B- and NK-cell reconstitution following conventional or nonmyeloablative conditioning and transplantation with bone marrow or peripheral blood stem cells from human leucocyte antigen identical sibling donors. Bone Marrow Transplant. 2003;32:65–72.CrossRefPubMed
9.
Zurück zum Zitat Maris M, Boeckh M, Storer B, Dawson M, White K, Keng M, Sandmaier B, Maloney D, Storb R, Storek J. Immunologic recovery after hematopoietic cell transplantation with nonmyeloablative conditioning. Exp Hematol. 2003;31:941–52.CrossRefPubMed Maris M, Boeckh M, Storer B, Dawson M, White K, Keng M, Sandmaier B, Maloney D, Storb R, Storek J. Immunologic recovery after hematopoietic cell transplantation with nonmyeloablative conditioning. Exp Hematol. 2003;31:941–52.CrossRefPubMed
10.
Zurück zum Zitat Kalwak K, Gorczynska E, Toporski J, Turkiewicz D, Slociak M, Ussowicz M, Latos-Grazynska E, Krol M, Boguslawska-Jaworska J, Chybicka A. Immune reconstitution after haematopoietic cell transplantation in children: immunophenotype analysis with regard to factors affecting the speed of recovery. Br J Haematol. 2002;118:74–89.CrossRefPubMed Kalwak K, Gorczynska E, Toporski J, Turkiewicz D, Slociak M, Ussowicz M, Latos-Grazynska E, Krol M, Boguslawska-Jaworska J, Chybicka A. Immune reconstitution after haematopoietic cell transplantation in children: immunophenotype analysis with regard to factors affecting the speed of recovery. Br J Haematol. 2002;118:74–89.CrossRefPubMed
11.
Zurück zum Zitat Porrata LF, Litzow MR, Tefferi A, Letendre L, Kumar S, Geyer SM, Markovic SN. Early lymphocyte recovery is a predictive factor for prolonged survival after autologous hematopoietic stem cell transplantation for acute myelogenous leukemia. Leukemia. 2002;16:1311–8.CrossRefPubMed Porrata LF, Litzow MR, Tefferi A, Letendre L, Kumar S, Geyer SM, Markovic SN. Early lymphocyte recovery is a predictive factor for prolonged survival after autologous hematopoietic stem cell transplantation for acute myelogenous leukemia. Leukemia. 2002;16:1311–8.CrossRefPubMed
12.
Zurück zum Zitat Porrata LF, Ingle JN, Litzow MR, Geyer S, Markovic SN. Prolonged survival associated with early lymphocyte recovery after autologous hematopoietic stem cell transplantation for patients with metastatic breast cancer. Bone Marrow Transplant. 2001;28:865–71.CrossRefPubMed Porrata LF, Ingle JN, Litzow MR, Geyer S, Markovic SN. Prolonged survival associated with early lymphocyte recovery after autologous hematopoietic stem cell transplantation for patients with metastatic breast cancer. Bone Marrow Transplant. 2001;28:865–71.CrossRefPubMed
13.
Zurück zum Zitat Porrata LF, Gertz MA, Inwards DJ, Litzow MR, Lacy MQ, Tefferi A, Gastineau DA, Dispenzieri A, Ansell SM, Micallef IN, et al. Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood. 2001;98:579–85.CrossRefPubMed Porrata LF, Gertz MA, Inwards DJ, Litzow MR, Lacy MQ, Tefferi A, Gastineau DA, Dispenzieri A, Ansell SM, Micallef IN, et al. Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood. 2001;98:579–85.CrossRefPubMed
14.
Zurück zum Zitat Lucas KG, Small TN, Heller G, Dupont B, O’Reilly RJ. The development of cellular immunity to Epstein-Barr virus after allogeneic bone marrow transplantation. Blood. 1996;87:2594–603.PubMed Lucas KG, Small TN, Heller G, Dupont B, O’Reilly RJ. The development of cellular immunity to Epstein-Barr virus after allogeneic bone marrow transplantation. Blood. 1996;87:2594–603.PubMed
15.
Zurück zum Zitat Parkman R, Cohen G, Carter SL, Weinberg KI, Masinsin B, Guinan E, Kurtzberg J, Wagner JE, Kernan NA. Successful immune reconstitution decreases leukemic relapse and improves survival in recipients of unrelated cord blood transplantation. Biol Blood Marrow Transplant. 2006;12:919–27.CrossRefPubMed Parkman R, Cohen G, Carter SL, Weinberg KI, Masinsin B, Guinan E, Kurtzberg J, Wagner JE, Kernan NA. Successful immune reconstitution decreases leukemic relapse and improves survival in recipients of unrelated cord blood transplantation. Biol Blood Marrow Transplant. 2006;12:919–27.CrossRefPubMed
16.
Zurück zum Zitat Porrata LF, Inwards DJ, Micallef IN, Ansell SM, Geyer SM, Markovic SN. Early lymphocyte recovery post-autologous haematopoietic stem cell transplantation is associated with better survival in Hodgkin’s disease. Br J Haematol. 2002;117:629–33.CrossRefPubMed Porrata LF, Inwards DJ, Micallef IN, Ansell SM, Geyer SM, Markovic SN. Early lymphocyte recovery post-autologous haematopoietic stem cell transplantation is associated with better survival in Hodgkin’s disease. Br J Haematol. 2002;117:629–33.CrossRefPubMed
17.
Zurück zum Zitat Savani BN, Rezvani K, Mielke S, Montero A, Kurlander R, Carter CS, Leitman S, Read EJ, Childs R, Barrett AJ. Factors associated with early molecular remission after T cell-depleted allogeneic stem cell transplantation for chronic myelogenous leukemia. Blood. 2006;107:1688–95.CrossRefPubMedPubMedCentral Savani BN, Rezvani K, Mielke S, Montero A, Kurlander R, Carter CS, Leitman S, Read EJ, Childs R, Barrett AJ. Factors associated with early molecular remission after T cell-depleted allogeneic stem cell transplantation for chronic myelogenous leukemia. Blood. 2006;107:1688–95.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Krause H, Hebart H, Jahn G, Muller CA, Einsele H. Screening for CMV-specific T cell proliferation to identify patients at risk of developing late onset CMV disease. Bone Marrow Transplant. 1997;19:1111–6.CrossRefPubMed Krause H, Hebart H, Jahn G, Muller CA, Einsele H. Screening for CMV-specific T cell proliferation to identify patients at risk of developing late onset CMV disease. Bone Marrow Transplant. 1997;19:1111–6.CrossRefPubMed
19.
Zurück zum Zitat Storek J, Espino G, Dawson MA, Storer B, Flowers ME, Maloney DG. Low B-cell and monocyte counts on day 80 are associated with high infection rates between days 100 and 365 after allogeneic marrow transplantation. Blood. 2000;96:3290–3.PubMed Storek J, Espino G, Dawson MA, Storer B, Flowers ME, Maloney DG. Low B-cell and monocyte counts on day 80 are associated with high infection rates between days 100 and 365 after allogeneic marrow transplantation. Blood. 2000;96:3290–3.PubMed
20.
Zurück zum Zitat Storek J, Gooley T, Witherspoon RP, Sullivan KM, Storb R. Infectious morbidity in long-term survivors of allogeneic marrow transplantation is associated with low CD4 T cell counts. Am J Hematol. 1997;54:131–8.CrossRefPubMed Storek J, Gooley T, Witherspoon RP, Sullivan KM, Storb R. Infectious morbidity in long-term survivors of allogeneic marrow transplantation is associated with low CD4 T cell counts. Am J Hematol. 1997;54:131–8.CrossRefPubMed
22.
Zurück zum Zitat van den Brink MR, Alpdogan O, Boyd RL. Strategies to enhance T-cell reconstitution in immunocompromised patients. Nat Rev Immunol. 2004;4:856–67.CrossRefPubMed van den Brink MR, Alpdogan O, Boyd RL. Strategies to enhance T-cell reconstitution in immunocompromised patients. Nat Rev Immunol. 2004;4:856–67.CrossRefPubMed
23.
Zurück zum Zitat Joao C, Porrata LF, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Gastineau DA, Markovic SN. Early lymphocyte recovery after autologous stem cell transplantation predicts superior survival in mantle-cell lymphoma. Bone Marrow Transplant. 2006;37:865–71.CrossRefPubMed Joao C, Porrata LF, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Gastineau DA, Markovic SN. Early lymphocyte recovery after autologous stem cell transplantation predicts superior survival in mantle-cell lymphoma. Bone Marrow Transplant. 2006;37:865–71.CrossRefPubMed
24.
Zurück zum Zitat Porrata LF, Ristow K, Witzig TE, Tuinistra N, Habermann TM, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Markovic SN. Absolute lymphocyte count predicts therapeutic efficacy and survival at the time of radioimmunotherapy in patients with relapsed follicular lymphomas. Leukemia. 2007;21:2554–6.CrossRefPubMed Porrata LF, Ristow K, Witzig TE, Tuinistra N, Habermann TM, Inwards DJ, Ansell SM, Micallef IN, Johnston PB, Markovic SN. Absolute lymphocyte count predicts therapeutic efficacy and survival at the time of radioimmunotherapy in patients with relapsed follicular lymphomas. Leukemia. 2007;21:2554–6.CrossRefPubMed
25.
Zurück zum Zitat Mangel J, Leitch HA, Connors JM, Buckstein R, Imrie K, Spaner D, Crump M, Pennell N, Boudreau A, Berinstein NL. Intensive chemotherapy and autologous stem-cell transplantation plus rituximab is superior to conventional chemotherapy for newly diagnosed advanced stage mantle-cell lymphoma: a matched pair analysis. Ann Oncol. 2004;15:283–90.CrossRefPubMed Mangel J, Leitch HA, Connors JM, Buckstein R, Imrie K, Spaner D, Crump M, Pennell N, Boudreau A, Berinstein NL. Intensive chemotherapy and autologous stem-cell transplantation plus rituximab is superior to conventional chemotherapy for newly diagnosed advanced stage mantle-cell lymphoma: a matched pair analysis. Ann Oncol. 2004;15:283–90.CrossRefPubMed
26.
Zurück zum Zitat Brugger W, Hirsch J, Grunebach F, Repp R, Brossart P, Vogel W, Kopp HG, Manz MG, Bitzer M, Schlimok G, et al. Rituximab consolidation after high-dose chemotherapy and autologous blood stem cell transplantation in follicular and mantle cell lymphoma: a prospective, multicenter phase II study. Ann Oncol. 2004;15:1691–8.CrossRefPubMed Brugger W, Hirsch J, Grunebach F, Repp R, Brossart P, Vogel W, Kopp HG, Manz MG, Bitzer M, Schlimok G, et al. Rituximab consolidation after high-dose chemotherapy and autologous blood stem cell transplantation in follicular and mantle cell lymphoma: a prospective, multicenter phase II study. Ann Oncol. 2004;15:1691–8.CrossRefPubMed
27.
Zurück zum Zitat Spolski R, Leonard WJ. Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008;26:57–79.CrossRefPubMed Spolski R, Leonard WJ. Interleukin-21: basic biology and implications for cancer and autoimmunity. Annu Rev Immunol. 2008;26:57–79.CrossRefPubMed
28.
Zurück zum Zitat Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, Oukka M, Kuchroo VK. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature. 2007;448:484–7.CrossRefPubMedPubMedCentral Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, Oukka M, Kuchroo VK. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature. 2007;448:484–7.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Vosshenrich CA, Ranson T, Samson SI, Corcuff E, Colucci F, Rosmaraki EE, Di Santo JP. Roles for common cytokine receptor gamma-chain-dependent cytokines in the generation, differentiation, and maturation of NK cell precursors and peripheral NK cells in vivo. J Immunol. 2005;174:1213–21.CrossRefPubMed Vosshenrich CA, Ranson T, Samson SI, Corcuff E, Colucci F, Rosmaraki EE, Di Santo JP. Roles for common cytokine receptor gamma-chain-dependent cytokines in the generation, differentiation, and maturation of NK cell precursors and peripheral NK cells in vivo. J Immunol. 2005;174:1213–21.CrossRefPubMed
30.
Zurück zum Zitat Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. J Immunol. 2005;175:2261–9.CrossRefPubMed Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the antigen-specific CD8 T cell response. J Immunol. 2005;175:2261–9.CrossRefPubMed
31.
Zurück zum Zitat Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, Schluns K, Tian Q, Watowich SS, Jetten AM, Dong C. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. 2007;448:480–3.CrossRefPubMed Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, Schluns K, Tian Q, Watowich SS, Jetten AM, Dong C. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. 2007;448:480–3.CrossRefPubMed
32.
Zurück zum Zitat Ozaki K, Spolski R, Feng CG, Qi CF, Cheng J, Sher A, Morse 3rd HC, Liu C, Schwartzberg PL, Leonard WJ. A critical role for IL-21 in regulating immunoglobulin production. Science. 2002;298:1630–4.CrossRefPubMed Ozaki K, Spolski R, Feng CG, Qi CF, Cheng J, Sher A, Morse 3rd HC, Liu C, Schwartzberg PL, Leonard WJ. A critical role for IL-21 in regulating immunoglobulin production. Science. 2002;298:1630–4.CrossRefPubMed
33.
Zurück zum Zitat Ozaki K, Hishiya A, Hatanaka K, Nakajima H, Wang G, Hwu P, Kitamura T, Ozawa K, Leonard WJ, Nosaka T. Overexpression of interleukin 21 induces expansion of hematopoietic progenitor cells. Int J Hematol. 2006;84:224–30.CrossRefPubMed Ozaki K, Hishiya A, Hatanaka K, Nakajima H, Wang G, Hwu P, Kitamura T, Ozawa K, Leonard WJ, Nosaka T. Overexpression of interleukin 21 induces expansion of hematopoietic progenitor cells. Int J Hematol. 2006;84:224–30.CrossRefPubMed
34.
Zurück zum Zitat Simard N, Konforte D, Tran AH, Esufali J, Leonard WJ, Paige CJ. Analysis of the role of IL-21 in development of murine B cell progenitors in the bone marrow. J Immunol. 2011;186:5244–53.CrossRefPubMedPubMedCentral Simard N, Konforte D, Tran AH, Esufali J, Leonard WJ, Paige CJ. Analysis of the role of IL-21 in development of murine B cell progenitors in the bone marrow. J Immunol. 2011;186:5244–53.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Rafei M, Rouette A, Brochu S, Vanegas JR, Perreault C. Differential effects of gammac cytokines on postselection differentiation of CD8 thymocytes. Blood. 2013;121:107–17.CrossRefPubMed Rafei M, Rouette A, Brochu S, Vanegas JR, Perreault C. Differential effects of gammac cytokines on postselection differentiation of CD8 thymocytes. Blood. 2013;121:107–17.CrossRefPubMed
36.
Zurück zum Zitat Rafei M, Dumont-Lagace M, Rouette A, Perreault C. Interleukin-21 accelerates thymic recovery from glucocorticoid-induced atrophy. PLoS One. 2013;8:e72801.CrossRefPubMedPubMedCentral Rafei M, Dumont-Lagace M, Rouette A, Perreault C. Interleukin-21 accelerates thymic recovery from glucocorticoid-induced atrophy. PLoS One. 2013;8:e72801.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Al-Chami E, Tormo A, Pasquin S, Kanjarawi R, Ziouani S, Rafei M. Interleukin-21 administration to aged mice rejuvenates their peripheral T-cell pool by triggering de novo thymopoiesis. Aging Cell. 2016;15:349–60.CrossRefPubMedPubMedCentral Al-Chami E, Tormo A, Pasquin S, Kanjarawi R, Ziouani S, Rafei M. Interleukin-21 administration to aged mice rejuvenates their peripheral T-cell pool by triggering de novo thymopoiesis. Aging Cell. 2016;15:349–60.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Yoshizaki A, Miyagaki T, DiLillo DJ, Matsushita T, Horikawa M, Kountikov EI, Spolski R, Poe JC, Leonard WJ, Tedder TF. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature. 2012;491:264–8.CrossRefPubMedPubMedCentral Yoshizaki A, Miyagaki T, DiLillo DJ, Matsushita T, Horikawa M, Kountikov EI, Spolski R, Poe JC, Leonard WJ, Tedder TF. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature. 2012;491:264–8.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Tanaka S, Saito Y, Kunisawa J, Kurashima Y, Wake T, Suzuki N, Shultz LD, Kiyono H, Ishikawa F. Development of mature and functional human myeloid subsets in hematopoietic stem cell-engrafted NOD/SCID/IL2rgammaKO mice. J Immunol. 2012;188:6145–55.CrossRefPubMedPubMedCentral Tanaka S, Saito Y, Kunisawa J, Kurashima Y, Wake T, Suzuki N, Shultz LD, Kiyono H, Ishikawa F. Development of mature and functional human myeloid subsets in hematopoietic stem cell-engrafted NOD/SCID/IL2rgammaKO mice. J Immunol. 2012;188:6145–55.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, Kotb M, Gillies SD, King M, Mangada J, 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;174:6477–89.CrossRefPubMed Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, Kotb M, Gillies SD, King M, Mangada J, 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;174:6477–89.CrossRefPubMed
41.
Zurück zum Zitat Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol. 2007;7:118–30.CrossRefPubMed Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol. 2007;7:118–30.CrossRefPubMed
42.
Zurück zum Zitat Boursalian TE, Golob J, Soper DM, Cooper CJ, Fink PJ. Continued maturation of thymic emigrants in the periphery. Nat Immunol. 2004;5:418–25.CrossRefPubMed Boursalian TE, Golob J, Soper DM, Cooper CJ, Fink PJ. Continued maturation of thymic emigrants in the periphery. Nat Immunol. 2004;5:418–25.CrossRefPubMed
43.
Zurück zum Zitat Hazenberg MD, Verschuren MC, Hamann D, Miedema F, van Dongen JJ. T cell receptor excision circles as markers for recent thymic emigrants: basic aspects, technical approach, and guidelines for interpretation. J Mol Med (Berl). 2001;79:631–40.CrossRef Hazenberg MD, Verschuren MC, Hamann D, Miedema F, van Dongen JJ. T cell receptor excision circles as markers for recent thymic emigrants: basic aspects, technical approach, and guidelines for interpretation. J Mol Med (Berl). 2001;79:631–40.CrossRef
44.
Zurück zum Zitat Hazenberg MD, Otto SA, de Pauw ES, Roelofs H, Fibbe WE, Hamann D, Miedema F. T-cell receptor excision circle and T-cell dynamics after allogeneic stem cell transplantation are related to clinical events. Blood. 2002;99:3449–53.CrossRefPubMed Hazenberg MD, Otto SA, de Pauw ES, Roelofs H, Fibbe WE, Hamann D, Miedema F. T-cell receptor excision circle and T-cell dynamics after allogeneic stem cell transplantation are related to clinical events. Blood. 2002;99:3449–53.CrossRefPubMed
45.
Zurück zum Zitat Hazenberg MD, Borghans JA, de Boer RJ, Miedema F. Thymic output: a bad TREC record. Nat Immunol. 2003;4:97–9.CrossRefPubMed Hazenberg MD, Borghans JA, de Boer RJ, Miedema F. Thymic output: a bad TREC record. Nat Immunol. 2003;4:97–9.CrossRefPubMed
46.
Zurück zum Zitat Rafei M, Hardy MP, Williams P, Vanegas JR, Forner KA, Dulude G, Labrecque N, Galipeau J, Perreault C. Development and function of innate polyclonal TCRalphabeta+ CD8+ thymocytes. J Immunol. 2011;187:3133–44.CrossRefPubMed Rafei M, Hardy MP, Williams P, Vanegas JR, Forner KA, Dulude G, Labrecque N, Galipeau J, Perreault C. Development and function of innate polyclonal TCRalphabeta+ CD8+ thymocytes. J Immunol. 2011;187:3133–44.CrossRefPubMed
47.
Zurück zum Zitat Moran AE, Holzapfel KL, Xing Y, Cunningham NR, Maltzman JS, Punt J, Hogquist KA. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J Exp Med. 2011;208:1279–89.CrossRefPubMedPubMedCentral Moran AE, Holzapfel KL, Xing Y, Cunningham NR, Maltzman JS, Punt J, Hogquist KA. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J Exp Med. 2011;208:1279–89.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol. 2011;11:330–42.CrossRefPubMed Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol. 2011;11:330–42.CrossRefPubMed
49.
Zurück zum Zitat Zlotoff DA, Sambandam A, Logan TD, Bell JJ, Schwarz BA, Bhandoola A. CCR7 and CCR9 together recruit hematopoietic progenitors to the adult thymus. Blood. 2010;115:1897–905.CrossRefPubMedPubMedCentral Zlotoff DA, Sambandam A, Logan TD, Bell JJ, Schwarz BA, Bhandoola A. CCR7 and CCR9 together recruit hematopoietic progenitors to the adult thymus. Blood. 2010;115:1897–905.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Krueger A, Willenzon S, Lyszkiewicz M, Kremmer E, Forster R. CC chemokine receptor 7 and 9 double-deficient hematopoietic progenitors are severely impaired in seeding the adult thymus. Blood. 2010;115:1906–12.CrossRefPubMed Krueger A, Willenzon S, Lyszkiewicz M, Kremmer E, Forster R. CC chemokine receptor 7 and 9 double-deficient hematopoietic progenitors are severely impaired in seeding the adult thymus. Blood. 2010;115:1906–12.CrossRefPubMed
51.
Zurück zum Zitat Zlotoff DA, Zhang SL, De Obaldia ME, Hess PR, Todd SP, Logan TD, Bhandoola A. Delivery of progenitors to the thymus limits T-lineage reconstitution after bone marrow transplantation. Blood. 2011;118:1962–70.CrossRefPubMedPubMedCentral Zlotoff DA, Zhang SL, De Obaldia ME, Hess PR, Todd SP, Logan TD, Bhandoola A. Delivery of progenitors to the thymus limits T-lineage reconstitution after bone marrow transplantation. Blood. 2011;118:1962–70.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Zlotoff DA, Schwarz BA, Bhandoola A. The long road to the thymus: the generation, mobilization, and circulation of T-cell progenitors in mouse and man. Semin Immunopathol. 2008;30:371–82.CrossRefPubMed Zlotoff DA, Schwarz BA, Bhandoola A. The long road to the thymus: the generation, mobilization, and circulation of T-cell progenitors in mouse and man. Semin Immunopathol. 2008;30:371–82.CrossRefPubMed
54.
Zurück zum Zitat Zhang SL, Bhandoola A. Trafficking to the thymus. Curr Top Microbiol Immunol. 2014;373:87–111.PubMed Zhang SL, Bhandoola A. Trafficking to the thymus. Curr Top Microbiol Immunol. 2014;373:87–111.PubMed
56.
Zurück zum Zitat Chi AW, Chavez A, Xu L, Weber BN, Shestova O, Schaffer A, Wertheim G, Pear WS, Izon D, Bhandoola A. Identification of Flt3(+)CD150(-) myeloid progenitors in adult mouse bone marrow that harbor T lymphoid developmental potential. Blood. 2011;118:2723–32.CrossRefPubMedPubMedCentral Chi AW, Chavez A, Xu L, Weber BN, Shestova O, Schaffer A, Wertheim G, Pear WS, Izon D, Bhandoola A. Identification of Flt3(+)CD150(-) myeloid progenitors in adult mouse bone marrow that harbor T lymphoid developmental potential. Blood. 2011;118:2723–32.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Bhandoola A, von Boehmer H, Petrie HT, Zuniga-Pflucker JC. Commitment and developmental potential of extrathymic and intrathymic T cell precursors: plenty to choose from. Immunity. 2007;26:678–89.CrossRefPubMed Bhandoola A, von Boehmer H, Petrie HT, Zuniga-Pflucker JC. Commitment and developmental potential of extrathymic and intrathymic T cell precursors: plenty to choose from. Immunity. 2007;26:678–89.CrossRefPubMed
58.
Zurück zum Zitat Bell JJ, Bhandoola A. The earliest thymic progenitors for T cells possess myeloid lineage potential. Nature. 2008;452:764–7.CrossRefPubMed Bell JJ, Bhandoola A. The earliest thymic progenitors for T cells possess myeloid lineage potential. Nature. 2008;452:764–7.CrossRefPubMed
59.
Zurück zum Zitat Cabezas-Wallscheid N, Klimmeck D, Hansson J, Lipka DB, Reyes A, Wang Q, Weichenhan D, Lier A, von Paleske L, Renders S, et al. Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell. 2014;15:507–22.CrossRefPubMed Cabezas-Wallscheid N, Klimmeck D, Hansson J, Lipka DB, Reyes A, Wang Q, Weichenhan D, Lier A, von Paleske L, Renders S, et al. Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell. 2014;15:507–22.CrossRefPubMed
60.
Zurück zum Zitat Chen BJ, Cui X, Sempowski GD, Domen J, Chao NJ. Hematopoietic stem cell dose correlates with the speed of immune reconstitution after stem cell transplantation. Blood. 2004;103:4344–52.CrossRefPubMed Chen BJ, Cui X, Sempowski GD, Domen J, Chao NJ. Hematopoietic stem cell dose correlates with the speed of immune reconstitution after stem cell transplantation. Blood. 2004;103:4344–52.CrossRefPubMed
61.
Zurück zum Zitat Liu C, Chen BJ, Deoliveira D, Sempowski GD, Chao NJ, Storms RW. Progenitor cell dose determines the pace and completeness of engraftment in a xenograft model for cord blood transplantation. Blood. 2010;116:5518–27.CrossRefPubMedPubMedCentral Liu C, Chen BJ, Deoliveira D, Sempowski GD, Chao NJ, Storms RW. Progenitor cell dose determines the pace and completeness of engraftment in a xenograft model for cord blood transplantation. Blood. 2010;116:5518–27.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Wils EJ, Braakman E, Verjans GM, Rombouts EJ, Broers AE, Niesters HG, Wagemaker G, Staal FJ, Lowenberg B, Spits H, Cornelissen JJ. Flt3 ligand expands lymphoid progenitors prior to recovery of thymopoiesis and accelerates T cell reconstitution after bone marrow transplantation. J Immunol. 2007;178:3551–7.CrossRefPubMed Wils EJ, Braakman E, Verjans GM, Rombouts EJ, Broers AE, Niesters HG, Wagemaker G, Staal FJ, Lowenberg B, Spits H, Cornelissen JJ. Flt3 ligand expands lymphoid progenitors prior to recovery of thymopoiesis and accelerates T cell reconstitution after bone marrow transplantation. J Immunol. 2007;178:3551–7.CrossRefPubMed
63.
Zurück zum Zitat Al-Chami E, Tormo A, Khodayarian F, Rafei M. Therapeutic utility of the newly discovered properties of interleukin-21. Cytokine. 2016;82:33–7.CrossRefPubMed Al-Chami E, Tormo A, Khodayarian F, Rafei M. Therapeutic utility of the newly discovered properties of interleukin-21. Cytokine. 2016;82:33–7.CrossRefPubMed
64.
Zurück zum Zitat Attridge K, Wang CJ, Wardzinski L, Kenefeck R, Chamberlain JL, Manzotti C, Kopf M, Walker LS. IL-21 inhibits T cell IL-2 production and impairs Treg homeostasis. Blood. 2012;119:4656–64.CrossRefPubMed Attridge K, Wang CJ, Wardzinski L, Kenefeck R, Chamberlain JL, Manzotti C, Kopf M, Walker LS. IL-21 inhibits T cell IL-2 production and impairs Treg homeostasis. Blood. 2012;119:4656–64.CrossRefPubMed
65.
Zurück zum Zitat Jamil MO, Mineishi S. State-of-the-art acute and chronic GVHD treatment. Int J Hematol. 2015;101:452–66.CrossRefPubMed Jamil MO, Mineishi S. State-of-the-art acute and chronic GVHD treatment. Int J Hematol. 2015;101:452–66.CrossRefPubMed
66.
Zurück zum Zitat Gelebart P, Zak Z, Anand M, Dien-Bard J, Amin HM, Lai R. Interleukin-21 effectively induces apoptosis in mantle cell lymphoma through a STAT1-dependent mechanism. Leukemia. 2009;23:1836–46.CrossRefPubMed Gelebart P, Zak Z, Anand M, Dien-Bard J, Amin HM, Lai R. Interleukin-21 effectively induces apoptosis in mantle cell lymphoma through a STAT1-dependent mechanism. Leukemia. 2009;23:1836–46.CrossRefPubMed
67.
Zurück zum Zitat Sarosiek KA, Malumbres R, Nechushtan H, Gentles AJ, Avisar E, Lossos IS. Novel IL-21 signaling pathway up-regulates c-Myc and induces apoptosis of diffuse large B-cell lymphomas. Blood. 2010;115:570–80.CrossRefPubMedPubMedCentral Sarosiek KA, Malumbres R, Nechushtan H, Gentles AJ, Avisar E, Lossos IS. Novel IL-21 signaling pathway up-regulates c-Myc and induces apoptosis of diffuse large B-cell lymphomas. Blood. 2010;115:570–80.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Carlyle JR, Zuniga-Pflucker JC. Requirement for the thymus in alphabeta T lymphocyte lineage commitment. Immunity. 1998;9:187–97.CrossRefPubMed Carlyle JR, Zuniga-Pflucker JC. Requirement for the thymus in alphabeta T lymphocyte lineage commitment. Immunity. 1998;9:187–97.CrossRefPubMed
69.
Zurück zum Zitat Jamieson BD, Douek DC, Killian S, Hultin LE, Scripture-Adams DD, Giorgi JV, Marelli D, Koup RA, Zack JA. Generation of functional thymocytes in the human adult. Immunity. 1999;10:569–75.CrossRefPubMed Jamieson BD, Douek DC, Killian S, Hultin LE, Scripture-Adams DD, Giorgi JV, Marelli D, Koup RA, Zack JA. Generation of functional thymocytes in the human adult. Immunity. 1999;10:569–75.CrossRefPubMed
70.
Zurück zum Zitat Zinkernagel RM, Althage A. On the role of thymic epithelium vs. bone marrow-derived cells in repertoire selection of T cells. Proc Natl Acad Sci U S A. 1999;96:8092–7.CrossRefPubMedPubMedCentral Zinkernagel RM, Althage A. On the role of thymic epithelium vs. bone marrow-derived cells in repertoire selection of T cells. Proc Natl Acad Sci U S A. 1999;96:8092–7.CrossRefPubMedPubMedCentral
71.
Zurück zum Zitat Dooley J, Liston A. Molecular control over thymic involution: from cytokines and microRNA to aging and adipose tissue. Eur J Immunol. 2012;42:1073–9.CrossRefPubMed Dooley J, Liston A. Molecular control over thymic involution: from cytokines and microRNA to aging and adipose tissue. Eur J Immunol. 2012;42:1073–9.CrossRefPubMed
73.
Zurück zum Zitat Wu X, Tan Y, Xing Q, Wang S. IL-21 accelerates xenogeneic graft-versus-host disease correlated with increased B-cell proliferation. Protein Cell. 2013;4:863–71.CrossRefPubMedPubMedCentral Wu X, Tan Y, Xing Q, Wang S. IL-21 accelerates xenogeneic graft-versus-host disease correlated with increased B-cell proliferation. Protein Cell. 2013;4:863–71.CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat Takahama Y. Journey through the thymus: stromal guides for T-cell development and selection. Nat Rev Immunol. 2006;6:127–35.CrossRefPubMed Takahama Y. Journey through the thymus: stromal guides for T-cell development and selection. Nat Rev Immunol. 2006;6:127–35.CrossRefPubMed
75.
Zurück zum Zitat King C, Ilic A, Koelsch K, Sarvetnick N. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell. 2004;117:265–77.CrossRefPubMed King C, Ilic A, Koelsch K, Sarvetnick N. Homeostatic expansion of T cells during immune insufficiency generates autoimmunity. Cell. 2004;117:265–77.CrossRefPubMed
76.
Zurück zum Zitat Moses CT, Thorstenson KM, Jameson SC, Khoruts A. Competition for self ligands restrains homeostatic proliferation of naive CD4 T cells. Proc Natl Acad Sci U S A. 2003;100:1185–90.CrossRefPubMedPubMedCentral Moses CT, Thorstenson KM, Jameson SC, Khoruts A. Competition for self ligands restrains homeostatic proliferation of naive CD4 T cells. Proc Natl Acad Sci U S A. 2003;100:1185–90.CrossRefPubMedPubMedCentral
77.
Zurück zum Zitat Storek J, Gillespy 3rd T, Lu H, Joseph A, Dawson MA, Gough M, Morris J, Hackman RC, Horn PA, Sale GE, et al. Interleukin-7 improves CD4 T-cell reconstitution after autologous CD34 cell transplantation in monkeys. Blood. 2003;101:4209–18.CrossRefPubMed Storek J, Gillespy 3rd T, Lu H, Joseph A, Dawson MA, Gough M, Morris J, Hackman RC, Horn PA, Sale GE, et al. Interleukin-7 improves CD4 T-cell reconstitution after autologous CD34 cell transplantation in monkeys. Blood. 2003;101:4209–18.CrossRefPubMed
78.
Zurück zum Zitat Perales MA, Goldberg JD, Yuan J, Koehne G, Lechner L, Papadopoulos EB, Young JW, Jakubowski AA, Zaidi B, Gallardo H, et al. Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation. Blood. 2012;120:4882–91.CrossRefPubMedPubMedCentral Perales MA, Goldberg JD, Yuan J, Koehne G, Lechner L, Papadopoulos EB, Young JW, Jakubowski AA, Zaidi B, Gallardo H, et al. Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogeneic stem cell transplantation. Blood. 2012;120:4882–91.CrossRefPubMedPubMedCentral
79.
Zurück zum Zitat Mackall CL, Fry TJ, Bare C, Morgan P, Galbraith A, Gress RE. IL-7 increases both thymic-dependent and thymic-independent T-cell regeneration after bone marrow transplantation. Blood. 2001;97:1491–7.CrossRefPubMed Mackall CL, Fry TJ, Bare C, Morgan P, Galbraith A, Gress RE. IL-7 increases both thymic-dependent and thymic-independent T-cell regeneration after bone marrow transplantation. Blood. 2001;97:1491–7.CrossRefPubMed
80.
Zurück zum Zitat Li A, Zhang Q, Jiang J, Yuan G, Feng Y, Hao J, Li C, Gao X, Wang G, Xie S. Co-transplantation of bone marrow stromal cells transduced with IL-7 gene enhances immune reconstitution after allogeneic bone marrow transplantation in mice. Gene Ther. 2006;13:1178–87.CrossRefPubMed Li A, Zhang Q, Jiang J, Yuan G, Feng Y, Hao J, Li C, Gao X, Wang G, Xie S. Co-transplantation of bone marrow stromal cells transduced with IL-7 gene enhances immune reconstitution after allogeneic bone marrow transplantation in mice. Gene Ther. 2006;13:1178–87.CrossRefPubMed
81.
Zurück zum Zitat Gasnault J, de Goer de Herve MG, Michot JM, Hendel-Chavez H, Seta V, Mazet AA, Croughs T, Stankoff B, Bourhis JH, Lambotte O, et al. Efficacy of recombinant human interleukin 7 in a patient with severe lymphopenia-related progressive multifocal leukoencephalopathy. Open Forum Infect Dis. 2014;1:ofu074.CrossRefPubMedPubMedCentral Gasnault J, de Goer de Herve MG, Michot JM, Hendel-Chavez H, Seta V, Mazet AA, Croughs T, Stankoff B, Bourhis JH, Lambotte O, et al. Efficacy of recombinant human interleukin 7 in a patient with severe lymphopenia-related progressive multifocal leukoencephalopathy. Open Forum Infect Dis. 2014;1:ofu074.CrossRefPubMedPubMedCentral
82.
Zurück zum Zitat Chung B, Min D, Joo LW, Krampf MR, Huang J, Yang Y, Shashidhar S, Brown J, Dudl EP, Weinberg KI. Combined effects of interleukin-7 and stem cell factor administration on lymphopoiesis after murine bone marrow transplantation. Biol Blood Marrow Transplant. 2011;17:48–60.CrossRefPubMed Chung B, Min D, Joo LW, Krampf MR, Huang J, Yang Y, Shashidhar S, Brown J, Dudl EP, Weinberg KI. Combined effects of interleukin-7 and stem cell factor administration on lymphopoiesis after murine bone marrow transplantation. Biol Blood Marrow Transplant. 2011;17:48–60.CrossRefPubMed
83.
Zurück zum Zitat Chung B, Barbara-Burnham L, Barsky L, Weinberg K. Radiosensitivity of thymic interleukin-7 production and thymopoiesis after bone marrow transplantation. Blood. 2001;98:1601–6.CrossRefPubMed Chung B, Barbara-Burnham L, Barsky L, Weinberg K. Radiosensitivity of thymic interleukin-7 production and thymopoiesis after bone marrow transplantation. Blood. 2001;98:1601–6.CrossRefPubMed
84.
Zurück zum Zitat Broers AE, Posthumus-van Sluijs SJ, Spits H, van der Holt B, Lowenberg B, Braakman E, Cornelissen JJ. Interleukin-7 improves T-cell recovery after experimental T-cell-depleted bone marrow transplantation in T-cell-deficient mice by strong expansion of recent thymic emigrants. Blood. 2003;102:1534–40.CrossRefPubMed Broers AE, Posthumus-van Sluijs SJ, Spits H, van der Holt B, Lowenberg B, Braakman E, Cornelissen JJ. Interleukin-7 improves T-cell recovery after experimental T-cell-depleted bone marrow transplantation in T-cell-deficient mice by strong expansion of recent thymic emigrants. Blood. 2003;102:1534–40.CrossRefPubMed
85.
Zurück zum Zitat Bolotin E, Smogorzewska M, Smith S, Widmer M, Weinberg K. Enhancement of thymopoiesis after bone marrow transplant by in vivo interleukin-7. Blood. 1996;88:1887–94.PubMed Bolotin E, Smogorzewska M, Smith S, Widmer M, Weinberg K. Enhancement of thymopoiesis after bone marrow transplant by in vivo interleukin-7. Blood. 1996;88:1887–94.PubMed
86.
Zurück zum Zitat Alpdogan O, Schmaltz C, Muriglan SJ, Kappel BJ, Perales MA, Rotolo JA, Halm JA, Rich BE, van den Brink MR. Administration of interleukin-7 after allogeneic bone marrow transplantation improves immune reconstitution without aggravating graft-versus-host disease. Blood. 2001;98:2256–65.CrossRefPubMed Alpdogan O, Schmaltz C, Muriglan SJ, Kappel BJ, Perales MA, Rotolo JA, Halm JA, Rich BE, van den Brink MR. Administration of interleukin-7 after allogeneic bone marrow transplantation improves immune reconstitution without aggravating graft-versus-host disease. Blood. 2001;98:2256–65.CrossRefPubMed
87.
Zurück zum Zitat Alpdogan O, Muriglan SJ, Eng JM, Willis LM, Greenberg AS, Kappel BJ, van den Brink MR. IL-7 enhances peripheral T cell reconstitution after allogeneic hematopoietic stem cell transplantation. J Clin Invest. 2003;112:1095–107.CrossRefPubMedPubMedCentral Alpdogan O, Muriglan SJ, Eng JM, Willis LM, Greenberg AS, Kappel BJ, van den Brink MR. IL-7 enhances peripheral T cell reconstitution after allogeneic hematopoietic stem cell transplantation. J Clin Invest. 2003;112:1095–107.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Abdul-Hai A, Weiss L, Ben-Yehuda A, Ergas D, Shapira MY, Slavin S. Interleukin-7 induced facilitation of immunological reconstitution of sublethally irradiated mice following treatment with alloreactive spleen cells in a murine model of B-cell leukemia/lymphoma (BCL1). Bone Marrow Transplant. 2007;40:881–9.CrossRefPubMed Abdul-Hai A, Weiss L, Ben-Yehuda A, Ergas D, Shapira MY, Slavin S. Interleukin-7 induced facilitation of immunological reconstitution of sublethally irradiated mice following treatment with alloreactive spleen cells in a murine model of B-cell leukemia/lymphoma (BCL1). Bone Marrow Transplant. 2007;40:881–9.CrossRefPubMed
89.
Zurück zum Zitat Abdul-Hai A, Or R, Slavin S, Friedman G, Weiss L, Matsa D, Ben-Yehuda A. Stimulation of immune reconstitution by interleukin-7 after syngeneic bone marrow transplantation in mice. Exp Hematol. 1996;24:1416–22.PubMed Abdul-Hai A, Or R, Slavin S, Friedman G, Weiss L, Matsa D, Ben-Yehuda A. Stimulation of immune reconstitution by interleukin-7 after syngeneic bone marrow transplantation in mice. Exp Hematol. 1996;24:1416–22.PubMed
90.
Zurück zum Zitat Oh I, Ozaki K, Meguro A, Hatanaka K, Kadowaki M, Matsu H, Tatara R, Sato K, Iwakura Y, Nakae S, et al. Altered effector CD4+ T cell function in IL-21R-/- CD4+ T cell-mediated graft-versus-host disease. J Immunol. 2010;185:1920–6.CrossRefPubMedPubMedCentral Oh I, Ozaki K, Meguro A, Hatanaka K, Kadowaki M, Matsu H, Tatara R, Sato K, Iwakura Y, Nakae S, et al. Altered effector CD4+ T cell function in IL-21R-/- CD4+ T cell-mediated graft-versus-host disease. J Immunol. 2010;185:1920–6.CrossRefPubMedPubMedCentral
91.
Zurück zum Zitat Meguro A, Ozaki K, Oh I, Hatanaka K, Matsu H, Tatara R, Sato K, Leonard WJ, Ozawa K. IL-21 is critical for GVHD in a mouse model. Bone Marrow Transplant. 2010;45:723–9.CrossRefPubMed Meguro A, Ozaki K, Oh I, Hatanaka K, Matsu H, Tatara R, Sato K, Leonard WJ, Ozawa K. IL-21 is critical for GVHD in a mouse model. Bone Marrow Transplant. 2010;45:723–9.CrossRefPubMed
92.
Zurück zum Zitat Gudmundsdottir H, Turka LA. A closer look at homeostatic proliferation of CD4+ T cells: costimulatory requirements and role in memory formation. J Immunol. 2001;167:3699–707.CrossRefPubMed Gudmundsdottir H, Turka LA. A closer look at homeostatic proliferation of CD4+ T cells: costimulatory requirements and role in memory formation. J Immunol. 2001;167:3699–707.CrossRefPubMed
93.
Zurück zum Zitat Min B, Yamane H, Hu-Li J, Paul WE. Spontaneous and homeostatic proliferation of CD4 T cells are regulated by different mechanisms. J Immunol. 2005;174:6039–44.CrossRefPubMed Min B, Yamane H, Hu-Li J, Paul WE. Spontaneous and homeostatic proliferation of CD4 T cells are regulated by different mechanisms. J Immunol. 2005;174:6039–44.CrossRefPubMed
94.
Zurück zum Zitat Hechinger AK, Smith BA, Flynn R, Hanke K, McDonald-Hyman C, Taylor PA, Pfeifer D, Hackanson B, Leonhardt F, Prinz G, et al. Therapeutic activity of multiple common gamma-chain cytokine inhibition in acute and chronic GVHD. Blood. 2015;125:570–80.CrossRefPubMedPubMedCentral Hechinger AK, Smith BA, Flynn R, Hanke K, McDonald-Hyman C, Taylor PA, Pfeifer D, Hackanson B, Leonhardt F, Prinz G, et al. Therapeutic activity of multiple common gamma-chain cytokine inhibition in acute and chronic GVHD. Blood. 2015;125:570–80.CrossRefPubMedPubMedCentral
95.
Zurück zum Zitat Duffner UA, Maeda Y, Cooke KR, Reddy P, Ordemann R, Liu C, Ferrara JL, Teshima T. Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease. J Immunol. 2004;172:7393–8.CrossRefPubMed Duffner UA, Maeda Y, Cooke KR, Reddy P, Ordemann R, Liu C, Ferrara JL, Teshima T. Host dendritic cells alone are sufficient to initiate acute graft-versus-host disease. J Immunol. 2004;172:7393–8.CrossRefPubMed
96.
Zurück zum Zitat Horikawa M, Minard-Colin V, Matsushita T, Tedder TF. Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J Clin Invest. 2011;121:4268–80.CrossRefPubMedPubMedCentral Horikawa M, Minard-Colin V, Matsushita T, Tedder TF. Regulatory B cell production of IL-10 inhibits lymphoma depletion during CD20 immunotherapy in mice. J Clin Invest. 2011;121:4268–80.CrossRefPubMedPubMedCentral
97.
Zurück zum Zitat Iwata Y, Matsushita T, Horikawa M, Dilillo DJ, Yanaba K, Venturi GM, Szabolcs PM, Bernstein SH, Magro CM, Williams AD, et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood. 2011;117:530–41.CrossRefPubMedPubMedCentral Iwata Y, Matsushita T, Horikawa M, Dilillo DJ, Yanaba K, Venturi GM, Szabolcs PM, Bernstein SH, Magro CM, Williams AD, et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood. 2011;117:530–41.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Shlomchik WD, Couzens MS, Tang CB, McNiff J, Robert ME, Liu J, Shlomchik MJ, Emerson SG. Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 1999;285:412–5.CrossRefPubMed Shlomchik WD, Couzens MS, Tang CB, McNiff J, Robert ME, Liu J, Shlomchik MJ, Emerson SG. Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 1999;285:412–5.CrossRefPubMed
99.
Zurück zum Zitat Zhang Y, Shlomchik WD, Joe G, Louboutin JP, Zhu J, Rivera A, Giannola D, Emerson SG. APCs in the liver and spleen recruit activated allogeneic CD8+ T cells to elicit hepatic graft-versus-host disease. J Immunol. 2002;169:7111–8.CrossRefPubMed Zhang Y, Shlomchik WD, Joe G, Louboutin JP, Zhu J, Rivera A, Giannola D, Emerson SG. APCs in the liver and spleen recruit activated allogeneic CD8+ T cells to elicit hepatic graft-versus-host disease. J Immunol. 2002;169:7111–8.CrossRefPubMed
100.
Zurück zum Zitat Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, Watanabe T, Akashi K, Shultz LD, Harada M. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood. 2005;106:1565–73.CrossRefPubMedPubMedCentral Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, Watanabe T, Akashi K, Shultz LD, Harada M. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood. 2005;106:1565–73.CrossRefPubMedPubMedCentral
Metadaten
Titel
Interleukin-21 promotes thymopoiesis recovery following hematopoietic stem cell transplantation
verfasst von
Aurélie Tormo
Fatemeh Khodayarian
Yun Cui
Edouard Al-Chami
Reem Kanjarawi
Beatriz Noé
Huijie Wang
Moutih Rafei
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2017
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-017-0490-3

Weitere Artikel der Ausgabe 1/2017

Journal of Hematology & Oncology 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.