Although several cytokines have been demonstrated to be critical regulators of development of multiple blood cell lineages, it remains disputed to what degree they act through instructive or permissive mechanisms. Signaling through the FMS-like tyrosine kinase 3 (FLT3) receptor and the hematopoietin IL-7 receptor α (IL-7Rα) has been demonstrated to be of critical importance for sustained thymopoiesis. Signaling triggered by IL-7 and thymic stromal lymphopoietin (TSLP) is dependent on IL-7Rα, and both ligands have been implicated in T-cell development. However, we demonstrate that, whereas thymopoiesis is abolished in adult mice doubly deficient in IL-7 and FLT3 ligand (FLT3L), TSLP does not play a key role in IL-7–independent or FLT3L-independent T lymphopoiesis. Furthermore, whereas previous studies implicated that the role of other cytokine tyrosine kinase receptors in T lymphopoiesis might not involve permissive actions, we demonstrate that ectopic expression of BCL2 is sufficient not only to partially correct the T-cell phenotype of Flt3l−/− mice but also to rescue the virtually complete loss of all discernable stages of early T lymphopoiesis in Flt3l−/−Il7r−/− mice. These findings implicate a permissive role of cytokine receptors of the hematopoietin and tyrosine kinase families in early T lymphopoiesis.

Growth factor dependent hematopoietic cell lines can on overexpression of the antiapoptotic regulator B-cell lymphoma-2 (BCL2) become independent of cytokines of the hematopoietin family.1  In vivo studies demonstrating that defective thymopoiesis in interleukin-7 (IL-7)– and IL-7Rα–deficient (Il7−/− and Il7r−/−) mice can be rescued on overexpression of BCL22,4  or by deleting the proapoptotic regulator Bax,5  further support the hypothesis that hematopoietin family cytokine receptors play a permissive role in thymopoiesis. Hematopoiesis is also regulated by a distinct family of cytokines acting through tyrosine kinase receptors, such as KIT oncogene (KIT) and FMS-like tyrosine kinase 3 (FLT3).6  Notably, although KIT, like IL-7Rα, is critically involved in the regulation of thymopoiesis, BCL2 overexpression failed to rescue the impaired T lymphopoiesis in KIT-deficient mice.7  This implies that at least some members of this family of cytokine receptors and ligands, unlike the hematopoietin family, might not promote hematopoiesis through permissive actions

More recently, FLT3 ligand (FLT3L) has emerged as a key regulator of T lymphopoiesis, including of IL-7Rα–independent thymopoiesis,8,,,12  but it has not been investigated to what degree FLT3 might act through permissive actions. Furthermore, as both IL-7 and thymic stromal lymphopoietin (TSLP) share the IL-7Rα, it is unclear to what degree the almost completely arrested T lymphopoiesis in adult Flt3l−/−Il7r−/− mice8  reflects a role of FLT3L in conjunction with IL-7 and/or TSLP. TSLP also depends on and acts through a second IL-2R gamma chain (IL-2Rγ)-like TSLP receptor (TSLPR) subunit13  and has been shown to modulate immunologic responses by activating T cells through regulation of dendritic cells.14  TSLP has also been implicated to play an important role in IL-7–independent B lymphopoiesis, as Il7r−/− mice have a more severe reduction in B lymphopoiesis than Il7−/− mice.15,16  Similarly although TSLP- and TSLPR-deficient (Tpte2−/−) mice appear to have normal T lymphopoiesis,17,18  there are indications that T lymphopoiesis is more severely affected in Il7r−/− than Il7−/− mice, compatible with a role of TSLP in IL-7–independent T lymphopoiesis.19,,22  This finding is supported by studies of TSLPR and IL-2Rγ deficient (Tpte2−/−Il2rg−/−) mice, in which a further reduction of total thymic cellularity compared with single Il2rg−/− mice has been reported.18 

In the present studies, we first sought to establish the relative roles of IL-7, TSLP, and FLT3L in T lymphopoiesis. Notably, although Il7r−/− mice had a more severe block in thymopoiesis than Il7−/− mice, studies of Tpte2−/−, Il7−/−Tpte2−/−, and Flt3l−/−Tpte2−/− mice established that IL-7 and FLT3L are key regulators of normal thymopoiesis, with no discernable key role of TSLP. We also investigated to what degree the role of FLT3L in thymopoiesis is mediated through permissive actions. Through correction of the T-cell phenotype of Flt3l−/− mice, most notably through partial rescue of the almost complete loss of thymocytes in adult Flt3l−/−Il7r−/− mice by BCL2 overexpression, we provide evidence for the critical role and interaction of IL-7 and FLT3L in T lymphopoiesis, being mediated at least in part through permissive actions.

Mice

Flt3l−/− mice9  were on a pure C57BL/6 background. Il7r−/−21  (Jackson Laboratory, Bar Harbor, ME), Il7−/−23  and BCL2 transgenic mice24  were backcrossed for a total of 10 generations to C57BL/6. Tpte2−/− mice17  were kept on the original mixed 129/SvJ/C57BL/6 background. Double-deficient mice were generated through intercrossing the above mentioned strains except for the mice double-deficient in FLT3L and IL-7Rα, which were obtained by cross breeding of Flt3l−/− (pure C57BL/6) mice with Il7r−/− mice backcrossed 5 generations to C57BL/6.8  All experiments were approved by the Ethical Committee at Lund University.

Antibodies

Antibodies used for cell-surface staining were 2.4G2 (CD16/32), RA3-6B2 (B220), RB6-8C5 (Gr1), M1/70 (CD11b, Mac1), H129.19 (CD4), 53-7 (CD5), 53-6.7 (CD8α), LY-76 (Ter-119), 2B8 (CD117, c-kit), A20 (CD45.1), 104 (CD45.2), 3C7 (CD25), IM7 (CD44; all from BD PharMingen, San Diego, CA) and polyclonal goat-antirat Tricolor (Caltag, Invitrogen, Carlsbad, CA).

Fluorescence-activated cell sorting

For thymic SP and DP cell staging and splenic T-cell analysis, cells were incubated with anti–mouse-CD8 FITC and -CD4 APC. For DN1-4 staging, thymocytes were stained with a cocktail of purified rat antibodies against lineage markers B220, CD4, CD5, CD8α, CD11b, Gr1, and Ter119. Lineage-positive cells were visualized with a goat-antirat Tricolor staining. Thereafter, cells were incubated with anti–mouse-CD44 FITC, -CD25 PE, and -KIT APC. 7 Amino-Actinomycin (Sigma-Aldrich, St Louis, MO) staining was used to exclude dead cells. Analyses were performed on a FACSCalibur (BD). Data analysis was carried out using FlowJo software (Tree Star, Ashland, OR).

Polymerase chain reaction analysis

Genomic DNA and RNA were prepared using Trizol (Invitrogen) according to the manufacturer's instructions. Tcrb D-J, VDJ rearrangements were assayed by polymerase chain reaction (PCR), as previously described,25,26  using 38 cycles (94°C 1 minute, 61°C 1.5 minutes, and 72°C 2 minutes) of PCR.

The PCR products were blotted onto Hybond N+ nylon membranes (GE Healthcare, Little Chalfont, United Kingdom) using capillary blotting. Membranes were prehybridized in 5× Denhardts (Ficoll 10 g/L, polyvinylpyrrolidone 10 g/L, bovine serum albumin 10 g/L), 6× SSPE, 0.1% SDS, and 100 μg/mL Salmon Sperm DNA, at 45°C for 60 minutes and hybridized with a specific γ[32P] labeled oligonucleotide for 12 hours at 45°C in the same solution. Membranes were washed at room temperature in 2× SSC (0.015 M Na citrate, pH 7.0, containing 0.15 M NaCL) supplemented with 0.1% SDS for 15 minutes and 0.1× SSC with 0.1% SDS for 5 to 10 minutes. The hybridized membrane was then subject to auto radiography.

The following oligonucleotides were used for PCR: Tcrb-J2: 5′-TCTCCTACTATCGATTTCCCTCCCG-3′; Tcrb-D1: 5′-GGTAGACCTATG-3; Tcrb-V5.2: 5′-GTGGAGAGAGACAAAGGATT-3′; Tcrb-V8.3: 5′-TTGGCTTCTCCCTCTCAGACATCTT-3′. The following oligonucleotide was used for hybridization: Jβ2: 5′-TTTCCCTC-CCGGAGATTCCCTAA-3′; Gata-1 PCR: Gata1 sense: 5′-CCACTAGT-CCTGGCCTAGTACC-3′. Gata1 anti sense: 5′-CTGGCCTTGGCTC-TGAGTCTCC-3′.

RNA was prepared from cells using RNeasy Micro Kit (QIAGEN, VWR International, Stockholm, Sweden), and cDNA was generated by annealing 1 μg total RNA to 0.5 μg of random hexamers in 10 μL diethyl pyrocarbonate-treated water. Reverse transcriptase (RT) reactions were performed with 200 U SuperScriptII (Invitrogen) in accordance with the manufacturer's recommendations. One-twentieth of the RT reaction was used in the PCR assays. PCR was performed with 1 U Taq polymerase (Invitrogen) in the manufacture's buffer supplemented with 0.2 mM dNTP, in a total volume 25 μL. β-actin (actb) was amplified by 25 cycles, whereas 35 cycles were used to amplify Lck, Ptcra, Il7r, and Rag2 cDNA (94°C, 30 s, 60°C, 30 s, and 72°C, 30 s). The following primers were added to a final concentration of 1 mM: Actb sense: 5′-GTTTGAGACCTTCAACACC-3′; Actb anti sense: 5′-GTGGCCATCCCTGCTCGAAGTC-3′; Lck sense: 5′-GGTCAGAGACTTCGACCAGAAC-3′; Lck anti sense: 5′-CCACTGCATAAAGCCGGACTAG-3′; Ptcra sense: 5′-CACACGCTGGTAGATGGAAGGC-3′; Ptcra anti sense: 5′-GTCAGGAGCACATCGAGCAGAAG-3′; Il7r sense: 5′-AGCTGTTTCTGGAGAAAGTGG-3′; Il7r anti sense: 5′-AACGATTTCAGGTCAGAGGG-3′; Rag2 sense: 5′-GGTGTGCT-ACATCATATATTCTCCCAG-3′ Rag2 anti sense: 5′-TTCAATCGTGT-TGTCCCCTAGAG-3′.

In vivo reconstitution experiments

A total of 5 × 106 unfractionated BM cells from Flt3l−/−Il7r−/− or Flt3l−/−Il7r−/−BCL2 (CD45.2) littermates were transplanted into lethally irradiated Flt3l−/− (CD45.1) recipients. As positive controls, 5 × 106 littermate WT or BCL2 unfractionated BM cells were transplanted into WT (C57, CD45.1) recipients. Recipient mice were analyzed for the presence of donor-derived (CD45.2) T (CD4 or CD8) cell reconstitution at 6 weeks after transplantation. Briefly, thymocytes were stained with antibodies against CD45.1 and CD45.2 to determine the level of donor cell reconstitution and CD4 and CD8 to determine T-cell reconstitution.

Statistics

All results were expressed as mean (SD). The statistical significance between 2 groups of mice with deletion of single versus multiple signaling pathways was determined using a 1-tailed Student t test as removal of additional signaling pathways was hypothesized to result in the same or a more severe T-cell phenotype. Aspin-Welch correction was used in cases of unequal variances between groups.

For the BCL2-mediated rescue studies we tested, as an approximation to investigating if μ12 > μ34, the null hypothesis H′0: μ′1 -μ′2 ≤ μ′3 -μ′4 versus the alternative H′1: μ′1 -μ′2 > μ′3 -μ′4. μ1, μ2, μ3 and μ4 denote population means of the WT mice, cytokine knockout mice on WT background, BCL2 mice and cytokine knockout mice on BCL2 background, respectively, and μ′1, μ′2, μ′3 and μ′4 denote population means of the corresponding logarithmically transformed quantities. To test H′0 we used a t-type statistic with an Aspin-Welch approximation to the number of degrees of freedom. (For further details see Document S1, available on the Blood website; see the Supplemental Materials link at the top of the online article.)

The number of mice included in each statistical analysis is specified in the figure legends.

Critical roles of IL-7 and FLT3L but not TSLP in thymopoiesis

In accordance with previous studies17,18  we found no changes at any stages of thymocyte development in adult Tpte2−/− mice (Figure 1A,B). TSLP has been suggested to be important for IL-7-independent lymphopoiesis, largely based on a more severe lymphoid phenotype of IL-7Rα than IL-7 signaling deficient mice.15,16,20,22  To further explore whether TSLP might have a role in IL-7-independent thymopoiesis, we first compared thymopoiesis in age-matched Il7r−/− and Il7−/− mice (both on a C57BL/6 background) because Il7r−/− mice are deficient in IL-7 and TSLP-mediated signaling. In comparison to Il7−/− mice, Il7r−/− mice had further reductions in total (Figure 1C), double negative (DN; CD4, CD8) (Figure 1D), double positive (DP; CD4+, CD8+) and single positive (SP; CD4+ or CD8+; Figure 1E) thymocytes, indirectly supporting the idea that TSLP might play a role in IL-7–independent T lymphopoiesis. In an attempt to get direct evidence for a role of TSLP in IL-7–independent thymopoiesis, we next investigated Il7−/− and Il7−/−Tpte2−/− mice. Notably, no differences were observed in cellularity or at any stage of thymocyte development (Figure 1F-H), providing evidence for TSLP playing no key role in steady state adult IL-7–independent T lymphopoiesis.

Figure 1

Lack of role of TSLP in IL-7-dependent and independent T lymphopoiesis. (A,B) fluorescence-acivated cell sorting (FACS) profiles and mean (SD) numbers of (A) DN subsets and (B) CD4 and CD8 SP and DP cells in thymus of 9- to 11-week-old littermate WT and Tpte2−/− mice. Data are from 5 littermate WT and 7 Tpte2−/− mice from 3 separate litters. Numbers in FACS profiles show the mean percentages of cells within the indicated gates of total thymocytes. (C-E) Mean (SD) numbers of (C) total thymocytes, (D) DN cells, and (E) CD4 and CD8 SP and DP thymocytes of 3-week-old WT, Il7−/− and Il7r−/− mice. Data are from 4 WT, 7 Il7−/−, and 8 Il7r−/− mice from at least 2 separate litters. (F-H) Mean (SD) numbers of (F) total thymocytes, (G) DN cells, and (H) CD4+ and CD8+ thymocytes of 3-week-old littermate Il7−/− and Il7−/−Tpte2−/− mice. Data are from 7 littermate Il7−/− and 11 Il7−/−Tpte2−/− mice from 4 separate litters. All groups were investigated for statistical differences by Student t test (*P < .05, **P < .01, ***P < .001). Unless otherwise indicated differences were nonsignificant.

Figure 1

Lack of role of TSLP in IL-7-dependent and independent T lymphopoiesis. (A,B) fluorescence-acivated cell sorting (FACS) profiles and mean (SD) numbers of (A) DN subsets and (B) CD4 and CD8 SP and DP cells in thymus of 9- to 11-week-old littermate WT and Tpte2−/− mice. Data are from 5 littermate WT and 7 Tpte2−/− mice from 3 separate litters. Numbers in FACS profiles show the mean percentages of cells within the indicated gates of total thymocytes. (C-E) Mean (SD) numbers of (C) total thymocytes, (D) DN cells, and (E) CD4 and CD8 SP and DP thymocytes of 3-week-old WT, Il7−/− and Il7r−/− mice. Data are from 4 WT, 7 Il7−/−, and 8 Il7r−/− mice from at least 2 separate litters. (F-H) Mean (SD) numbers of (F) total thymocytes, (G) DN cells, and (H) CD4+ and CD8+ thymocytes of 3-week-old littermate Il7−/− and Il7−/−Tpte2−/− mice. Data are from 7 littermate Il7−/− and 11 Il7−/−Tpte2−/− mice from 4 separate litters. All groups were investigated for statistical differences by Student t test (*P < .05, **P < .01, ***P < .001). Unless otherwise indicated differences were nonsignificant.

Close modal

Compared with mice deficient in FLT3L or IL-7Rα, thymopoiesis in mice double-deficient in FLT3L and IL-7Rα (Flt3l−/−Il7r−/−) is severely impaired and almost completely arrested already in young adult mice.8  To establish the relative role of IL-7 and TSLP in FLT3L-independent thymopoiesis, we compared thymopoiesis in Flt3l−/−Il7−/− and Flt3l−/−Tpte2−/− mice. Notably, whereas Flt3l−/−Il7r−/− and Flt3l−/−Il7−/− mice had indistinguishable thymocyte deficiencies with a virtually complete loss of all thymocyte subsets (Figure 2A-D), Flt3l−/−Tpte2−/− mice had a thymocyte phenotype indistinguishable from Flt3l−/− mice (Figure 2E,F). Thus, whereas FLT3L and IL-7 play critical and complementary roles in T lymphopoiesis, TSLP appears to have no important role in IL-7–independent or FLT3L-independent thymopoiesis.

Figure 2

Critical roles of FLT3L and IL-7 but not TSLP in adult thymopoiesis. (A) Mean (SD) thymic cellularity, (B) FACS profiles of DN development, and mean (SD) numbers of (C) DN cells and (D) CD4 and CD8 SP and DP thymocytes of 9- to 11-week-old WT, Flt3l−/−Il7−/− and Flt3l−/−Il7r−/− mice. Numbers in FACS profiles show the mean percentages (of total thymocytes) of cells within the indicated gates. Data are from 5 WT, 6 to 13 Flt3l−/−Il7−/− and 4-10 Flt3l−/−Il7r−/− mice from at least 2 separate litters. (E,F) Mean (SD) numbers of (E) DN cells and (F) CD4 and CD8 SP and DP thymocytes of 9- to 11 week-old littermate Flt3l−/− and Flt3l−/−Tpte2−/− mice. Data are from 8 littermate Flt3l−/− and 10 Flt3l−/−Tpte2−/− mice from 2 separate litters. All groups were investigated for statistical differences by Student t test (*P < .05, **P < .01). Unless otherwise indicated, differences were nonsignificant.

Figure 2

Critical roles of FLT3L and IL-7 but not TSLP in adult thymopoiesis. (A) Mean (SD) thymic cellularity, (B) FACS profiles of DN development, and mean (SD) numbers of (C) DN cells and (D) CD4 and CD8 SP and DP thymocytes of 9- to 11-week-old WT, Flt3l−/−Il7−/− and Flt3l−/−Il7r−/− mice. Numbers in FACS profiles show the mean percentages (of total thymocytes) of cells within the indicated gates. Data are from 5 WT, 6 to 13 Flt3l−/−Il7−/− and 4-10 Flt3l−/−Il7r−/− mice from at least 2 separate litters. (E,F) Mean (SD) numbers of (E) DN cells and (F) CD4 and CD8 SP and DP thymocytes of 9- to 11 week-old littermate Flt3l−/− and Flt3l−/−Tpte2−/− mice. Data are from 8 littermate Flt3l−/− and 10 Flt3l−/−Tpte2−/− mice from 2 separate litters. All groups were investigated for statistical differences by Student t test (*P < .05, **P < .01). Unless otherwise indicated, differences were nonsignificant.

Close modal

Overexpression of BCL2 partially rescues all stages of T-cell development in Flt3l−/−Il7r−/− mice

We next investigated to what degree the role of FLT3L in thymopoiesis is mediated through permissive actions. Although somewhat contentious,7  IL-2Rγ and IL-7Rα have, through overexpression of BCL2, been suggested to largely act in a permissive manner in αβ T lymphopoiesis.2,4  In contrast, and through a similar strategy, it was not possible to obtain support for a permissive role of the cytokine tyrosine kinase receptor KIT in thymopoiesis.7  To investigate to what degree the critical importance of FLT3 signaling in early T-cell development might involve permissive actions, we examined thymopoiesis in Flt3l−/−, Il7r−/− and Flt3l−/−Il7r−/− mice on a BCL2 transgenic (BCL2) background, obtained by intercrossing each of the strains with transgenic mice with high pan-hematopoietic expression of BCL2.24  Because the loss of thymocytes in all these strains increases with age, we performed this analysis in 7- to 9-week-old mice at which time only minute numbers of DN cells are sustained in Flt3l−/−Il7r−/− mice (Figure 3A-H). Notably, not only could we demonstrate an extensive rescue (although not completely to the levels in BCL2 mice) of the earliest as well as later stages of T lymphopoiesis in the thymus of Il7r−/− mice, but also the reduction in DN progenitors in Flt3l−/− mice were largely rescued on BCL2 overexpression, including rescue of KIT-positive DN1 and DN2 immature T-cell progenitors27,29  (Figure 3A-H). Most notably, the virtually complete loss of all stages of DN as well as DP and SP thymocytes in Flt3l−/−Il7r−/− mice was substantially rescued by BCL2, resulting in the recovery of all stages of thymocyte progenitors to the same levels observed in Il7r−/− mice in which BCL2 had been overexpressed (Figure 3A-K). For all populations, statistical analyses were performed to test whether the reduction of lymphocyte populations in the cytokine deficient mice on a WT background was more severe than on a BCL2 background, using data transformed to a logarithmic scale (see “Methods” and Document S1 for details). Based on these calculations, the reductions of DN1KIT+, DN2-DN4, DP and SP CD4/8 thymocyte populations were all significantly (P < .01) reduced by BCL2 in IL-7Rα-deficient mice (Figure 3A-K). Similarly, reductions of DN2, DN2KIT+, and DN3, were significantly (P < .05) reduced by BCL2 in FLT3L-deficient mice (Figure 3E-G). Comparing the double deficiency of FLT3L and IL-7Rα on a WT and BCL2 background, the reductions of thymocytes on the BCL2 background was much less severe for all thymocyte populations (Figure 3A-K, P < .01). Moreover, in Flt3l−/−Il7r−/− mice, numbers of CD4+ and CD8+ T cells in spleen were increased 99- and 15-fold, respectively, in mice on a BCL2 compared with WT background (Figure S1). Notably, numbers of splenic T cells in Flt3l−/−Il7r−/−BCL2 mice were not significantly different from T cells in Il7r−/−BCL2 mice (Figure S1).

Figure 3

BCL2-mediated rescue of defective thymopoiesis in adult Flt3l−/−Il7r−/− mice. (A) Mean (SD) thymic cellularities, (B) FACS profiles of DN staging, and mean (SD) numbers of (C) DN1, (D) DN1KIT+, (E) DN2, (F) DN2KIT+, (G) DN3, (H) DN4, (I) DP, (J) CD4 SP, and (K) CD8 SP cells in the thymus of 7- to 9-week-old WT, Flt3l−/−, Il7r−/− and Flt3l−/−Il7r−/− mice on WT or BCL2 transgenic background. Data are from 3 to 4 WT, 12 Flt3l−/−, 8 to 12 Il7r−/−, 4 to 10 Flt3l−/−Il7r−/−, 7 to 9 BCL2, 14 Flt3l−/−BCL2, 12 Il7r−/−BCL2, and 6 to 7 Flt3l−/−Il7r−/−BCL2 mice, in all cases from at least 2 separate litters. Numbers in FACS profiles show the mean percentage (of total thymocytes) of cells within the indicated gates. For each strain and cell type, cutoff numbers in graphs are indicated. However, for one mouse in the Flt3l−/−Il7r−/− group, in which no DN1KIT+ and DN2KIT+ cells were detected, too few events were acquired to be applicable to the cutoff value.

Figure 3

BCL2-mediated rescue of defective thymopoiesis in adult Flt3l−/−Il7r−/− mice. (A) Mean (SD) thymic cellularities, (B) FACS profiles of DN staging, and mean (SD) numbers of (C) DN1, (D) DN1KIT+, (E) DN2, (F) DN2KIT+, (G) DN3, (H) DN4, (I) DP, (J) CD4 SP, and (K) CD8 SP cells in the thymus of 7- to 9-week-old WT, Flt3l−/−, Il7r−/− and Flt3l−/−Il7r−/− mice on WT or BCL2 transgenic background. Data are from 3 to 4 WT, 12 Flt3l−/−, 8 to 12 Il7r−/−, 4 to 10 Flt3l−/−Il7r−/−, 7 to 9 BCL2, 14 Flt3l−/−BCL2, 12 Il7r−/−BCL2, and 6 to 7 Flt3l−/−Il7r−/−BCL2 mice, in all cases from at least 2 separate litters. Numbers in FACS profiles show the mean percentage (of total thymocytes) of cells within the indicated gates. For each strain and cell type, cutoff numbers in graphs are indicated. However, for one mouse in the Flt3l−/−Il7r−/− group, in which no DN1KIT+ and DN2KIT+ cells were detected, too few events were acquired to be applicable to the cutoff value.

Close modal

We next investigated whether the BCL2-rescued thymocytes in Flt3l−/−Il7r−/− mice expressed genes critical for T-cell development. RT-PCR analysis demonstrated that thymocytes from Flt3l−/−Il7r−/−BCL2 mice expressed recombination activating gene 2 (Rag2) and Pre-T-cell antigen receptor α (Ptcra), required for T-cell receptor (TCR) rearrangements and assembly of the pre-TCR. In contrast, expression of these genes was undetectable in residual thymocytes from Flt3l−/−Il7r−/− mice (Figure 4A). Furthermore, Tcrb-D-J (Figure 4B) and Tcrb-V-DJ (Figure 4C) rearrangement diversity was observed in Flt3l−/−Il7r−/−BCL2 thymocytes in a pattern comparable with WT thymocytes, supporting the hypothesis that BCL2 rescues polyclonal thymopoiesis in adult Flt3l−/−Il7r−/− mice.

Figure 4

Molecular evidence for BCL2-induced rescue of FLT3L and IL-7Rα–dependent thymopoiesis in Flt3l−/−Il7r−/− mice. (A) Ethidium bromide-stained agarose gels with resulting PCR products from RT-PCR analysis of Rag2, Ptcra, Lck, and Il7r message in thymus from mice of the indicated genotypes. Autoradiograms of (B) Tcrb-D1-J rearrangements and (C) Tcrb-V-DJ rearrangements in the thymus of WT, BCL2, Flt3l−/−Il7r−/−, and Flt3l−/−Il7r−/−BCL2 mice. Amplification of a region in the Gata1 gene was performed to verify that the DNA preparations were of comparable quality. Results are from representative mice of each genotype. Arrows indicate rearrangement products. Please note that lines visible on reproduction of gels represent reproduction artifacts.

Figure 4

Molecular evidence for BCL2-induced rescue of FLT3L and IL-7Rα–dependent thymopoiesis in Flt3l−/−Il7r−/− mice. (A) Ethidium bromide-stained agarose gels with resulting PCR products from RT-PCR analysis of Rag2, Ptcra, Lck, and Il7r message in thymus from mice of the indicated genotypes. Autoradiograms of (B) Tcrb-D1-J rearrangements and (C) Tcrb-V-DJ rearrangements in the thymus of WT, BCL2, Flt3l−/−Il7r−/−, and Flt3l−/−Il7r−/−BCL2 mice. Amplification of a region in the Gata1 gene was performed to verify that the DNA preparations were of comparable quality. Results are from representative mice of each genotype. Arrows indicate rearrangement products. Please note that lines visible on reproduction of gels represent reproduction artifacts.

Close modal

Multipotent progenitor cells in the BM are required to continuously replace short-lived thymocytes.30  However, Flt3l−/−Il7r−/− BM cells fail to reconstitute thymopoiesis.8  Strikingly, whereas transplantation of Flt3l−/−Il7r−/− BM cells into lethally irradiated Flt3l−/− recipients failed to reconstitute thymopoiesis, thymic reconstitution was observed with Flt3l−/−Il7r−/−BCL2 BM cells (Figure 5A,B). Specifically, total donor-derived thymic cellularity, DP, CD4 SP, and CD8 SP thymocytes were increased 37-fold, 98-fold, 163-fold, and 49-fold, respectively, in recipients of Flt3l−/−Il7r−/−BCL2 BM compared with Flt3l−/−Il7r−/− BM cells (Figure 5A,B).

Figure 5

Ectopic expression of BCL2 enhances the T-cell reconstitution potential of Flt3l−/−Il7r−/− BM cells. 5 × 106 littermate Flt3l−/−Il7r−/− or Flt3l−/−Il7r−/−BCL2 (CD45.2) unfractionated BM cells were transplanted into lethally irradiated Flt3l−/− (CD45.1) recipients. As positive controls, 5 × 106 littermate WT or BCL2 unfractionated BM cells were transplanted into WT (CD45.1) recipients. (A) FACS profiles showing donor (CD45.2)-derived CD4 and CD8 DP and SP cells in thymus 6 weeks after transplantation. Numbers in FACS profiles represent the mean percentage of total donor cells relative to total thymocytes and the contribution of CD4 and CD8 cells within donor cells. (B) Mean (SD) numbers of donor-derived total, DP, and SP CD4+ and CD8+ thymocytes per thymus, at 6 weeks after transplantation. Data are from 4 recipients of WT, Flt3l−/−Il7r−/−, Flt3l−/−Il7r−/−BCL2 and 3 recipients of BCL2 cells.

Figure 5

Ectopic expression of BCL2 enhances the T-cell reconstitution potential of Flt3l−/−Il7r−/− BM cells. 5 × 106 littermate Flt3l−/−Il7r−/− or Flt3l−/−Il7r−/−BCL2 (CD45.2) unfractionated BM cells were transplanted into lethally irradiated Flt3l−/− (CD45.1) recipients. As positive controls, 5 × 106 littermate WT or BCL2 unfractionated BM cells were transplanted into WT (CD45.1) recipients. (A) FACS profiles showing donor (CD45.2)-derived CD4 and CD8 DP and SP cells in thymus 6 weeks after transplantation. Numbers in FACS profiles represent the mean percentage of total donor cells relative to total thymocytes and the contribution of CD4 and CD8 cells within donor cells. (B) Mean (SD) numbers of donor-derived total, DP, and SP CD4+ and CD8+ thymocytes per thymus, at 6 weeks after transplantation. Data are from 4 recipients of WT, Flt3l−/−Il7r−/−, Flt3l−/−Il7r−/−BCL2 and 3 recipients of BCL2 cells.

Close modal

Mice doubly deficient in FLT3L and IL-7Rα have virtually no existing adult thymopoiesis.8  Because the IL-7Rα has 2 ligands, IL-7 and TSLP, Flt3l−/−Il7r−/− mice lack FLT3L, IL-7, as well as TSLP-mediated signaling. In the present studies, we explored the relative importance of these key cytokines, all implicated in early T-cell development. TSLP has, by comparative studies in IL-7Rα and IL-7 signaling deficient mice, been suggested to have a major role in IL-7–independent B lymphopoiesis.15,16  Furthermore, findings in support of a role for TSLP in IL-7–independent T-cell development have been reported, as mice double-deficient in IL-2Rγ and TSLPR show an additional thymic phenotype compared with single IL-2Rγ–deficient mice.18  Our comparative studies of IL-7Rα– and IL-7–deficient mice confirmed a more severe T-cell phenotype in Il7r−/− than Il7−/− mice, further supporting the idea that TSLP might play a role in IL-7–independent thymopoiesis. However, when directly investigating the role for TSLP in IL-7–independent thymopoiesis, we found no clear differences at any stage of thymocyte development between Il7−/− and Il7−/−Tpte2−/− mice, clearly indicating that TSLP does not play a key role in IL-7–independent T lymphopoiesis. In light of these findings, the more aggravated T-cell phenotype in Il7r−/− compared with Il7−/− mice may instead be explained by alternative, TSLP-independent, mechanisms. Such mechanisms may potentially include yet unidentified, IL-7Rα ligands or cross-activation of IL-7Rα in Il7−/− mice through other pathways, such as KIT, as recently suggested.31  Although our findings do not support a key role of TSLP in IL-7–dependent or – independent steady state thymopoiesis, it remains possible that TSLP might be involved in T lymphopoiesis in conditions other than steady state.

We also failed to obtain any evidence in support of TSLP playing a role in FLT3L-independent T lymphopoiesis as Flt3l−/− and Flt3l−/−Tpte2−/− mice revealed indistinguishable T-cell phenotypes. Furthermore, the lack of active thymopoiesis in adult Flt3l−/−Il7−/− as well as Flt3l−/−Il7r−/− mice unequivocally demonstrated that intact TSLP function is insufficient for sustaining active thymopoiesis in the absence of FLT3L and IL-7–mediated signaling. Taken together, FLT3L and IL-7 are critical extrinsic regulators of thymopoiesis, whereas TSLP appears to plays no critical role.

Previous studies have questioned whether the important functions of cytokine tyrosine kinase receptors in T lymphopoiesis are mediated through permissive actions because overexpression of BCL2 had no rescuing effect on defective T lymphopoiesis in KIT-deficient mice.7  In contrast, we demonstrated here that the defect in early T-cell progenitors in Flt3l−/− mice, and the virtually complete loss of all stages of thymocytes in adult Flt3l−/−Il7r−/− mice is largely rescued (although not fully to the levels of BCL2 transgenic mice), and all developmental stages become clearly detectable, on enforced expression of BCL2. In further support of a BCL2-mediated rescue of normal polyclonal thymopoiesis in adult Flt3l−/−Il7r−/− mice, rearrangement diversity and expression of key genes required for normal T-cell development comparable with that of WT thymocytes were restored on overexpression of BCL2 in Flt3l−/−Il7r−/− mice. Furthermore, the inability of adult Flt3l−/−Il7r−/− BM cells to reconstitute the thymus of FLT3L deficient mice was partially corrected on enforced expression of BCL2.

In conclusion, our studies demonstrate for the first time that the requirement for ligands of cytokine tyrosine kinase receptors (ie, FLT3), as ligands of the hematopoietin receptor family (ie, IL-7), can largely be substituted by the potent antiapoptotic regulator BCL2, supporting the hypothesis that both of these cytokine receptor pathways at least in part mediate critical functions in T-cell development through permissive actions.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank Lilian Wittman, Sudi Wu, and Olof Gidlöv for expert technical assistance, Dr Ana Cumano and Dr James Ihle for generously providing the Il7−/− and the Tpte2−/− mice, and Dr Helen Ferry for critically reviewing this manuscript.

This work was supported by grants from the Swedish Research Council, Juvenile Diabetes Research Foundation (JDRF), and the Göran Gustafsson Foundation. The Lund Stem Cell Center is supported by a Center of Excellence grant from the Swedish Foundation for Strategic Research.

Contribution: C.T.J., M.S., E.S., and S.E.W.J. designed and conceptualized the research, analyzed the data, and wrote the manuscript; C.T.J., S.K., C.B., M.S., and A.L. did the characterization of the different mouse strains; and T.R. analyzed the data and performed statistical analysis.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Sten Eirik W. Jacobsen, Hematopoietic Stem Cell Laboratory, Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University, Klinikgatan 26, 221 84, Lund, Sweden; e-mail: sten.jacobsen@med.lu.se.

1
Fairbairn
LJ
Cowling
GJ
Reipert
BM
Dexter
TM
Suppression of apoptosis allows differentiation and development of a multipotent hemopoietic cell line in the absence of added growth factors.
Cell
1993
74
823
832
2
Akashi
K
Kondo
M
von Freeden-Jeffry
U
Murray
R
Weissman
IL
Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice.
Cell
1997
89
1033
1041
3
Kondo
M
Akashi
K
Domen
J
Sugamura
K
Weissman
IL
Bcl-2 rescues T lymphopoiesis, but not B or NK cell development, in common gamma chain-deficient mice.
Immunity
1997
7
155
162
4
Maraskovsky
E
O'Reilly
LA
Teepe
M
Corcoran
LM
Peschon
JJ
Strasser
A
Bcl-2 can rescue T lymphocyte development in interleukin-7 receptor-deficient mice but not in mutant rag-1−/− mice.
Cell
1997
89
1011
1019
5
Khaled
AR
Li
WQ
Huang
J
et al
Bax deficiency partially corrects interleukin-7 receptor alpha deficiency.
Immunity
2002
17
561
573
6
Lyman
SD
Jacobsen
SE
c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities.
Blood
1998
91
1101
1134
7
Rodewald
HR
Waskow
C
Haller
C
Essential requirement for c-kit and common gamma chain in thymocyte development cannot be overruled by enforced expression of Bcl-2.
J Exp Med
2001
193
1431
1437
8
Sitnicka
E
Buza-Vidas
N
Ahlenius
H
et al
Critical role of FLT3 ligand in IL-7 receptor independent T lymphopoiesis and regulation of lymphoid-primed multipotent progenitors.
Blood
2007
110
2955
2964
9
McKenna
HJ
Stocking
KL
Miller
RE
et al
Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells.
Blood
2000
95
3489
3497
10
Sambandam
A
Maillard
I
Zediak
VP
et al
Notch signaling controls the generation and differentiation of early T lineage progenitors.
Nat Immunol
2005
6
663
670
11
Schwarz
BA
Sambandam
A
Maillard
I
Harman
BC
Love
PE
Bhandoola
A
Selective thymus settling regulated by cytokine and chemokine receptors.
J Immunol
2007
178
2008
2017
12
Sitnicka
E
Bryder
D
Theilgaard-Monch
K
Buza-Vidas
N
Adolfsson
J
Jacobsen
SE
Key role of flt3 ligand in regulation of the common lymphoid progenitor but not in maintenance of the hematopoietic stem cell pool.
Immunity
2002
17
463
472
13
Pandey
A
Ozaki
K
Baumann
H
et al
Cloning of a receptor subunit required for signaling by thymic stromal lymphopoietin.
Nat Immunol
2000
1
59
64
14
Liu
YJ
Thymic stromal lymphopoietin: master switch for allergic inflammation.
J Exp Med
2006
203
269
273
15
Vosshenrich
CA
Cumano
A
Muller
W
Di Santo
JP
Vieira
P
Thymic stromal-derived lymphopoietin distinguishes fetal from adult B cell development.
Nat Immunol
2003
4
773
779
16
Vosshenrich
CA
Cumano
A
Muller
W
Di Santo
JP
Vieira
P
Pre-B cell receptor expression is necessary for thymic stromal lymphopoietin responsiveness in the bone marrow but not in the liver environment.
Proc Natl Acad Sci U S A
2004
101
11070
11075
17
Carpino
N
Thierfelder
WE
Chang
MS
et al
Absence of an essential role for thymic stromal lymphopoietin receptor in murine B-cell development.
Mol Cell Biol
2004
24
2584
2592
18
Al-Shami
A
Spolski
R
Kelly
J
et al
A role for thymic stromal lymphopoietin in CD4(+) T cell development.
J Exp Med
2004
200
159
168
19
Fry
TJ
Mackall
CL
The many faces of IL-7: from lymphopoiesis to peripheral T cell maintenance.
J Immunol
2005
174
6571
6576
20
Moore
TA
von Freeden-Jeffry
U
Murray
R
Zlotnik
A
Inhibition of gamma delta T cell development and early thymocyte maturation in IL-7 −/− mice.
J Immunol
1996
157
2366
2373
21
Peschon
JJ
Morrissey
PJ
Grabstein
KH
et al
Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice.
J Exp Med
1994
180
1955
1960
22
von Freeden-Jeffry
U
Vieira
P
Lucian
LA
McNeil
T
Burdach
SE
Murray
R
Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine.
J Exp Med
1995
181
1519
1526
23
Dias
S
Silva
H
Jr
Cumano
A
Vieira
P
Interleukin-7 is necessary to maintain the B cell potential in common lymphoid progenitors.
J Exp Med
2005
201
971
979
24
Domen
J
Gandy
KL
Weissman
IL
Systemic overexpression of BCL-2 in the hematopoietic system protects transgenic mice from the consequences of lethal irradiation.
Blood
1998
91
2272
2282
25
van Meerwijk
JP
Bluthmann
H
Steinmetz
M
T-cell specific rearrangement of T-cell receptor beta transgenes in mice.
EMBO J
1990
9
1057
1062
26
Okamura
RM
Sigvardsson
M
Galceran
J
Verbeek
S
Clevers
H
Grosschedl
R
Redundant regulation of T cell differentiation and TCRalpha gene expression by the transcription factors LEF-1 and TCF-1.
Immunity
1998
8
11
20
27
Godfrey
DI
Kennedy
J
Suda
T
Zlotnik
A
A developmental pathway involving four phenotypically and functionally distinct subsets of CD3-CD4-CD8-triple-negative adult mouse thymocytes defined by CD44 and CD25 expression.
J Immunol
1993
150
4244
4252
28
Porritt
HE
Rumfelt
LL
Tabrizifard
S
Schmitt
TM
Zuniga-Pflucker
JC
Petrie
HT
Heterogeneity among DN1 prothymocytes reveals multiple progenitors with different capacities to generate T cell and non-T cell lineages.
Immunity
2004
20
735
745
29
Ceredig
R
Rolink
T
A positive look at double-negative thymocytes.
Nat Rev Immunol
2002
2
888
897
30
Bhandoola
A
Sambandam
A
Allman
D
Meraz
A
Schwarz
B
Early T lineage progenitors: new insights, but old questions remain.
J Immunol
2003
171
5653
5658
31
Jahn
T
Sindhu
S
Gooch
S
et al
Direct interaction between Kit and the interleukin-7 receptor.
Blood
2007
110
1840
1847
Sign in via your Institution