Myeloablative conditioning results in thymic epithelial cell (TEC) injury, slow T-cell reconstitution, and a high risk of opportunistic infections. Keratinocyte growth factor (KGF) stimulates TEC proliferation and, when given preconditioning, reduces TEC injury. Thymocytes and TECs express androgen receptors, and exposure to androgen inhibits thymopoiesis. In this study, we have investigated whether TEC stimulation via preconditioning treatment with KGF and leuprolide acetate (Lupron), 2 clinically approved agents, given only before conditioning would circumvent the profound TEC and associated T-cell deficiency seen in allogeneic bone marrow transplant (BMT) recipients. Only combined treatment with KGF plus leuprolide acetate normalized TEC subset numbers and thymic architecture. Thymopoiesis and thymic output were supranormal, leading to the accelerated peripheral reconstitution of naive CD4 and CD8 T cells with a broad Vβ repertoire and decreased homeostatic T-cell proliferation. Combined therapy facilitated T:B cooperativity and enabled a B-cell humoral response to a CD4 T cell–dependent neoantigen challenge soon after BMT. In vivo antigen-specific CD8 T-cell responses and clearance of a live pathogen was superior with combined versus individual agent therapy. Thus, KGF combined with androgen blockade represents a novel approach to restore thymic function and facilitates the rapid recovery of peripheral T-cell function after allogeneic BMT.

Allogeneic bone marrow transplantation (BMT) is a valuable treatment option for malignant and nonmalignant disorders.1,2  After myeloablative conditioning, a favorable outcome depends upon successful immune reconstitution, including the de novo generation of a polyclonal population of naive T cells in the thymus.2-6  Mature T-cell generation is substantially delayed after BMT, primarily because of thymic injury induced by pre-BMT chemoradiotherapy and graft-versus-host disease (GVHD).3,5  Fungal and viral infections normally controlled by T cells can occur at high frequency in BMT patients, resulting in significant morbidity and mortality.7  Thus, strategies are needed to speed thymopoiesis after BMT.

Normal thymopoiesis involves a program of thymocyte differentiation and maturation through sequential stages characterized by CD4 and CD8 expression—CD4CD8 (“double negative”), CD4+CD8+ (“double positive”), and CD4+ or CD8+ (“single positive”)—culminating in the export of mature CD4+ and CD8+ T cells into the periphery.8  The thymic stroma is composed primarily of a 3-dimensional matrix of cortical and medullary thymic epithelial cells (TECs).9  TECs directly support thymocyte development and selection but are susceptible to BMT-conditioning-induced damage, impairing the ability of the thymus to produce T cells for prolonged periods of time after BMT.10-13 

Several growth factors regulate the development, proliferation, and function of TECs throughout life, including fibroblast growth factor-7 (FGF-7), also known as keratinocyte growth factor (KGF).14-16  KGF is an epithelial growth factor mainly produced by mesenchymal cells in the thymus and binds exclusively to a specific member of the fibroblast growth factor receptor-2 family, FGFR2-IIIb (KGFR), which is expressed in the thymus by TEC.17  KGF can aid in the protection and/or repair of epithelial cells in murine models of radiation- and chemotherapy-induced injury and is FDA-approved for the prevention of oral mucositis associated with chemoradiotherapy and BMT.18-20  Murine studies have demonstrated that thymic injury and prolonged immune deficiency can be prevented by KGF pretreatment in models with and without GVHD.11,21,22  KGF also has been shown to facilitate engraftment and abrogate GVHD-induced lethality in murine BMT recipients.23 

The thymic atrophy that occurs with advancing age has been partly linked to physiologic changes in sex steroid hormone production.24-26  Androgen receptors (ARs) are expressed on TECs, certain thymocyte subsets, and mature T cells, although the exact mechanisms by which androgens exert their effects on thymopoiesis and T-cell homeostasis/function are not fully understood.27-31  Physical castration of aged mice results in a complete restoration of thymic size and function to prepubertal levels, and mice castrated pre-BMT restore thymopoiesis and peripheral T-cell numbers more rapidly than sham-castrated recipients.32-36  Although physical castration has been proven effective in the murine model, methods of chemically induced castration are more directly translatable to the human BMT setting. Disrupting sex steroid production using a luteinizing hormone-releasing hormone agonist (LHRH-A) rapidly results in long-lasting changes in sex steroids similar to that of surgical castration.37  Leuprolide acetate (Lupron) is a potent LHRH-A that is currently used in the clinic to treat prostate cancer, and LHRH-A has been tested as a single agent in a pilot study of autologous and allogeneic hematopoietic stem cell transplant recipients and shown to increase levels of naive CD4+ T cells in the periphery in a cohort of patients.38 

The receptor distribution of FGFR2-IIIb and ARs on TECs indicates the potential for additive effects from combined treatment with KGF and leuprolide acetate. We hypothesized that pre-BMT androgen blockade via chemical castration could act in an additive fashion with KGF to enhance thymic recovery and T-cell reconstitution in allogeneic BMT recipients. This study focuses on 2 currently FDA-approved agents (recombinant human KGF [Kepivance] and leuprolide acetate). We report that combined pre-BMT treatment resulted in a restoration of thymic architecture, number, and subset distribution of TECs. These changes led not only to additive effects on restoring thymopoiesis, thymic output, and recovery of peripheral naive CD4 and CD8 T-cell numbers but also in vivo responses to neoantigen and challenges with a live pathogen. These findings suggest a novel, clinically translatable approach to accelerate the recovery of functional immune system recovery after BMT.

Animals

C57BL/6 (H-2b; termed B6) and [C57BL/6xBALB/c]F1 (H-2d/b; termed CB6F1) male mice were purchased from The Jackson Laboratory (Bar Harbor, ME) and used at 8 weeks of age as BMT recipients or control animals (non-BMT control mice). Donor female BALB/c (H-2d) or C57BL/6.Ly5.1 mice of the same age were purchased from the National Cancer Institute (Frederick, MD). Mice were housed in specific pathogen-free facilities. All protocols were approved by the Institutional Animal Care and Use Committee at the University of Minnesota.

KGF administration and chemical castration

Recombinant human KGF (kindly provided by Amgen, Thousand Oaks, CA) was administered subcutaneously for 3 consecutive days (5 mg/kg per day) before radiation treatment as previously reported.11  Leuprolide acetate (Lupron; TAP Pharmaceuticals, Lake Forest, IL), an LHRH agonist that ablates sex steroid hormone production, was injected as a 3-month deposition into the hind leg of B6 recipients 13 days before total body irradiation (TBI) at a dose of 0.8 mg/mouse based upon dose-response studies in the same model demonstrating equivalency in day 28 after BMT thymopoiesis in doses of 0.4, 0.8, and 1.2 mg/mouse34  (and data not shown).

BM transplantation

Single-cell suspensions of BM cells obtained from femurs and tibiae of BALB/c (allogeneic) or B6.Ly5.1 (congenic) donors were CD4/8-depleted as described previously39  and 107 (allogeneic) or 5 × 106 (congenic) CD4/8-depleted BM cells were intravenously administered to recipients that had received 11-Gy TBI from a cesium source 24 h before BMT.

Lymphocyte flow cytometry

Thymocytes, splenocytes, and lymph nodes were suspended in 2% fetal calf serum/phosphate-buffered saline (PBS), and 106 cells were incubated with appropriate fluorochrome-conjugated monoclonal antibodies (BD Pharmingen, San Jose, CA) for 30 minutes at 4°C. A total of 104-105 live events were acquired on a FACScalibur flow cytometer (BD Pharmingen) and analyzed with FlowJo software (TreeStar, San Jose, CA).

TEC analysis by FACS

TECs were isolated and analyzed as described previously.40,41  Individual thymi were removed and small capsule incisions were made, followed by gentle disruption in ice-cold RPMI 1640 medium to deplete majority of thymocytes. Thymi were then incubated twice for 15 minutes in 0.1% collagenase-D plus 0.125% DNase-I (Roche, Indianapolis, IN), followed by incubation for 30 minutes in 0.1% collagenase/dispase plus 0.125% DNase-I (Roche). Supernatants from the final 2 enzymatic digestions were pooled and analyzed with the following antibodies: CD45-PerCp, Ly51/CDR1-fluorescein isothiocyanate, major histocompatibility complex-II-phycoerythrin (MHC-II-PE; BD Pharmingen), and biotinylated-Ulex-europaeus-agglutinin-1 (UEA-1; Vector Labs, Burlingame, CA) plus streptavidin-conjugated-Cy5 (Invitrogen, Carlsbad, CA). Anti-AIRE antibody was a generous gift of H. Scott (Monash University, Victoria, Australia) and was detected with mouse anti-rat IgG2c-Cy5 (BD Pharmingen).

BrdU incorporation assays

Mice were injected intraperitoneally with 200 μL of a 5 mg/mL solution of 5-bromo-2-deoxyuridine (BrdU)/PBS on days 3, 10, or 22 after BMT and subsequently given BrdU in their drinking water for 5 days (0.8 mg/mL). Recipients were killed after 5 days, and TECs or T cells were assessed for BrdU incorporation using the BrdU Flow Kit (BD Pharmingen) and the Alexa Fluor 647-conjugated monoclonal anti-BrdU antibody (clone PRB-1; Invitrogen). At least 5 × 104 CD45 (for TEC analyses) or total live events (for lymphocyte analyses) were acquired by fluorescence-activated cell sorting (FACS).

Detection of recent thymic emigrants

Anesthetized mice were injected in one thymic lobe with 10 μL of a 5 mg/mL solution of sulfosuccinimidyl-6-(biotinamido) hexanoate (sulfo-NHS-LC biotin) in PBS (Pierce, Rockford IL). After 24 hours, thymus and spleen were stained with streptavidin-conjugated-Cy5 and other markers and analyzed by flow cytometry as described previously.42 

Immunofluorescence microscopy

Intact thymi embedded in optimum cutting temperature (OCT) compound (Sakura, Tokyo, Japan) were snap-frozen in liquid nitrogen and stored at −80°C. Cryosections (7 μm) were fixed by air-drying overnight, blocking with 10% normal horse serum/PBS (Jackson Immunoresearch Laboratories, West Grove, PA), and staining with rabbit-anti-mouse cytokeratin-5 (K5) antibody (Covance, Berkeley, CA) plus a cychrome-5-conjugated goat-anti-rabbit antibody (Invitrogen) and biotinylated mouse-anti-mouse K18 (Progen Biotechnik, Heidelberg, Germany) plus Alexa Fluor 555-conjugated streptavidin (Invitrogen). Sections were mounted under a cover slip with 4,6-diamidino-2-phenylindole anti-fade solution (Invitrogen) and imaged on the following day at room temperature using an Olympus FluoView 500 Confocal Scanning Laser Microscope (Olympus, Center Valley, PA). Identification of TEC subsets according to marker expression was done as reported previously.22 

Immunization with KLH and quantification of serum immunoglobulin

Mice were injected intraperitoneally with 50 μg keyhole-limpet hemocyanin (KLH; CalBiochem, La Jolla, CA) in Complete Freund adjuvant (Sigma-Aldrich, St Louis, MO) followed 3 weeks later with 50 μg KLH in Incomplete Freund adjuvant (Sigma-Aldrich). After 7 days, peripheral blood was collected by retro-orbital bleeds and analyzed with the use of enzyme-linked immunosorbent assay (ELISA) for total and KLH-specific IgG1 levels.43 

Listeria monocytogenes infection

The recombinant L monocytogenes strains Lm-OVA and ΔactA-Lm-OVA44  (attenuated) expressing full-length chicken ovalbumin were kindly provided by Dr S. S. Way (University of Minnesota). Mice were inoculated with early logarithmic phase (OD600 of 0.1) bacteria grown in brain heart infusion broth at 37°C. Mice were injected intravenously with 106 colony-forming units (CFU) of ΔactA-Lm-OVA (primary) or 105 CFU of Lm-OVA (secondary) diluted in 200 μL PBS.

Quantification of Lm-OVA-specific CD8 T cells

MHC-I-DimerX:mouse-Ig-PE was purchased from BD Biosciences, and purified OVA257-64 (SIINFEKL) peptide was purchased from Anaspec (San Jose, CA). MHC-I-DimerX:mouse-Ig:OVA257-64 conjugates were prepared according to manufacturer's instructions (BD Biosciences). Peripheral blood was incubated with DimerX:mouse-Ig:OVA257-64-PE plus antibodies for other markers (all from BD Biosciences), and 5 × 103 donor CD8 T cells were collected and analyzed by flow cytometry.

Determination of L monocytogenes CFU

Mice were immunized intravenously with 105 CFU and rechallenged with 2 × 106 CFU of Lm strain 2C45  diluted in 200 μL PBS. Four days after secondary infection, livers were removed and homogenized in 0.05% Triton X-100/PBS (Sigma-Aldrich). Serial dilutions were plated onto brain-heart infusion plates, and Lm colonies were enumerated after 24 to 48 hours at 37°C.

Statistical analysis

Differences between treated and untreated BMT groups were analyzed by a 2-tailed, paired Student t test with unequal distribution.

Combined pretreatment with KGF and leuprolide acetate additively restored thymopoiesis soon after allogeneic BMT

KGF administered before TBI and BMT enhanced thymopoiesis and peripheral T-cell reconstitution after BMT; maximal benefit was observed in the periphery 2 months after BMT.11  To determine whether KGF and androgen blockade before BMT could act in an additive fashion to more rapidly restore thymopoiesis, allogeneic murine BMT recipients of rigorously T cell–depleted BM cells were either left untreated (BMT control) or were pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate before transplant. On day 28 after BMT, total thymocyte cellularity was reduced by approximately 50% in BMT control mice compared with age/sex-matched, unmanipulated B6 control animals (non-BMT Control; Figure 1A). Mice treated with KGF or leuprolide acetate abrogated the reduction in thymocyte cellularity seen in BMT control mice. Combined treatment with KGF + leuprolide acetate showed an additive increase in thymic cellularity to numbers that were significantly greater than untreated or KGF-/leuprolide acetate-treated BMT recipients and non-BMT control mice (Figure 1A). The relative distribution of thymocyte subsets was not affected by any treatment with KGF and/or leuprolide acetate; therefore, concomitant increases in the DN, DP, CD4+ SP, and CD8+ SP subsets were observed (Figure 1A-E). Additive effects of KGF + leuprolide acetate were maintained through at least day 56 after BMT (Figure S1A-E, available on the Blood website; see the Supplemental Materials link at the top of the online article).

Figure 1

Pretreatment with KGF combined with androgen blockade additively restored thymopoiesis early after allogeneic BMT. Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and analyzed for thymocyte cellularity at day 28 after BMT alongside age-/sex-matched, unmanipulated B6 control animals (non-BMT Control). Data shown are mean absolute numbers (± SEM) of (A) total thymocytes and of the thymocyte subsets: (B) CD8CD4 double-negative, (C) CD8+CD4+ double-positive, (D) CD4+CD8 single-positive, and (E) CD4CD8+ single-positive. The data are representative of 4 independent experiments with 4 to 5 mice per group. *P < .05 compared with untreated BMT recipients; #P < .05 compared with KGF-treated or leuprolide acetate-treated BMT recipients.

Figure 1

Pretreatment with KGF combined with androgen blockade additively restored thymopoiesis early after allogeneic BMT. Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and analyzed for thymocyte cellularity at day 28 after BMT alongside age-/sex-matched, unmanipulated B6 control animals (non-BMT Control). Data shown are mean absolute numbers (± SEM) of (A) total thymocytes and of the thymocyte subsets: (B) CD8CD4 double-negative, (C) CD8+CD4+ double-positive, (D) CD4+CD8 single-positive, and (E) CD4CD8+ single-positive. The data are representative of 4 independent experiments with 4 to 5 mice per group. *P < .05 compared with untreated BMT recipients; #P < .05 compared with KGF-treated or leuprolide acetate-treated BMT recipients.

Close modal

Because both donor-derived and residual host-derived thymocytes might contribute to thymic cellularity early after BMT, we assessed thymocyte chimerism. On day 28 after BMT, more than 95% of thymocytes were of donor origin, increasing to more than 99% by day 56 (data not shown). Therefore, increased thymic cellularity was due to increased numbers of donor-derived thymocytes and not to preservation of host-derived thymocytes.

Combined pretreatment with KGF and leuprolide acetate maximally restored TECs after BMT

To determine whether protection/restoration of TECs was responsible for the enhancement of thymopoietic recovery observed on day 28 after BMT, absolute numbers of TECs, defined as CD45MHC-II+ cells, were quantified in thymi of allogeneic BMT recipients left untreated or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and compared with non-BMT control mice. Regardless of pretreatment, TECs were severely depleted as early as day 7 after BMT (>75%), and TEC numbers remained significantly reduced through day 14 after BMT compared with non-BMT control mice (data not shown). On day 26 after BMT, all BMT groups still had reduced total TEC numbers compared with non-BMT control mice except for KGF + leuprolide acetate-treated BMT recipients that had restored TECs to levels above untreated BMT recipients and to similar numbers compared with non-BMT control mice (Figure 2A). With the exception of leuprolide acetate-treated BMT recipients, cortical TECs (cTEC; CD45MHC-II+Ly51+) were restored to normal levels at day 26 (Figure 2B). Medullary TECs (mTECs; CD45MHC-II+Ly51) remained depleted in all groups except for the KGF + leuprolide acetate BMT recipients, which had similar mTEC numbers compared with non-BMT control mice (Figure 2C).

Figure 2

Combined pretreatment with KGF and androgen blockade maximally restores numbers of totalTECsand mTEC subsets by day 26afterBMT. (A-F) Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and analyzed for absolute numbers of TEC and TEC subsets at day 26 after BMT. Single-cell suspensions were prepared from enzymatic digests of individual thymi and used to determine total thymic epithelial cells (TEC; CD45MHC-II+) (A), cortical TEC (cTEC; CD45MHC-II+Ly51+) (B), medullary TEC (mTEC; CD45MHC-II+Ly51) (C), UEA-1+ mTEClo (CD45MHC-IIlowLy51 UEA-1+) (D), UEA-1+ mTEChi (CD45MHC-IIhighLy51 UEA-1+) (E), AIRE+ mTEChi (CD45MHC-IIhighLy51 AIRE+) (F). (G-J) Total TECs and individual TEC subsets were assessed for proliferation by continuous administration of BrdU in the drinking water (0.8 mg/mL) between day 10 and 14 after BMT at which point Total TECs (G), cTEC (H), mTEClo (I), and mTEChi subsets (J) were analyzed for BrdU incorporation. Data shown are the mean numbers of TEC (± SEM) or mean percentages of BrdU+ TEC (± SEM) and are representative of one experiment of 4 mice per group. *P < .05 compared with BMT control mice; #P < .05 compared with KGF- or leuprolide acetate-treated BMT recipients.

Figure 2

Combined pretreatment with KGF and androgen blockade maximally restores numbers of totalTECsand mTEC subsets by day 26afterBMT. (A-F) Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and analyzed for absolute numbers of TEC and TEC subsets at day 26 after BMT. Single-cell suspensions were prepared from enzymatic digests of individual thymi and used to determine total thymic epithelial cells (TEC; CD45MHC-II+) (A), cortical TEC (cTEC; CD45MHC-II+Ly51+) (B), medullary TEC (mTEC; CD45MHC-II+Ly51) (C), UEA-1+ mTEClo (CD45MHC-IIlowLy51 UEA-1+) (D), UEA-1+ mTEChi (CD45MHC-IIhighLy51 UEA-1+) (E), AIRE+ mTEChi (CD45MHC-IIhighLy51 AIRE+) (F). (G-J) Total TECs and individual TEC subsets were assessed for proliferation by continuous administration of BrdU in the drinking water (0.8 mg/mL) between day 10 and 14 after BMT at which point Total TECs (G), cTEC (H), mTEClo (I), and mTEChi subsets (J) were analyzed for BrdU incorporation. Data shown are the mean numbers of TEC (± SEM) or mean percentages of BrdU+ TEC (± SEM) and are representative of one experiment of 4 mice per group. *P < .05 compared with BMT control mice; #P < .05 compared with KGF- or leuprolide acetate-treated BMT recipients.

Close modal

Within the medulla, 2 distinct mTEC subpopulations can be identified based upon MHC-II expression and binding to the lectin Ulex Europaeus Agglutinin-1 (UEA-1)–mTEClo (CD45MHC-IIloLy51UEA-1+) and mTEChi (CD45MHC-IIhiLy51UEA-1+).40,41  Both mTEC populations were reduced in number by 50% or more in untreated, KGF- or leuprolide acetate-treated BMT recipients compared with non-BMT control mice on day 26 after BMT (Figure 2D,E). However, KGF + leuprolide acetate treatment resulted in mTEClo and mTEChi numbers comparable with those of non-BMT control mice and 1.5-fold (mTEClo) and 4-fold (mTEChi) higher than untreated BMT control mice (Figure 2D,E). Numbers of AIRE+ mTEChi, known to be critical for the negative selection of thymocytes with autoreactive specificities,46  were significantly reduced (∼3-fold) in untreated and leuprolide acetate-treated BMT recipients compared with non-BMT control mice. In KGF- and KGF + leuprolide acetate-treated BMT recipients, AIRE+ mTEChi numbers were restored to levels similar to those of non-BMT control mice (Figure 2F). Medullary dendritic cells in the thymus, also involved in negative selection,46  were severely depleted after BMT and were significantly and maximally restored by day 26 after BMT in KGF + leuprolide acetate-treated BMT recipients (data not shown).

To determine whether the increased numbers of TECs observed on day 26 after BMT were due to enhanced TEC proliferation at an earlier time point, mice were injected between days 10 and 14 after BMT with the thymidine analog BrdU, which is incorporated into DNA of replicating cells. In agreement with the relative TEC numbers observed on day 26 after BMT, total TECs, cTECs, and mTEChi showed the greatest levels of proliferation in the thymi of BMT recipients treated with KGF + leuprolide acetate (Figure 2G-J).

In addition to the loss of TECs, immunofluorescence microscopy showed a loss of clear compartmentalization of cortex and medulla in the thymi of untreated BMT recipients on day 28 after BMT (Figure S2). In contrast, non-BMT control mice displayed well organized and densely clustered cortical and medullary regions with a distinct corticomedullary boundary, a finding largely recapitulated in KGF + leuprolide acetate-treated BMT recipients (Figure S2). All groups had restored thymic architecture by day 56 after BMT (Figure S2).

Pretreatment with KGF and leuprolide acetate resulted in enhanced T-cell reconstitution in the lymph nodes and spleen by day 35 after BMT

To determine whether improved thymopoiesis induced by KGF and leuprolide acetate resulted in higher numbers of peripheral T cells, lymph nodes of BMT recipients were analyzed for CD4+ and CD8+ T cells on day 35 after BMT. CD4+ and CD8+ T-cell numbers in untreated BMT recipients were reduced approximately 50% and approximately 75%, respectively, compared with non-BMT control mice and were partially restored in BMT recipients pretreated with KGF or leuprolide acetate alone (Figure 3A,C). Combined, these treatments provided an additive increase in CD4+ and CD8+ T cells that was significantly greater than either agent alone (Figure 3A,C), completely eliminating CD4+ and virtually eliminating CD8+ T-cell lymphopenia. Because small numbers of radioresistant host-derived peripheral T cells still remained at this time point, we specifically quantified naive, donor-derived, CD4+ and CD8+ T cells to assess the contribution of thymus-derived T cells to reconstitution of peripheral T cells. Additive increases of donor-derived, naive CD4+ (H2Kd+CD4+CD45RBhiCD44lo) and CD8+ (H2Kd+CD8+CD62LhiCD44lo) T cells were observed in BMT recipients treated with KGF + leuprolide acetate that were significantly greater than was observed in untreated, KGF-treated, and leuprolide acetate-treated BMT recipients, respectively (Figure 3B,D). By day 60 after BMT, CD4+ T-cell compartments in untreated BMT recipients were restored to their non-BMT control levels, whereas leuprolide acetate- and KGF + leuprolide acetate treated BMT recipients maintained comparatively higher numbers of total and naive CD4+ T cells (Figure S3A,B). Donor-derived naive CD8+ T cells remained significantly reduced through day 60 after BMT in all BMT groups (compared with non-BMT control mice), except those pretreated with KGF + leuprolide acetate (Figure S3C,D).

Figure 3

Combined pretreatment with KGF and androgen blockade significantly restore numbers of total and donor-derived naive CD4+ and CD8+ T cells in lymph node and spleen by day 35 after BMT. Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and analyzed for the presence of T cells in the lymph nodes and spleen at day 35 after BMT alongside unmanipulated age/sex-matched B6 control animals (non-BMT Control). Mean absolute num-bers (± SEM) of total CD4+ T cells (A,E), naive (CD45RBhighCD44low) CD4+ T cells (B,F), total CD8+ T cells (C,G), and naive (CD62LhighCD44low) CD8+ T cells (D,H) in the lymph nodes and spleen are shown. Lymph node data are represented here by pooled cells from 2 inguinal, 2 axillary, and mesenteric lymph nodes. Data are representative of 3 experiments, each with 4 mice per group; *P < .05 compared with BMT control mice; #P < .05 compared with KGF-treated BMT recipients.

Figure 3

Combined pretreatment with KGF and androgen blockade significantly restore numbers of total and donor-derived naive CD4+ and CD8+ T cells in lymph node and spleen by day 35 after BMT. Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and analyzed for the presence of T cells in the lymph nodes and spleen at day 35 after BMT alongside unmanipulated age/sex-matched B6 control animals (non-BMT Control). Mean absolute num-bers (± SEM) of total CD4+ T cells (A,E), naive (CD45RBhighCD44low) CD4+ T cells (B,F), total CD8+ T cells (C,G), and naive (CD62LhighCD44low) CD8+ T cells (D,H) in the lymph nodes and spleen are shown. Lymph node data are represented here by pooled cells from 2 inguinal, 2 axillary, and mesenteric lymph nodes. Data are representative of 3 experiments, each with 4 mice per group; *P < .05 compared with BMT control mice; #P < .05 compared with KGF-treated BMT recipients.

Close modal

Similar to the lymph node findings, treatment with KGF + leuprolide acetate resulted in complete reconstitution of splenic T-cell numbers at day 35 after BMT. Whereas there were highly significant (>4-fold) reductions in total CD4+ and CD8+ T cells in the spleen of untreated BMT recipients compared with non-BMT control mice (Figure 3E,G), combined treatment completely prevented the state of T-cell lymphopenia, resulting in the highest increase in total CD4+ and CD8+ T cells, which appeared additive for total CD8+ T cells but not for total CD4+ T cells (Figure 3E,G). Compared with BMT control mice, naive, donor-derived CD4+ T cells were significantly increased by KGF (3-fold), leuprolide acetate (4-fold), and KGF + leuprolide acetate (5-fold), reaching levels that were no longer significantly lower than those in non-BMT control mice (Figure 3F). Significant increases in naive, donor-derived CD8+ T cells were observed with KGF or leuprolide acetate (3-fold) and KGF + leuprolide acetate (5-fold) compared with untreated BMT recipients (Figure 3H). Total and naive donor-derived CD8+ T-cell numbers remained deficient through day 60 after BMT; only the combined treatment resulted in a significant restoration of these cells compared with untreated control animals (Figure S3G,H). These data demonstrate that combined treatment with KGF + leuprolide acetate before BMT additively and durably enhanced recovery of donor-derived naive CD4+ and CD8+ T cells in both lymph nodes and spleen after BMT. Analysis of TCR Vβ repertoire by flow cytometric analysis representing 12 Vβ alleles confirmed that KGF and leuprolide acetate did not affect the diversity of TCR Vβ usage in donor-derived T cells compared with non-BMT control mice (Figure S4).

Combined pretreatment with KGF and leuprolide acetate resulted in enhanced thymic output and less homeostatic proliferation early after BMT

Because KGF and leuprolide acetate each may increase peripheral T-cell expansion, we sought to determine the relative contributions of thymic export and peripheral homeostatic expansion to the observed increased numbers of total and naive peripheral CD4+ and CD8+ T cells found in secondary lymphoid organs. Thymic export was quantified at day 28 after BMT by detection of intrathymically biotin-labeled donor-derived T cells that had recently emigrated to the spleen.42  Consistent with higher naive splenic CD4+ T-cell numbers, we observed a significant and almost significant increase in the export of donor-derived CD4+ T cells into the periphery in BMT recipients treated with KGF and leuprolide acetate, respectively, compared with the untreated BMT control mice (Figure 4A). Although thymic export of donor-derived CD8+ T cells was not significantly improved with KGF or leuprolide acetate alone, there was a significant and additive increase in the export of donor-derived CD4+ and CD8+ T cells into the periphery in KGF + leuprolide acetate-treated BMT recipients on day 28 after BMT, consistent with higher naive CD4+ and CD8+ T-cell numbers in this group (Figure 4A,B).

Figure 4

Enhanced thymocyte cellularity in KGF + leuprolide acetate-treated BMT recipients correlates with increased T-cell export into the periphery and diminished homeostatic proliferation early after BMT. (A,B) Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow that were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate were intrathymically injected (one thymic lobe) with biotin at 28 days after BMT. After 24 hours of in vivo labeling, the export of thymus-derived CD4 (A) and CD8 (B) T cells into the periphery was assessed by staining total splenocytes with fluorescently labeled streptavidin in combination with monoclonal antibodies specific for CD4 and CD8. Absolute numbers of recent thymic emigrants (RTE) were normalized to absolute numbers of donor-derived CD4+ or CD8+ T cells in panels A and B, respectively. (C-F) Homeostatic proliferation in the CD4+ and CD8+ T-cell compartment was also assessed between days 30 and 35 after BMT. Treated and untreated BMT recipients and non-BMT control mice were injected intraperitoneally with BrdU (1 mg) on day 30, followed by continuous administration of BrdU in the drinking water (0.8 mg/mL) through day 35 after BMT, at which point CD4+ and CD8+ T cells were analyzed for BrdU incorporation and concomitant up-regulation of CD44, thus indicating that proliferation had occurred within the 5-day labeling window. Data shown are the mean percentages of BrdU+CD44+ (± SEM) of total CD4+ T cells (C,E) and CD8+ T cells (D,F) isolated from the spleens and lymph nodes of these mice. These data are representative of one experiment with 4 mice per group; *P < .05 compared with BMT control mice; #P < .05 compared with KGF- or leuprolide acetate-treated BMT recipients.

Figure 4

Enhanced thymocyte cellularity in KGF + leuprolide acetate-treated BMT recipients correlates with increased T-cell export into the periphery and diminished homeostatic proliferation early after BMT. (A,B) Lethally irradiated B6 recipients of allogeneic (BALB/c) bone marrow that were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate were intrathymically injected (one thymic lobe) with biotin at 28 days after BMT. After 24 hours of in vivo labeling, the export of thymus-derived CD4 (A) and CD8 (B) T cells into the periphery was assessed by staining total splenocytes with fluorescently labeled streptavidin in combination with monoclonal antibodies specific for CD4 and CD8. Absolute numbers of recent thymic emigrants (RTE) were normalized to absolute numbers of donor-derived CD4+ or CD8+ T cells in panels A and B, respectively. (C-F) Homeostatic proliferation in the CD4+ and CD8+ T-cell compartment was also assessed between days 30 and 35 after BMT. Treated and untreated BMT recipients and non-BMT control mice were injected intraperitoneally with BrdU (1 mg) on day 30, followed by continuous administration of BrdU in the drinking water (0.8 mg/mL) through day 35 after BMT, at which point CD4+ and CD8+ T cells were analyzed for BrdU incorporation and concomitant up-regulation of CD44, thus indicating that proliferation had occurred within the 5-day labeling window. Data shown are the mean percentages of BrdU+CD44+ (± SEM) of total CD4+ T cells (C,E) and CD8+ T cells (D,F) isolated from the spleens and lymph nodes of these mice. These data are representative of one experiment with 4 mice per group; *P < .05 compared with BMT control mice; #P < .05 compared with KGF- or leuprolide acetate-treated BMT recipients.

Close modal

To correlate thymic output with the degree of homeostatic proliferation (HP) occurring in peripheral lymphoid organs, BrdU was given continuously in the drinking water between days 30 and 35 after BMT. BrdU incorporation with up-regulation of CD44 in CD4+ and CD8+ T cells in the spleen and lymph nodes was measured. An inverse relationship was observed between the number of recently exported CD4+ and CD8+ T cells into the periphery around day 28 after BMT and the degree of HP detected in peripheral lymphoid organs between days 30 and 35 after BMT (Figure 4C-E). The higher donor-derived naive CD4+ and CD8+ T-cell numbers are probably due to increased thymic output rather than higher levels of HP.

Pretreatment with KGF and leuprolide acetate enhanced T cell–dependent antibody responses early after BMT

The prolonged CD4+ T-cell deficiency after BMT can preclude the generation of normal immune responses to T cell–dependent B-cell antigens.47  Because KGF + leuprolide acetate-treated BMT recipients had normalization of donor naive CD4+ T cells and donor B cells by day 35 after BMT (data not shown), we sought to determine whether this treatment could enhance a humoral immune response against a T cell–dependent neoantigen challenge given on day 28 after BMT. Mice were immunized with KLH plus adjuvant, rechallenged 2 weeks later with KLH, and serum immunoglobulin levels were measured after 7 days. Total and KLH-specific IgG1 levels were approximately 50% lower in untreated BMT recipients compared with non-BMT control mice, and pretreatment with KGF or leuprolide acetate only marginally improved T cell–dependent B-cellisotype switching as measured by anti-KLH specific IgG1 antibody levels (Figure 5A,B). BMT recipients treated with KGF + leuprolide acetate produced significantly greater amounts of total and KLH-specific IgG1 antibody, reaching levels comparable with non-BMT control mice (Figure 5A,B). The early increases of peripheral donor T cells and B cells in BMT recipients pretreated with KGF + leuprolide acetate permitted the normal generation of a CD4+ T cell–dependent B-cell immunoglobulin isotype-switched response to KLH even when given as early as day 28 after BMT.

Figure 5

KGF treatment and androgen blockadebeforeBMT enhance the secondary humoral immune response to KLH after allogeneic BMT. Lethally irradiated C57BL/6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and immunized at day 28 after BMT with 50 μg of KLH in Complete Freund's adjuvant (KLH/CFA) alongside unmanipulated age/sex-matched C57BL/6 control animals (non-BMT Control). Two weeks after primary immunization (equivalent to day 42 after BMT), mice were rechallenged with 50 μg of KLH in Incomplete Freund's adjuvant (KLH/IFA). Serum was then collected after 7 days and analyzed with the use of ELISA for total IgG1 (A) and KLH-specific IgG1 (B) antibody levels. Data shown are mean micrograms of IgG1 per milliliter of serum (± SEM) from one experiment with 4 mice per group; *P < .05 compared with untreated BMT control mice.

Figure 5

KGF treatment and androgen blockadebeforeBMT enhance the secondary humoral immune response to KLH after allogeneic BMT. Lethally irradiated C57BL/6 recipients of allogeneic (BALB/c) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and immunized at day 28 after BMT with 50 μg of KLH in Complete Freund's adjuvant (KLH/CFA) alongside unmanipulated age/sex-matched C57BL/6 control animals (non-BMT Control). Two weeks after primary immunization (equivalent to day 42 after BMT), mice were rechallenged with 50 μg of KLH in Incomplete Freund's adjuvant (KLH/IFA). Serum was then collected after 7 days and analyzed with the use of ELISA for total IgG1 (A) and KLH-specific IgG1 (B) antibody levels. Data shown are mean micrograms of IgG1 per milliliter of serum (± SEM) from one experiment with 4 mice per group; *P < .05 compared with untreated BMT control mice.

Close modal

Pretreatment with KGF and leuprolide acetate enhances functional immune response against L monocytogenes after BMT

To determine whether improved immune reconstitution induced by pre-BMT treatment with KGF and/or leuprolide acetate would permit a functional immune response to challenge with a live intracellular pathogen after BMT, mice were immunized with 106 CFU of an attenuated strain of L monocytogenes. To monitor pathogen specific immune responses, a congenic BMT system was used with L monocytogenes (Lm) engineered to express chicken ovalbumin (ΔactA-Lm-OVA). Thus, activated Lm-specific CD8 T-cell responses were quantifiable in peripheral blood by MHC class-I:OVA tetramer binding to donor-derived CD44+ CD8 T cells (Ly5.1+CD8+CD44+Kb:OVA257-264+). Seven days after primary infection, KGF- or leuprolide acetate-treated BMT recipients contained modestly higher numbers of donor-derived, OVA-specific CD44+ CD8 T cells compared with untreated BMT recipients, and treatment with KGF + leuprolide acetate resulted in additively higher numbers that were significantly greater than in all other BMT groups (Figure 6A). Postmortem necroscopic analysis revealed that all BMT and non-BMT groups had effectively cleared this attenuated strain by 3 weeks after infection (data not shown).

Figure 6

Combined pretreatment with KGF and androgen blockade before BMT significantly improves CD8 T-cell responses against L monocytogenes after allogeneic BMT. Lethally irradiated C57BL/6 Ly5.2+ recipients of congenic (C57BL/6 Ly5.1+) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and immunized at day 42 after BMT alongside unmanipulated age/sex-matched B6 control animals (non-BMT Control). For primary immunization, 106 CFU of an attenuated strain of L monocytogenes that express recombinant full-length chicken ovalbumin (ΔactA-Lm-OVA) was intravenously injected. (A) Absolute numbers of donor-derived Ly5.1+CD44+CD8+ Kb-OVA257-64–specific T cells were quantified in peripheral blood of infected animals by FACS 7 days after primary infection. (B) Immunized mice were then rechallenged with 105 CFU of the virulent parent strain, Lm-OVA, 42 days after primary infection. After 3 days, isolated splenocytes of infected animals were restimulated ex vivo for 5 h with OVA257-64, and donor-derived CD8 T cells were analyzed by FACS for IFNγ production. *P < .05 compared with untreated BMT control mice; #P < .05 compared with KGF- and leuprolide acetate–treated BMT recipients.

Figure 6

Combined pretreatment with KGF and androgen blockade before BMT significantly improves CD8 T-cell responses against L monocytogenes after allogeneic BMT. Lethally irradiated C57BL/6 Ly5.2+ recipients of congenic (C57BL/6 Ly5.1+) bone marrow were left untreated (BMT Control) or pretreated with KGF, leuprolide acetate, or KGF + leuprolide acetate and immunized at day 42 after BMT alongside unmanipulated age/sex-matched B6 control animals (non-BMT Control). For primary immunization, 106 CFU of an attenuated strain of L monocytogenes that express recombinant full-length chicken ovalbumin (ΔactA-Lm-OVA) was intravenously injected. (A) Absolute numbers of donor-derived Ly5.1+CD44+CD8+ Kb-OVA257-64–specific T cells were quantified in peripheral blood of infected animals by FACS 7 days after primary infection. (B) Immunized mice were then rechallenged with 105 CFU of the virulent parent strain, Lm-OVA, 42 days after primary infection. After 3 days, isolated splenocytes of infected animals were restimulated ex vivo for 5 h with OVA257-64, and donor-derived CD8 T cells were analyzed by FACS for IFNγ production. *P < .05 compared with untreated BMT control mice; #P < .05 compared with KGF- and leuprolide acetate–treated BMT recipients.

Close modal

To investigate the effects of KGF and leuprolide acetate on a secondary immune response against Lm-OVA, mice were rechallenged 42 days after primary infection with 105 CFU of the nonattenuated parent strain, Lm-OVA. Three days later, we restimulated splenocytes from infected animals ex vivo with soluble OVA257-264 and found that only KGF + leuprolide acetate-treated BMT recipients demonstrated a comparable number of interferon-γ (IFNγ)–positive CD8 T cells as non-BMT control mice, at least 2-fold higher than all other groups (Figure 6B).

In separate experiments, untreated, KGF-, leuprolide acetate- or KGF + leuprolide acetate-treated allogeneic BMT recipients were assessed for clearance of Lm as measured by CFU determination in livers of infected animals 4 days after secondary infection. Untreated BMT recipients contained significantly higher bacterial burdens than non-BMT control mice and KGF or leuprolide acetate administered as single agents before BMT resulted in a marginal benefit for decreasing liver CFU (Figure S5). It is noteworthy that KGF + leuprolide acetate-pretreated BMT recipients significantly reduced the CFU burden from the liver to levels that were comparable with those in non-BMT control mice (Figure S5).

We report a novel therapy given entirely before the BMT conditioning regimen that combines the administration of KGF and the LHRH agonist (leuprolide acetate) to enhance thymopoiesis and peripheral T-cell recovery and function in BMT recipients. Compared with no treatment in BMT reccipients, KGF + leuprolide acetate treatment additively increased thymocyte and peripheral T-cell recovery. Thymic architecture and TECs were significantly restored in KGF + leuprolide acetate-treated recipients early after BMT, improving peripheral donor-derived T-cell reconstitution as a result of increased thymic output. KGF + leuprolide acetate-pretreated BMT recipients mounted a superior immune response to the neoantigen KLH and cleared a live infection with Lm more effectively than other treatment groups.

In rodents and nonhuman primate models of chemoradiotherapy and BMT, KGF pretreatment has been shown to augment thymopoiesis in an interleukin-7 (IL-7)–dependent fashion.11,21,48  Because TECs are KGFR+, the putative mechanism of action proposed was stimulation of TEC proliferation and/or repair from radiation-induced injury.11  In mice that did not receive transplants, KGF administration increased TEC numbers.14  Thus, KGF may induce the expansion of a TEC progenitor population that, in turn, may rapidly mature to restore TEC numbers after BMT conditioning. Androgen blockade by physical castration can reverse the thymic atrophy accompanying aging and can improve thymopoietic recovery in rodents undergoing allogeneic BMT.32-34,36  Although our experiments focused on male mice, gonadal hormone blockade should be effective in female subjects, because leuprolide acetate has been successfully used to treat hormone-related disorders in women.49  ARs are expressed on TECs, non-TEC thymic stromal cells, thymocytes, and peripheral T cells,36  although chimeric studies have indicated that the restorative effect of castration on the thymus required AR expression on TECs.27,28  Because castration-induced enhancement of thymopoietic recovery after BMT was KGF-independent,36  these data suggest that each agent acts through distinct mechanisms.

Thymic cellularity is tightly controlled by the availability of thymic stromal niches to support thymocyte maturation.9,50  Although studies have demonstrated TBI-induced TEC depletion,10-12  this is the first to quantitatively analyze TEC subset depletion and recovery by FACS after TBI and BMT. TBI-induced TEC depletion is not surprising in light of data illustrating that TEC, especially mTEC, are dynamically proliferating in the steady state.46  The equivalent loss of TEC observed in all BMT groups early after BMT (day 7, day 14) indicates that KGF + leuprolide acetate accelerates the restoration rather than the protection of TEC compartment from TBI-induced depletion. KGF and leuprolide acetate appeared to act in concert to restore more rapidly TEC numbers without disrupting thymic architecture and may have provided for a larger stromal scaffold to support thymopoiesis. These proliferation events may have occurred early after BMT based upon earlier studies demonstrating maximal TEC proliferation at 1-3 days after discontinuation of KGF.14  KGF has been shown to be mitogenic for both cTEC and mTEC and can act to restore the thymic disorganization associated with aging and BMT14  (G.A.H., unpublished data, August 2007). A large percentage of UEA-1+ mTEC express FGFR2-IIIb.14  Although expression patterns of ARs on TEC subsets have not been described, androgen removal is mitogenic for mTEC.51  It is thought that mTEChi encompass mature, postmitotic TECs that are derived from immature mTEClo.46  Our findings of a large number of BrdU+ mTEChi in KGF + leuprolide acetate-treated BMT recipients are consistent with the interpretation that these cells are the progeny of mTEClo recovering from depletion and maturing to reconstitute mTEChi cells. Thus, KGF + leuprolide acetate appears to maximally enhance mTEChi regeneration after BMT, including the AIRE+ subset, critical for the expression of tissue-restricted antigens needed for negative repertoire selection of autoreactive T-cell clones during thymopoiesis.52 

The additive increase in thymic cellularity in recipients pretreated with KGF + leuprolide acetate correlated with increased thymic export of T cells into the periphery. Previous studies have shown that KGF treatment and androgen blockade (physical castration), when analyzed individually, could increase thymic cellularity and enhance thymic output.14,35,36  In mice that did not receive transplants, thymic export is a direct function of thymic cellularity.53  Our data suggest that the same relationship holds true after BMT, even though the thymus is damaged and exports T cells into a lymphopenic environment. In agreement with an enhanced thymic output in KGF + leuprolide acetate-treated mice, we observed by day 35 after BMT additive increases in donor-derived naive CD4 and CD8 T cells in both spleen and lymph node. Because T cell–depleted BM was used, all donor-derived T cells with a naive surface phenotype must have been generated in the thymus in this transplant model. The BMT models used in these studies do not result in GVHD clinically or histologically. Because androgen blockade by physical castration pre-BMT does not exacerbate GVHD in other models and KGF has been shown to ameliorate thymic damage and lethality as a result of GVHD,22,23,36  KGF + leuprolide acetate may represent a viable clinical approach to speeding immune reconstitution even in the setting of GVHD risk.

Higher export of naive T cells in KGF + leuprolide acetate-treated BMT recipients also correlated with a decrease in HP observed in secondary lymphoid organs of these mice. T cells derived from lymphopenia-induced peripheral expansion or expansion driven by exogenous cytokines generally result in expansion of a limited number of peripheral T-cell clones and can create “holes” in the TCR repertoire.5,54-56  In our studies, TCR Vβ diversity was not skewed by KGF and/or leuprolide acetate. Therefore, KGF + leuprolide acetate in the clinical BMT setting may speed reconstitution of a diverse naive peripheral T-cell compartment capable of responding to a wider array of pathogens after BMT.

Poor immune responsiveness among BMT recipients leads to suboptimal responses to immunization for extended periods of time.5,47  In myeloablated recipients of autologous hematopoietic grafts, KLH responses were decreased up to 16 months.47  Although all BMT groups mounted similar primary anti-KLH IgM responses (data not shown), indicating that B-cell function was sufficient, only KGF + leuprolide acetate provided maximal benefit for enhancing T:B cooperativity with higher total IgG1 and KLH-specific IgG1 antibody titers indicative of isotype switching mechanisms requiring CD4 T-cell help.57  The demonstration that pretreatment with KGF + leuprolide acetate improved the T cell–dependent antibody response suggests a potential for KGF and leuprolide acetate as adjunct to immunization in BMT recipients.

Low peripheral CD4 and CD8 T-cell numbers results in poor immune responsiveness and increased susceptibility to and severity of infection among BMT recipients.7,58  Likewise, we observed a severe defect in the ability of untreated murine BMT recipients to clear Lm from the liver, whereas KGF + leuprolide acetate-pretreated BMT recipients cleared Lm from these sites as efficiently as non-BMT control mice. Consistent with this defective clearance, untreated BMT recipients showed less than 50% of the total number of Lm-reactive CD8 T cells compared with non-BMT control mice and BMT recipients pretreated with KGF + leuprolide acetate after a secondary infection. This same trend was observed when analyzing the percentage of IFNγ+ cells within the Kb:OVA-reactive CD8 T-cell compartment (data not shown). Because adaptive immunity against many intracellular pathogens involve highly conserved T-cell responses,59-61  the enhanced Lm immunity would likely be extended to a wider spectrum of viral and intracellular bacterial pathogens that pose a serious threat to BMT recipients. In this regard, we have shown that KGF-treated nonhuman primate BMT recipients develop improved responses to a challenge with simian immunodeficiency virus compared with untreated BMT recipients.48  T-cell responsiveness to CD3 signals also has been shown to be drastically diminished after BMT compared with that in healthy control animals, and cytomegalovirus reactivation after BMT has been associated with dysfunctional antigen-specific CD8 T cells.58,62  In contrast, peripheral T cells from castrated mice are hyperresponsive to antigen and CD3/28 signaling early after castration, although this effect is lost by 7 weeks after castration.35  Although heightened sensitivity to antigenic stimulation of T cells after androgen blockade may have played a role in driving early peripheral T-cell reconstitution, it is unlikely to be a major contributor to improved CD8 T-cell responses to Lm late after BMT.

In summary, combined KGF and androgen blockade was highly effective in speeding thymic recovery and peripheral T-cell reconstitution after BMT. Treated BMT recipients were resistant to a pathogenic challenge and were capable, relatively early after engraftment, of T:B-cell cooperativity in generating antibody responses to a neoantigen. Because KGF and leuprolide acetate are being used as single agents in the context of human BMT, our data suggest that future clinical trials designed to determine the safety and efficacy of combining these agents for facilitating immune recovery after BMT.

An Inside Blood analysis of this article appears at the front of this issue.

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.

We thank Chris Contag (Stanford University, Stanford, CA) for kindly providing L monocytogenes strain 2C and Hamish Scott (Monash University, Victoria, Australia) for providing the anti-AIRE antibody. We thank Emily Goren and Luna Liu for excellent animal care, Ryan Fremming for performing the ELISA analysis of KLH immune serum, and Jason Gill, Daniel Gray, Jonathan Hardy, and SingSing Way for kindly providing ΔactALmOVA and LmOVA, helpful discussions, and experimental advice.

This work was supported by National Institutes of Health grants R01-HL073794, R01-HL55209, R01-A1057477-01, and R01-A1057477-01, by SNF 3100-68310.02, by the Children's Cancer Research Fund, and by a grant from the European Community 6th Framework Program Eurothymaid Integrated Project. We would like to acknowledge the use of the confocal microscope made available through a National Center for Research Resources Shared Instrumentation Grant (1-S10-RR16851).

National Institutes of Health

Contribution: R.M.K. designed and performed research, analyzed and interpreted data, made the figures, and wrote the manuscript. S.L.H. performed research. A.P-M. provided advice and edited the manuscript. P.A.T. performed research. R.L.B. provided advice and edited the manuscript. G.A.H. advised on experimental design and edited the manuscript. B.R.B. designed research, advised on experimental design, and edited the manuscript.

Conflict-of-interest disclosure: R.L.B. is named as an inventor on patents pertaining to immune reconstitution after androgen blockade. The remaining authors declare no competing financial interests.

Correspondence: Dr Bruce R. Blazar, University of Minnesota, Room 460F, Cancer Center Research Building, 425 East River Road, Minneapolis, MN 55455; e-mail: blaza001@umn.edu.

1
Markova
 
M
Barker
 
JN
Miller
 
JS
, et al. 
Fludarabine vs cladribine plus busulfan and low-dose TBI as reduced intensity conditioning for allogeneic hematopoietic stem cell transplantation: a prospective randomized trial.
Bone Marrow Transplant
2007
, vol. 
39
 (pg. 
193
-
199
)
2
Weinberg
 
K
Blazar
 
BR
Wagner
 
JE
, et al. 
Factors affecting thymic function after allogeneic hematopoietic stem cell transplantation.
Blood
2001
, vol. 
97
 (pg. 
1458
-
1466
)
3
Douek
 
DC
Vescio
 
RA
Betts
 
MR
, et al. 
Assessment of thymic output in adults after haematopoietic stem-cell transplantation and prediction of T-cell reconstitution.
Lancet
2000
, vol. 
355
 (pg. 
1875
-
1881
)
4
Hakim
 
FT
Memon
 
SA
Cepeda
 
R
, et al. 
Age-dependent incidence, time course, and consequences of thymic renewal in adults.
J Clin Invest
2005
, vol. 
115
 (pg. 
930
-
939
)
5
Williams
 
KM
Hakim
 
FT
Gress
 
RE
T cell immune reconstitution following lymphodepletion.
Semin Immunol
2007
, vol. 
19
 (pg. 
318
-
330
)
6
Welniak
 
LA
Blazar
 
BR
Murphy
 
WJ
Immunobiology of allogeneic hematopoietic stem cell transplantation.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
139
-
170
)
7
Centers for Disease Control and Prevention
Infectious Diseases Society of America; American Society of Blood and Marrow Transplantation. Guidelines for preventing opportunistic infections among hematopoietic stem cell transplant recipients.
Biol Blood Marrow Transplant
2000
, vol. 
6
 (pg. 
659
-
713
715; 717-627; quiz 729–733
8
Starr
 
TK
Jameson
 
SC
Hogquist
 
KA
Positive and negative selection of T cells.
Annu Rev Immunol
2003
, vol. 
21
 (pg. 
139
-
176
)
9
Anderson
 
G
Lane
 
PJ
Jenkinson
 
EJ
Generating intrathymic microenvironments to establish T-cell tolerance.
Nat Rev Immunol
2007
, vol. 
7
 (pg. 
954
-
963
)
10
Adkins
 
B
Gandour
 
D
Strober
 
S
Weissman
 
I
Total lymphoid irradiation leads to transient depletion of the mouse thymic medulla and persistent abnormalities among medullary stromal cells.
J Immunol
1988
, vol. 
140
 (pg. 
3373
-
3379
)
11
Min
 
D
Taylor
 
PA
Panoskaltsis-Mortari
 
A
, et al. 
Protection from thymic epithelial cell injury by keratinocyte growth factor: a new approach to improve thymic and peripheral T-cell reconstitution after bone marrow transplantation.
Blood
2002
, vol. 
99
 (pg. 
4592
-
4600
)
12
Chung
 
B
Barbara-Burnham
 
L
Barsky
 
L
Weinberg
 
K
Radiosensitivity of thymic interleukin-7 production and thymopoiesis after bone marrow transplantation.
Blood
2001
, vol. 
98
 (pg. 
1601
-
1606
)
13
Gray
 
DH
Tull
 
D
Ueno
 
T
, et al. 
A unique thymic fibroblast population revealed by the monoclonal antibody MTS-15.
J Immunol
2007
, vol. 
178
 (pg. 
4956
-
4965
)
14
Rossi
 
SW
Jeker
 
LT
Ueno
 
T
, et al. 
Keratinocyte growth factor (KGF) enhances postnatal T- cell development via enhancements in proliferation and function of thymic epithelial cells.
Blood
2007
15
Min
 
D
Panoskaltsis-Mortari
 
A
Kuro
 
OM
Hollander
 
GA
Blazar
 
BR
Weinberg
 
KI
Sustained thymopoiesis and improvement in functional immunity induced by exogenous KGF administration in murine models of aging.
Blood
2007
, vol. 
109
 (pg. 
2529
-
2537
)
16
Rubin
 
JS
Osada
 
H
Finch
 
PW
Taylor
 
WG
Rudikoff
 
S
Aaronson
 
SA
Purification and characterization of a newly identified growth factor specific for epithelial cells.
Proc Natl Acad Sci U S A
1989
, vol. 
86
 (pg. 
802
-
806
)
17
Revest
 
JM
Suniara
 
RK
Kerr
 
K
Owen
 
JJ
Dickson
 
C
Development of the thymus requires signaling through the fibroblast growth factor receptor R2-IIIb.
J Immunol
2001
, vol. 
167
 (pg. 
1954
-
1961
)
18
Finch
 
PW
Rubin
 
JS
Keratinocyte growth factor/fibroblast growth factor 7, a homeostatic factor with therapeutic potential for epithelial protection and repair.
Adv Cancer Res
2004
, vol. 
91
 (pg. 
69
-
136
)
19
Spielberger
 
R
Stiff
 
P
Bensinger
 
W
, et al. 
Palifermin for oral mucositis after intensive therapy for hematologic cancers.
N Engl J Med
2004
, vol. 
351
 (pg. 
2590
-
2598
)
20
Stiff
 
PJ
Emmanouilides
 
C
Bensinger
 
WI
, et al. 
Palifermin reduces patient-reported mouth and throat soreness and improves patient functioning in the hematopoietic stem-cell transplantation setting.
J Clin Oncol
2006
, vol. 
24
 (pg. 
5186
-
5193
)
21
Alpdogan
 
O
Hubbard
 
VM
Smith
 
OM
, et al. 
Keratinocyte growth factor (KGF) is required for postnatal thymic regeneration.
Blood
2006
, vol. 
107
 (pg. 
2453
-
2460
)
22
Rossi
 
S
Blazar
 
BR
Farrell
 
CL
, et al. 
Keratinocyte growth factor preserves normal thymopoiesis and thymic microenvironment during experimental graft-versus-host disease.
Blood
2002
, vol. 
100
 (pg. 
682
-
691
)
23
Panoskaltsis-Mortari
 
A
Taylor
 
PA
Rubin
 
JS
, et al. 
Keratinocyte growth factor facilitates alloengraftment and ameliorates graft-versus-host disease in mice by a mechanism independent of repair of conditioning-induced tissue injury.
Blood
2000
, vol. 
96
 (pg. 
4350
-
4356
)
24
Taub
 
DD
Longo
 
DL
Insights into thymic aging and regeneration.
Immunol Rev
2005
, vol. 
205
 (pg. 
72
-
93
)
25
Hirokawa
 
K
Utsuyama
 
M
Kasai
 
M
Kurashima
 
C
Ishijima
 
S
Zeng
 
YX
Understanding the mechanism of the age-change of thymic function to promote T cell differentiation.
Immunol Lett
1994
, vol. 
40
 (pg. 
269
-
277
)
26
Utsuyama
 
M
Hirokawa
 
K
Mancini
 
C
Brunelli
 
R
Leter
 
G
Doria
 
G
Differential effects of gonadectomy on thymic stromal cells in promoting T cell differentiation in mice.
Mech Ageing Dev
1995
, vol. 
81
 (pg. 
107
-
117
)
27
Viselli
 
SM
Olsen
 
NJ
Shults
 
K
Steizer
 
G
Kovacs
 
WJ
Immunochemical and flow cytometric analysis of androgen receptor expression in thymocytes.
Mol Cell Endocrinol
1995
, vol. 
109
 (pg. 
19
-
26
)
28
Olsen
 
NJ
Olson
 
G
Viselli
 
SM
Gu
 
X
Kovacs
 
WJ
Androgen receptors in thymic epithelium modulate thymus size and thymocyte development.
Endocrinology
2001
, vol. 
142
 (pg. 
1278
-
1283
)
29
Kawashima
 
I
Seiki
 
K
Sakabe
 
K
Ihara
 
S
Akatsuka
 
A
Katsumata
 
Y
Localization of estrogen receptors and estrogen receptor-mRNA in female mouse thymus.
Thymus
1992
, vol. 
20
 (pg. 
115
-
121
)
30
Kohen
 
F
Abel
 
L
Sharp
 
A
, et al. 
Estrogen-receptor expression and function in thymocytes in relation to gender and age.
Dev Immunol
1998
, vol. 
5
 (pg. 
277
-
285
)
31
Pearce
 
PT
Khalid
 
BA
Funder
 
JW
Progesterone receptors in rat thymus.
Endocrinology
1983
, vol. 
113
 (pg. 
1287
-
1291
)
32
Heng
 
TS
Goldberg
 
GL
Gray
 
DH
Sutherland
 
JS
Chidgey
 
AP
Boyd
 
RL
Effects of castration on thymocyte development in two different models of thymic involution.
J Immunol
2005
, vol. 
175
 (pg. 
2982
-
2993
)
33
Goldberg
 
GL
Sutherland
 
JS
Hammet
 
MV
, et al. 
Sex steroid ablation enhances lymphoid recovery following autologous hematopoietic stem cell transplantation.
Transplantation
2005
, vol. 
80
 (pg. 
1604
-
1613
)
34
Sutherland
 
JS
Goldberg
 
GL
Hammett
 
MV
, et al. 
Activation of thymic regeneration in mice and humans following androgen blockade.
J Immunol
2005
, vol. 
175
 (pg. 
2741
-
2753
)
35
Roden
 
AC
Moser
 
MT
Tri
 
SD
, et al. 
Augmentation of T cell levels and responses induced by androgen deprivation.
J Immunol
2004
, vol. 
173
 (pg. 
6098
-
6108
)
36
Goldberg
 
GL
Alpdogan
 
O
Muriglan
 
SJ
, et al. 
Enhanced immune reconstitution by sex steroid ablation following allogeneic hemopoietic stem cell transplantation.
J Immunol
2007
, vol. 
178
 (pg. 
7473
-
7484
)
37
Matthews
 
PA
Hormone ablation therapy: lightening the load for today's prostate cancer patient.
Urol Nurs
2007
27
Suppl
(pg. 
3
-
11
)
38
Seach
 
N
Layton
 
D
Lim
 
J
Chidgey
 
A
Boyd
 
R
Thymic generation and regeneration: a new paradigm for establishing clinical tolerance of stem cell-based therapies.
Curr Opin Biotechnol
2007
, vol. 
18
 (pg. 
441
-
447
)
39
Jasperson
 
LK
Bucher
 
C
Panoskaltsis-Mortari
 
A
, et al. 
Indoleamine 2, 3-dioxygenase is a critical regulator of acute GVHD lethality.
Blood
2007
40
Gray
 
DH
Fletcher
 
AL
Hammett
 
M
, et al. 
Unbiased analysis, enrichment and purification of thymic stromal cells.
J Immunol Methods
2008
, vol. 
329
 (pg. 
56
-
66
)
41
Gray
 
DH
Chidgey
 
AP
Boyd
 
RL
Analysis of thymic stromal cell populations using flow cytometry.
J Immunol Methods
2002
, vol. 
260
 (pg. 
15
-
28
)
42
McCaughtry
 
TM
Wilken
 
MS
Hogquist
 
KA
Thymic emigration revisited.
J Exp Med
2007
, vol. 
204
 (pg. 
2513
-
2520
)
43
Waldschmidt
 
TJ
Panoskaltsis-Mortari
 
A
McElmurry
 
RT
Tygrett
 
LT
Taylor
 
PA
Blazar
 
BR
Abnormal T cell-dependent B-cell responses in SCID mice receiving allogeneic bone marrow in utero. Severe combined immune deficiency.
Blood
2002
, vol. 
100
 (pg. 
4557
-
4564
)
44
Kollmann
 
TR
Reikie
 
B
Blimkie
 
D
, et al. 
Induction of protective immunity to Listeria monocytogenes in neonates.
J Immunol
2007
, vol. 
178
 (pg. 
3695
-
3701
)
45
Hardy
 
J
Francis
 
KP
DeBoer
 
M
Chu
 
P
Gibbs
 
K
Contag
 
CH
Extracellular replication of Listeria monocytogenes in the murine gall bladder.
Science
2004
, vol. 
303
 (pg. 
851
-
853
)
46
Gray
 
D
Abramson
 
J
Benoist
 
C
Mathis
 
D
Proliferative arrest and rapid turnover of thymic epithelial cells expressing Aire.
J Exp Med
2007
, vol. 
204
 (pg. 
2521
-
2528
)
47
Miller
 
JS
Curtsinger
 
J
Berthold
 
M
, et al. 
Diminished neo-antigen response to keyhole limpet hemocyanin (KLH) vaccines in patients after treatment with chemotherapy or hematopoietic cell transplantation.
Clin Immunol
2005
, vol. 
117
 (pg. 
144
-
151
)
48
Seggewiss
 
R
Lore
 
K
Guenaga
 
FJ
, et al. 
Keratinocyte growth factor augments immune reconstitution after autologous hematopoietic progenitor cell transplantation in rhesus macaques.
Blood
2007
49
Wilson
 
AC
Meethal
 
SV
Bowen
 
RL
Atwood
 
CS
Leuprolide acetate: a drug of diverse clinical applications.
Expert Opin Investig Drugs
2007
, vol. 
16
 (pg. 
1851
-
1863
)
50
Prockop
 
SE
Petrie
 
HT
Regulation of thymus size by competition for stromal niches among early T cell progenitors.
J Immunol
2004
, vol. 
173
 (pg. 
1604
-
1611
)
51
Gray
 
DH
Seach
 
N
Ueno
 
T
, et al. 
Developmental kinetics, turnover, and stimulatory capacity of thymic epithelial cells.
Blood
2006
, vol. 
108
 (pg. 
3777
-
3785
)
52
Mathis
 
D
Benoist
 
C
A decade of AIRE.
Nat Rev Immunol
2007
, vol. 
7
 (pg. 
645
-
650
)
53
Berzins
 
SP
Boyd
 
RL
Miller
 
JF
The role of the thymus and recent thymic migrants in the maintenance of the adult peripheral lymphocyte pool.
J Exp Med
1998
, vol. 
187
 (pg. 
1839
-
1848
)
54
Prlic
 
M
Jameson
 
SC
Homeostatic expansion versus antigen-driven proliferation: common ends by different means?
Microbes Infect
2002
, vol. 
4
 (pg. 
531
-
537
)
55
Alpdogan
 
O
Muriglan
 
SJ
Eng
 
JM
, et al. 
IL-7 enhances peripheral T cell reconstitution after allogeneic hematopoietic stem cell transplantation.
J Clin Invest
2003
, vol. 
112
 (pg. 
1095
-
1107
)
56
Alpdogan
 
O
Eng
 
JM
Muriglan
 
SJ
, et al. 
Interleukin-15 enhances immune reconstitution after allogeneic bone marrow transplantation.
Blood
2005
, vol. 
105
 (pg. 
865
-
873
)
57
Stockinger
 
B
Bourgeois
 
C
Kassiotis
 
G
CD4+ memory T cells: functional differentiation and homeostasis.
Immunol Rev
2006
, vol. 
211
 (pg. 
39
-
48
)
58
Yamagami
 
M
McFadden
 
PW
Koethe
 
SM
Ratanatharathorn
 
V
Lum
 
LG
Failure of T cell receptor-anti-CD3 monoclonal antibody interaction in T cells from marrow recipients to induce increases in intracellular ionized calcium.
J Clin Invest
1990
, vol. 
86
 (pg. 
1347
-
1351
)
59
Pamer
 
EG
Immune responses to Listeria monocytogenes.
Nat Rev Immunol
2004
, vol. 
4
 (pg. 
812
-
823
)
60
Lara-Tejero
 
M
Pamer
 
EG
T cell responses to Listeria monocytogenes.
Curr Opin Microbiol
2004
, vol. 
7
 (pg. 
45
-
50
)
61
Prlic
 
M
Williams
 
MA
Bevan
 
MJ
Requirements for CD8 T-cell priming, memory generation and maintenance.
Curr Opin Immunol
2007
, vol. 
19
 (pg. 
315
-
319
)
62
Ozdemir
 
E
St John
 
LS
Gillespie
 
G
, et al. 
Cytomegalovirus reactivation following allogeneic stem cell transplantation is associated with the presence of dysfunctional antigen-specific CD8+ T cells.
Blood
2002
, vol. 
100
 (pg. 
3690
-
3697
)

Supplemental data

Sign in via your Institution