Thymic-deficient hosts rely primarily on antigen-driven expansion to restore the peripheral T-cell compartment following T-cell depletion (TCD). The degree to which this thymic-independent pathway can restore immune competence remains poorly understood but has important implications for a number of clinical conditions including stem cell transplantation and human immunodeficiency virus (HIV) infection. A model of HY-mediated skin graft rejection by athymic, TCD mice was used to show that restoration of naive and recall responses via peripheral expansion requires transfer of only 25 × 106 lymph node (LN) cells representing approximately 10% of the T-cell repertoire. Consitutive expression of bcl-2 in the expanding inocula restored recall responses to HY at a substantially lower LN cell dose (1 × 106), which is normally insufficient to induce HY-mediated graft rejection in athymic hosts. Interestingly, bcl-2 had no effect on primary responses. Interleukin-7 (IL-7) potently enhanced thymic-independent peripheral expansion and led to HY graft rejection using an LN cell dose of 1 × 106 in both primary and recall models. The restoration of immune competence by IL-7 appeared to be mediated through a combination of programmed cell death inhibition, improved costimulation, and modulation of antigen-presenting cell (APC) function. These results show that immune competence for even stringent antigens such as HY can be restored in the absence of thymic function and identify IL-7 as a potent modulator of thymic-independent T-cell regeneration.

Acute depletion of the T-cell compartment occurs in a number of clinical situations including stem cell transplantation, human immunodeficiency virus (HIV) infection, and after intensive cytotoxic therapy for cancer. Restoration of immune competence following T-cell depletion (TCD) requires the regeneration of a functionally intact, diverse T-cell pool that is capable of responding to foreign antigens and, potentially, altered self-antigens expressed by tumors. Previous studies have shown that there are 2 main pathways capable of substantial peripheral T-cell regeneration: thymic-dependent regeneration from bone marrow progenitors and thymic-independent peripheral expansion of mature T-cell populations.1 The relative contribution of these 2 pathways is dynamic and dependent on the degree of thymic function. Current concepts hold that the capacity of the host to restore immune competence depends primarily on the extent to which thymic pathways contribute to T-cell regeneration. However, due to disease, therapy-related toxicity, and age-related changes, thymic function is frequently limiting, resulting in a relative reliance on thymic-independent peripheral expansion in many clinical situations associated with TCD.2-8 The degree to which the peripheral expansion of mature T-cell populations can restore host immune competence following TCD remains poorly understood and is the focus of this report.

Peripheral expansion is predicted to give rise to limited immune competence for a variety of reasons. First, although dramatic expansions in cell number can occur,9-12 hosts reconstituted in this manner display chronically reduced T-cell numbers.10,13 Second, because the expansion of peripheral T cells via this process proceeds from limited numbers of cells and is heavily influenced by interactions with cognate antigen,14the regenerated repertoire is susceptible to dramatic skewing15 and shows limited T-cell receptor diversity.16-19 Third, the process of peripheral expansion leads to an accumulation of T cells that display a highly activated phenotype.13,20 Continuous or repeated exposure of these activated T cells to antigens that contribute to the process of expansion could also provide a signal for programmed cell death.21,22 This is suggested by studies in patients with TCD due to HIV infection showing increased susceptibility of T cells to apoptosis with restimulation in vitro23-27 and by direct labeling studies in vivo.28-30 Although these observations could potentially relate to direct or indirect effects of HIV, similar results have been observed in T-cell–depleted hosts after intensive chemotherapeutic regimens20 and after stem cell transplantation,31 suggesting that increased susceptibility to cell death is a common phenomenon in regenerating T-cell populations. Thus, a propensity for programmed cell death in peripherally expanding T-cell populations may also limit the capacity of peripheral expansion to restore host immune competence.

Interleukin-7 (IL-7) exerts potent effects on T- and B-cell progenitors and is absolutely required for early T-cell development.32-34 Because T-cell development can be largely restored in IL-7 knockout mice by the concurrent expression of a bcl-2 transgene,35,36 current models suggest that IL-7 acts primarily by inhibiting programmed cell death of immature thymocytes during development. Indeed, the administration of IL-7 after cyclophosphamide or bone marrow transplantation in mice leads to enhanced T-cell reconstitution, presumably via enhanced thymopoiesis.37-41 

Recent studies have shown that IL-7 is also capable of rescuing mature T cells from apoptosis induced by glucocorticoids, cytokine withdrawal, and radiation.42-47 These effects are correlated with alterations in intracellular levels of bcl-2, bcl-xL, and caspase 3 activity, suggesting modulation of intracellular pathways of programmed cell death. In this report, we sought to examine the capacity of peripheral expansion to restore immune competence following TCD and to investigate the role of programmed cell death in limiting immune competence following reconstitution by peripheral expansion. Our results show that programmed cell death plays an important role in limiting immune competence in thymic-deficient hosts undergoing T-cell regeneration and, in addition, we have identified IL-7 as a cytokine that dramatically enhances immune competence in thymic-deficient hosts reconstituted via this pathway.

Mice and thymectomies

C57BL/6 (B6/Thy-1.2, Ly-5.1), B6 PL Thy-1a (B6/Thy 1.1), and B6/Ly-5.2 mice were purchased from the Animal Production Unit, National Cancer Institute (NCI; Frederick, MD) and housed in a specific pathogen-free environment at the National Institutes of Health (NIH). At 4 to 6 weeks of age, mice were anesthetized and suction thymectomy was performed through a sternal incision according to approved protocol. Completeness of the thymectomies was confirmed by visual inspection at the completion of the experiments. C57BL/6-TgN(BCL2)36Wehi mice48 were originally purchased from Jackson Laboratories (Bar Harbor, ME) and bred at the National Institutes of Health. Mice were screened for human bcl-2 (hbcl-2) transgene expression at 8 weeks of age. A small aliquot of blood was obtained from the tail followed by separation of mononuclear cells over lymphocyte separation media (Biowhitaker, Walkersville, MD). The presence of hbcl-2 protein was determined by flow cytometry using intracellular labeling with a human-specific, monoclonal antibody (clone 6C8, Pharmingen, San Diego, CA) and detected with goat antihamster IgG fluorescein isothiocyanate (FITC; Southern Biotechnology Associates, Birmingham, AL). The validation of this screening technique was confirmed by polymerase chain reaction (PCR) analysis. Transgene-negative littermates were used as control lymph node donors. C57BL/6 IL-7Rα−− mice were purchased from Jackson Laboratories and bred at the NCI-Frederick.

TCD and skin grafting

Mice were depleted of T cells using rat antimouse anti-CD4 (clone GK1.5) and anti-CD8 (clone 2.43) purchased from the Biological Resources Branch, NCI (Frederick, MD). Injections were given intraperitoneally with schedules and doses determined for individual lots by in vivo experiments to achieve more than 98% depletion of CD4 and CD8 cells in spleen and lymph node (LN) at 4 days (data not shown). After 2 weeks, to allow clearance of the monoclonal antibodies, mice were injected with LN populations as indicated for the individual experiments. Skin grafting was performed using a modification of a protocol described elsewhere49 with a 0.5-cm patch of male tail skin grafted over the thorax and covered with a pressure dressing for 7 days. After 1 week, bandages were removed and a blinded observer monitored graft rejection. Complete rejection was defined as more than 80% of the graft surface area involved. Animal care was provided in accordance with procedures outlined in the “Guide for the Care and Use of Laboratory Animals” (NIH Publication no. 86-23, 1996) and all protocols were approved by the animal care and use committee at the NCI.

Cellinjections

Axillary and inguinal LNs were harvested from syngeneic female mice and placed in iced complete media (RPMI, with 10% heat-inactivated fetal bovine serum, penicillin, streptomycin,l-glutamine, Hepes buffer, nonessential amino acids, sodium pyruvate (all from Gibco Life Technologies, Gaithersburg, MD), and β-mercaptoethanol (Sigma, St. Louis, MO). LNs were teased apart with fine forceps, gently minced with the plunger of a syringe, and passed through a nylon mesh. The cells were washed twice in iced complete media, viable cell number was determined with trypan blue exclusion, and cells were resuspended at the appropriate concentration for injection via tail vein in RPMI without fetal calf serum at the doses described.

Male dendritic cells were purified from splenocytes following plate adherence and incubation with IL-4 and granulocyte-macrophage colony-stimulating factor (GM-CSF). Briefly, male spleens were minced with the plunger of a syringe in iced complete media and passed through a nylon mesh. Red blood cells (RBCs) were lysed with ammonium hydroxide lysing buffer, washed twice in complete media, resuspended in iced Dulbecco phosphate-buffered saline (DPBS; Gibco Life Technologies) and separated over a 50% Percoll gradient by centrifugation at 1800g for 12 minutes. The cell layer was removed, washed twice in complete media, resuspended, and counted using trypan blue exclusion. Viable cells (100 × 106) were loaded onto a 150 × 25-mm tissue culture dish (Falcon 3025, Becton Dickinson, Lincoln Park, NJ), incubated at room temperature for 20 minutes followed by incubation at 37°C/5% CO2 for 2 hours. Nonadherent cells were removed by washing with 50 mL warm complete media, the media was replaced with complete media containing 1 ng/mL GM-CSF and 0.1 μg/mL IL-4 (Peprotech, Rocky Hill, NJ), and the plate was incubated overnight at 37°C/5% CO2. On day 2, the nonadherent fraction was removed by gentle pipetting, washed twice in complete media, resuspended, and counted. A fraction of the cell suspension was analyzed with flow cytometry and the remainder was injected in RPMI intraperitoneally at a dose of 1 × 105cells per mouse.

In vitro studies

Single-cell suspensions of RBC-depleted splenocytes were made as described followed by passage over a negative selection T-cell enrichment column (R&D Systems, Minneapolis, MN) according to the manufacturer's instructions. The enriched fraction was washed, counted, and resuspended in complete media. The 24-well plates were coated for 18 hours with 0.1 μg/mL rat antimouse CD3 (clone 145-2C11) in DPBS or with DPBS alone. This concentration of 2C11 is insufficient to induce maximal T-cell proliferation (data not shown). T-cell–enriched splenocytes were plated at 4 × 106cells/well in 2 mL complete media. Recombinant human IL-7 (Peprotech) was added at a concentration of 10 ng/mL. At the time points indicated, wells were washed, pooled, and analyzed with flow cytometry. For proliferation studies, T-cell–enriched splenocytes were labeled with CFSE (5-[and 6]-carboxyfluorescein diacetate succinimidyl ester; Molecular Probes, Eugene, OR) according to the manufacturer's instructions. Labeled cells were incubated on a 96-well plate coated with anti-CD3 as described above.

Flow cytometric analysis

Cell suspensions were prepared in staining buffer (Hanks balanced salt solution without phenol red with 0.2% human serum albumin [Sigma] and 0.1% sodium azide [Sigma]). For intracellular labeling, cell suspensions were washed in staining buffer containing 0.03% saponin (Sigma). Prior to antibody labeling, FCγIII/II receptors were blocked with monoclonal antibody (clone 2.4G2). The monoclonal antibodies used were CD4 phycoerythrin (PE) (clone CT-CD4) and CD8a PE (clone CT-CD8a) (Caltag Laboratories, Burlingame, CA), TCR β chain PE (clone H57-597), CD45R/B220PE (clone RA3-6B2), CD11b PE (clone M1/70), CD11c FITC (clone HL3), CD45.1/Ly5.2 FITC (clone A20), CD45.2/Ly5.1 FITC (clone 104), CD54/ICAM-1 FITC (clone 3E2), CD80/B7-1 FITC (clone 1G10), CD86/B7-2 FITC (clone GL-1), CD90.1/Thy 1.1 FITC (clone HIS51), and CD90.2/Thy1.2 FITC (clone OX-7) (Pharmingen). Intracellular murine bcl-2 was detected with purified hamster antimouse bcl-2 (clone 3F11; Pharmingen) and detected with goat antihamster IgG FITC (Southern Biotechnology Associates). Appropriate isotype controls were used for all experiments. Annexin V labeling was performed in phosphate-buffered saline (PBS) containing 10 mM NaOH, 140 mM NaCl, and 2.5 mM CaCl2. Cells were then analyzed in buffer containing 2 μg/mL propidium iodide (Sigma).

Flow cytometric analysis was performed on a single laser FACScan or dual laser FACSCalibur (Becton Dickinson). Fluorescence data were collected and analyzed using CELLQuest software. Viable lymphocyte populations were gated based on forward-scatter and side-scatter characteristics.

Statisticalanalysis

Survival analysis was performed on the HY skin graft rejection experiments considering the time of complete rejection in days as the end-point. A dichotomous variable of 1 was assigned if a rejection occurred during the period of observation (100 days) and value of 0 was assigned if rejection did not occur and the event was censored. For each comparison, a log-rank test was performed and the 2-sided Pvalue is reported.

Restoration of immune competence in athymic hosts reconstituted via peripheral expansion is critically dependent on the size of the expanding inocula

To assess the capability of hosts reconstituted via peripheral expansion to respond to recall antigen, thymectomized, T-cell–depleted C57BL/6 females were grafted with male tail skin and injected intravenously with 1 × 106 primed LN cells as a source of mature T cells. In the recall model, primed cells refer to LN cells taken from syngeneic female mice within 3 weeks after complete male skin graft rejection. Thymus-bearing mice reject HY-disparate skin grafts within 8 weeks. In contrast, thymectomized, T-cell–depleted mice were unable to reject HY-disparate skin grafts despite administration of primed LN cells as a source for peripheral expansion. Importantly, these mice are capable of rejecting allogeneic skin grafts within 8 to 10 days (data not shown). Therefore, the impairment in T-cell immune competence following reconstitution via thymic-independent pathways is limited to responses to antigen with a low precursor frequency suggesting that a simple limitation in the number of cells bearing antigen-specific T-cell receptor specificities may play a central role.

To determine the degree to which the deficiency in HY graft rejection observed following thymic-independent T-cell regeneration was related to an inadequate starting T-cell inocula, and, if so, to establish the size of the inocula required to restore responses to recall antigen, thymectomized T-cell–depleted mice were reconstituted using progressively larger primed mature LN inocula. As the dose of primed LN cells was increased, there is an increase in the rate of graft rejection (Figure 1A). Indeed, T-cell–depleted mice reconstituted from 25 × 106 primed LN cells were able to reject HY grafts at a rate analogous to that observed in control groups.

Fig. 1.

Rejection of HY-disparate skin grafts via peripherally expanded lymph node cells is critically dependent on the size of the starting inocula.

(A) Adult thymectomized, C57BL/6 females were T cell depleted and grafted with male tail skin as described in “Materials and methods.” Primed cells were collected from the draining LN of T-cell–replete syngeneic female mice 3 weeks after successful male skin graft rejection, teased into a single-cell suspension, and injected via the tail vein 24 hours after skin grafting. Unprimed cells were collected from LN of T-cell–replete females naive to male antigen. Percent surviving grafts are shown as measured by visual inspection as described in “Materials and methods.” Thymectomized, T-cell–depleted mice (TXY/TCD mice) receiving 5 × 106primed LN cells are partially able to reject HY-disparate skin grafts (▪, n = 4) when compared to mice receiving no LN inocula (●, n = 5). When the LN cell dose is increased to 10 × 106, there is rejection in all mice but the rate of graft rejection remains delayed (♦, n = 5). At a dose of 25 × 106 LN cells (▴, solid line, n = 5), graft rejection occurs at a rate analogous to thymus-bearing control animals (▾, dashed line, n = 6) also receiving primed inocula. Note the modest effect of the primed inocula when this group is compared to thymus-bearing animals receiving an unprimed inocula (▿, dashed line, n = 5). (B) To assess primary immune responses, TXY/TCD mice were given graded numbers of LN cells from syngeneic females naive to male antigen via tail vein. The mice were then sensitized by intraperitoneal injection of 1 × 105 enriched male dendritic cells from male splenocytes as described in “Materials and methods.” These cells express B7-1, B7-2, CD11c, and major histocompatibility complex (MHC) class II and represent 50% to 60% of the cells injected in all experiments. Twenty-one days after LN cell transfer and enriched male dendritic cell sensitization, the mice were grafted with male tail skin as before and observed for rejection. As with the recall responses, mice receiving 1 × 106 naive cells were unable to reject HY-disparate skin grafts (●, n = 5). Administration of 10 × 106 naive cells led to graft rejection in all of the mice but at a delayed rate (▪, n = 7). Transfer of 25 × 106 LN cells (⧫, n = 7) completely restored responses so that rejection occurred at similar rate to thymus-bearing control groups receiving enriched dendritic cells (▴, dashed line, n = 6). Thymus-bearing control groups receiving no enriched dendritic cells (▾, dashed line, n = 5) required approximately 2 weeks longer to reject HY grafts.

Fig. 1.

Rejection of HY-disparate skin grafts via peripherally expanded lymph node cells is critically dependent on the size of the starting inocula.

(A) Adult thymectomized, C57BL/6 females were T cell depleted and grafted with male tail skin as described in “Materials and methods.” Primed cells were collected from the draining LN of T-cell–replete syngeneic female mice 3 weeks after successful male skin graft rejection, teased into a single-cell suspension, and injected via the tail vein 24 hours after skin grafting. Unprimed cells were collected from LN of T-cell–replete females naive to male antigen. Percent surviving grafts are shown as measured by visual inspection as described in “Materials and methods.” Thymectomized, T-cell–depleted mice (TXY/TCD mice) receiving 5 × 106primed LN cells are partially able to reject HY-disparate skin grafts (▪, n = 4) when compared to mice receiving no LN inocula (●, n = 5). When the LN cell dose is increased to 10 × 106, there is rejection in all mice but the rate of graft rejection remains delayed (♦, n = 5). At a dose of 25 × 106 LN cells (▴, solid line, n = 5), graft rejection occurs at a rate analogous to thymus-bearing control animals (▾, dashed line, n = 6) also receiving primed inocula. Note the modest effect of the primed inocula when this group is compared to thymus-bearing animals receiving an unprimed inocula (▿, dashed line, n = 5). (B) To assess primary immune responses, TXY/TCD mice were given graded numbers of LN cells from syngeneic females naive to male antigen via tail vein. The mice were then sensitized by intraperitoneal injection of 1 × 105 enriched male dendritic cells from male splenocytes as described in “Materials and methods.” These cells express B7-1, B7-2, CD11c, and major histocompatibility complex (MHC) class II and represent 50% to 60% of the cells injected in all experiments. Twenty-one days after LN cell transfer and enriched male dendritic cell sensitization, the mice were grafted with male tail skin as before and observed for rejection. As with the recall responses, mice receiving 1 × 106 naive cells were unable to reject HY-disparate skin grafts (●, n = 5). Administration of 10 × 106 naive cells led to graft rejection in all of the mice but at a delayed rate (▪, n = 7). Transfer of 25 × 106 LN cells (⧫, n = 7) completely restored responses so that rejection occurred at similar rate to thymus-bearing control groups receiving enriched dendritic cells (▴, dashed line, n = 6). Thymus-bearing control groups receiving no enriched dendritic cells (▾, dashed line, n = 5) required approximately 2 weeks longer to reject HY grafts.

Close modal

To examine the induction of primary immune responses after thymic-independent regeneration, thymectomized T-cell–depleted mice were injected with LN cells from syngeneic female mice naive to male antigen at the same time as enriched male dendritic cells as a sensitizing population. In this model sensitization must occur following transfer into T-cell–depleted hosts. In T-cell–replete mice, sensitization with enriched male dendritic cells accelerated the rate of graft rejection by approximately 2 weeks, confirming the ability of these cells to sensitize to HY antigen (Figure 1B). In contrast, as seen in the recall experiments, thymectomized T-cell–depleted mice reconstituted with 1 × 106 naive LN cells failed to reject male grafts despite sensitization with enriched male dendritic cells. Importantly, titration of the lymph node dose to 25 × 106 LN cells led to complete restoration of responses to HY antigen.

Therefore, in both the primary response and the recall model, limitations in the capacity of athymic hosts to respond to nominal antigen could be overcome by supplying increased numbers of T cells for peripheral expansion. These results confirm that the impairment in T-cell responses observed in thymic-deficient hosts is not absolute and can be induced in this setting if sufficient T-cell numbers are provided.

Inhibition of apoptosis during reconstitution from a limited T-cell inocula restores responses to recall antigen but not to naive antigen

To study the mechanisms that contribute to the limitation in T-cell responses following immune reconstitution via peripheral expansion, subsequent experiments attempted to identify factors limiting peripheral expansion in hosts reconstituted from suboptimal numbers of T cells. Based on observations made in clinical settings associated with TCD,20 31 we hypothesized that increased apoptosis of peripherally expanding T cells could limit immune competence in this setting. To determine whether expanding populations are susceptible to programmed cell death in our model, T cells from thymectomized, T-cell–depleted C57BL/6 (Thy-1.2) mice reconstituted from 10 × 106 congenic B6 PL Thy-1a(Thy-1.1) LN cells were analyzed for apoptosis at various time points after transfer. When analyzed on the day of collection there was no evidence for increased T-cell apoptosis in peripherally expanding cells when compared to normal, nonexpanding populations. However, as shown in Figure2, peripherally expanding congenic T cells from thymectomized, T-cell–depleted animals collected on day 15 after transfer and restimulated with concanavalin A for 18 hours show increased annexin V staining when compared to nonexpanding controls. This provides evidence that T cells undergoing peripheral expansion in thymectomized T-cell–depleted hosts are susceptible to apoptotic cell death on restimulation and suggested that a loss of T cells to apoptosis during peripheral expansion from an insufficient inocula could potentially play a role in limiting T-cell responses in these hosts.

Fig. 2.

Increased annexin V staining of peripherally expanding T cells.

TXY/TCD C57BL/6 (Thy 1.2) mice were injected with 10 × 106 congenic LN cells from B6 PL Thy-1a females (Thy 1.1). At 5, 11, and 21 days, mice were killed for flow cytometric analysis of apoptosis in Thy 1.1+ T cells. This figure is a representative histogram showing annexin V staining on Thy 1.1+ propidium iodide–negative splenocytes (gray line) at 11 days after an 18 hours incubation with concanavalin A compared to Thy 1.2+ splenocytes from thymus-bearing, T-cell–replete controls (black line) also incubated with con A for 18 hours. In a separate experiment there was no increased annexin V staining on Thy 1.2+ splenocytes from thymectomized T-cell–replete females when compared to sham thymectomized animals treated in the same manner (data not shown).

Fig. 2.

Increased annexin V staining of peripherally expanding T cells.

TXY/TCD C57BL/6 (Thy 1.2) mice were injected with 10 × 106 congenic LN cells from B6 PL Thy-1a females (Thy 1.1). At 5, 11, and 21 days, mice were killed for flow cytometric analysis of apoptosis in Thy 1.1+ T cells. This figure is a representative histogram showing annexin V staining on Thy 1.1+ propidium iodide–negative splenocytes (gray line) at 11 days after an 18 hours incubation with concanavalin A compared to Thy 1.2+ splenocytes from thymus-bearing, T-cell–replete controls (black line) also incubated with con A for 18 hours. In a separate experiment there was no increased annexin V staining on Thy 1.2+ splenocytes from thymectomized T-cell–replete females when compared to sham thymectomized animals treated in the same manner (data not shown).

Close modal

Although the finding of increased annexin V staining of restimulated, peripherally expanding T cells, along with the findings of other investigators,20,31 suggests that these cells are prone to apoptosis, it is unclear whether this process contributes to the limitation in immune responses observed in hosts reconstituted in this manner. If the loss of peripherally expanding T cells to apoptosis in our skin graft rejection model contributes to the inability to induce responses to HY antigen in animals reconstituted from a suboptimal T-cell number, we hypothesized that inhibition of apoptosis in vivo could restore HY-mediated graft rejection. Thymectomized T-cell–depleted mice were injected with 1 × 106 primed LN cells from female mice expressing an hbcl-2 transgene in lymphocytes. T cells from these animals are resistant to apoptotic cell death induced by a number of mechanisms including radiation and cytokine withdrawal.50 51 As shown in Figure3A, mice reconstituted via peripheral expansion of 1 × 106 primed, hbcl-2 transgenic LN cells were able to rapidly reject HY-disparate skin grafts, whereas mice reconstituted from the same size inocula of LN cells from nontransgenic littermate controls were unable to reject these grafts by 100 days. Therefore, inhibition of apoptosis via constitutive bcl-2 expression during peripheral expansion is sufficient to restore recall responses to nominal antigen, thereby confirming the role of apoptosis in limiting immune responses in this setting. Interestingly, no significant increase in the rate of graft rejection was observed in T-cell–replete hosts suggesting that programmed cell death is uniquely limiting in the setting of TCD.

Fig. 3.

Inhibition of programmed cell death by constitutive expression of hbcl-2 in peripherally expanding T cells restores recall responses to HY-disparate skin grafts at suboptimal inocula but fails to restore primary responses.

(A) TXY/TCD mice were reconstituted with 1 × 106 primed LN cells constitutively expressing hbcl-2 (●, n = 9) 24 hours after placement of a male tail skin graft. This resulted in restoration of HY-disparate skin graft rejection when compared to mice receiving unprimed hbcl-2 transgenic inocula (▪, n = 6) or primed cells from nontransgenic littermate controls (♦, n = 9). Primed hbcl-2 transgenic inocula did not enhance graft rejection in thymus-bearing controls (▾, dashed line, n = 5) when compared to primed inocula from nontransgenic littermate controls (▴, dashed line, n = 5,P = .49). (B) Reconstitution of TXY/TCD mice with 1 × 106 naive hbcl-2 transgenic LN cells followed by sensitization with enriched male dendritic cells does not restore rejection of HY-disparate skin grafts placed 3 weeks after cell transfers (●, n = 5). Titration of the inocula to 10 × 106 (▪, n = 5) and 25 × 106(♦, n = 5) leads to HY-disparate graft rejection at a rate comparable to thymus-bearing controls (▴, dashed line, n = 5) analogous to nontransgenic inocula (Figure 2B).

Fig. 3.

Inhibition of programmed cell death by constitutive expression of hbcl-2 in peripherally expanding T cells restores recall responses to HY-disparate skin grafts at suboptimal inocula but fails to restore primary responses.

(A) TXY/TCD mice were reconstituted with 1 × 106 primed LN cells constitutively expressing hbcl-2 (●, n = 9) 24 hours after placement of a male tail skin graft. This resulted in restoration of HY-disparate skin graft rejection when compared to mice receiving unprimed hbcl-2 transgenic inocula (▪, n = 6) or primed cells from nontransgenic littermate controls (♦, n = 9). Primed hbcl-2 transgenic inocula did not enhance graft rejection in thymus-bearing controls (▾, dashed line, n = 5) when compared to primed inocula from nontransgenic littermate controls (▴, dashed line, n = 5,P = .49). (B) Reconstitution of TXY/TCD mice with 1 × 106 naive hbcl-2 transgenic LN cells followed by sensitization with enriched male dendritic cells does not restore rejection of HY-disparate skin grafts placed 3 weeks after cell transfers (●, n = 5). Titration of the inocula to 10 × 106 (▪, n = 5) and 25 × 106(♦, n = 5) leads to HY-disparate graft rejection at a rate comparable to thymus-bearing controls (▴, dashed line, n = 5) analogous to nontransgenic inocula (Figure 2B).

Close modal

Given the similar results obtained in the LN cell titration experiments between recall and primary responses detailed above, we predicted that the constitutive expression of bcl-2 transgene in peripherally expanding T cells would also lead to restoration of primary immune responses to HY. Surprisingly, however, 1 × 106 naive hbcl-2 transgenic LN cells sensitized with enriched male dendritic cells failed to restore responses to HY antigen in athymic, T-cell–depleted hosts (Figure 3B). Indeed, the same size inocula of both transgenic and nontransgenic LN cells (25 × 106) was required to fully restore HY responses in this model. These results indicate that, unlike responses to recall antigen, constitutive bcl-2 expression in T cells undergoing thymic-independent regeneration from a limited inocula is insufficient to induce primary responses to HY antigens in thymic-deficient hosts and suggests that additional factors beyond the susceptibility to programmed cell death contribute to limitations in induction of primary T-cell responses in thymic-deficient hosts.

IL-7 has diverse effects on mature T cells in vitro, and in vivo administration leads to enhanced peripheral expansion

The T cells undergoing peripheral expansion display an activated phenotype regardless of activation status prior to injection into T-cell–depleted hosts.15 Therefore, modulation of T-cell activation could potentially enhance the process of peripheral expansion. To examine the effects of IL-7 on the activation of mature T cells in vitro, T-cell–enriched splenocytes were stimulated with plate-bound anti-CD3 antibody at a suboptimal concentration with and without recombinant human IL-7 (rhIL-7). By day 5, T cells incubated with rhIL-7 plus anti-CD3 displayed increased bcl-2 protein (Figure 4A) consistent with the antiapoptotic effect of this cytokine. In addition, prelabeling of T cells with CFSE indicated that rhIL-7 enhanced proliferation (Figure4B). By day 7, the number of viable T cells recovered after incubation with rhIL-7 plus anti-CD3 was significantly increased when compared to unstimulated groups or to groups incubated with anti-CD3 alone (Figure4C). Consistent with prior reports,42-46 these results indicate that IL-7 can have multiple effects on mature T cells including an antiapoptotic effect and a costimulatory effect and, therefore, may be particularly capable of modulating peripheral expansion in vivo.

Fig. 4.

IL-7 has diverse effects on mature T cells in vitro and leads to enhanced peripheral expansion in vivo.

Female C57BL/6 splenocytes were enriched for T cells using a positive selection column as described in “Materials and methods.” T-cell–enriched splenocytes were incubated in 24-well plates precoated with anti-CD3 at a suboptimal concentration of 0.1 μg/mL with or without rhIL-7 at 10 ng/mL. At days 2, 5, and 7, wells were harvested. Viable cells were counted with trypan blue exclusion and analyzed with flow cytometry. (A) By day 5 of culture, there is increased bcl-2 fluorescence intensity in cells incubated with rhIL-7 and stimulated by suboptimal anti-CD3 (heavy solid line) when compared to suboptimal αCD3 alone (gray dotted line) or media (light solid line). Isotype control shown as dark dotted line. (B) T-cell–enriched splenocytes were prelabeled with CFSE according to the manufacturer's instructions. At day 5, there is decreased CFSE intensity in a subset of T cells from wells incubated with suboptimal anti-CD3 in combination with rhIL-7 (solid line) when compared to suboptimal anti-CD3 alone or media only (dotted lines) indicating increased proliferation. Analogous results were obtained with H3 incorporation and the difference was not seen at optimal anti-CD3 stimulation (data not shown). (C) Incubation with suboptimal anti-CD3 and rhIL-7 (▪) results in increased viable T-cell number per well by days 5 and 7 compared to controls with suboptimal anti-CD3 (●) or media alone (♦). (D) TXY/TCD, C57BL/6 Ly 5.2 mice were injected intravenously with an inocula of 10 × 106 LN cells from normal or hbcl-2 transgenic donors (both Ly 5.1). One group receiving nontransgenic LN cells was treated daily with rhIL-7 at a dose of 5 μg/d intraperitoneally for 28 days. By day 28, there was a significant increase in the number of Ly 5.1+ T cells in the rhIL-7–treated group (n = 7, P = .004) or the group receiving hbcl-2 transgenic inocula (n = 7, P = .007) compared to control animals (n = 7). Data were analyzed using an unpaired t test.

Fig. 4.

IL-7 has diverse effects on mature T cells in vitro and leads to enhanced peripheral expansion in vivo.

Female C57BL/6 splenocytes were enriched for T cells using a positive selection column as described in “Materials and methods.” T-cell–enriched splenocytes were incubated in 24-well plates precoated with anti-CD3 at a suboptimal concentration of 0.1 μg/mL with or without rhIL-7 at 10 ng/mL. At days 2, 5, and 7, wells were harvested. Viable cells were counted with trypan blue exclusion and analyzed with flow cytometry. (A) By day 5 of culture, there is increased bcl-2 fluorescence intensity in cells incubated with rhIL-7 and stimulated by suboptimal anti-CD3 (heavy solid line) when compared to suboptimal αCD3 alone (gray dotted line) or media (light solid line). Isotype control shown as dark dotted line. (B) T-cell–enriched splenocytes were prelabeled with CFSE according to the manufacturer's instructions. At day 5, there is decreased CFSE intensity in a subset of T cells from wells incubated with suboptimal anti-CD3 in combination with rhIL-7 (solid line) when compared to suboptimal anti-CD3 alone or media only (dotted lines) indicating increased proliferation. Analogous results were obtained with H3 incorporation and the difference was not seen at optimal anti-CD3 stimulation (data not shown). (C) Incubation with suboptimal anti-CD3 and rhIL-7 (▪) results in increased viable T-cell number per well by days 5 and 7 compared to controls with suboptimal anti-CD3 (●) or media alone (♦). (D) TXY/TCD, C57BL/6 Ly 5.2 mice were injected intravenously with an inocula of 10 × 106 LN cells from normal or hbcl-2 transgenic donors (both Ly 5.1). One group receiving nontransgenic LN cells was treated daily with rhIL-7 at a dose of 5 μg/d intraperitoneally for 28 days. By day 28, there was a significant increase in the number of Ly 5.1+ T cells in the rhIL-7–treated group (n = 7, P = .004) or the group receiving hbcl-2 transgenic inocula (n = 7, P = .007) compared to control animals (n = 7). Data were analyzed using an unpaired t test.

Close modal

To test this hypothesis, rhIL-7 was administered at a dose of 5 μg/d intraperitoneally for 28 days to thymectomized, T-cell–depleted C57Bl/6 Ly5.2 mice during peripheral expansion of 10 × 106 Ly5.1+ LN cells. In addition, in the same experiment, the potential for peripheral expansion of hbcl-2 transgenic (Ly 5.1) inocula was determined. As seen in Figure4D, in vivo administration of rhIL-7 led to a significant increase in Ly5.1+ T-cell number. A similar result was observed in mice receiving hbcl-2 transgenic inocula, further confirming the role for apoptosis in limiting peripheral expansion. Therefore, both rhIL-7 treatment and constitutive expression of bcl-2 numerically enhance the peripheral expansion of mature T cells in athymic T-cell–depleted hosts.

IL-7 treatment restores recall and primary immune responses in athymic, T-cell–depleted hosts undergoing immune reconstitution from a limited T-cell inocula

Due to the in vitro effects of IL-7 and the potent enhancement of peripheral expansion observed in vivo, we tested whether IL-7 could restore recall responses during reconstitution from suboptimal inocula. As shown in Figure5A, administration of rhIL-7 during peripheral expansion from a limited primed LN inocula (1 × 106) leads to rapid rejection of HY-disparate skin grafts. Importantly, rejection of male skin grafts in all animals required administration of rhIL-7 along with a primed LN inocula, providing evidence that the effect of IL-7 in this model was dependent on the presence of a population of mature T cells undergoing peripheral expansion. We also tested whether the ability of IL-7 to restore immune responses extended to primary responses. As with recall responses, administration of rhIL-7 also led to rapid rejection of HY-disparate skin grafts in mice reconstituted from a limited naive LN inocula and sensitized with enriched male dendritic cells (Figure 5B). The rhIL-7 appeared to have no effect on responses in T-cell–replete hosts in both recall and primary models. Therefore, IL-7 potently restores immune competence in thymic-deficient hosts undergoing T-cell reconstitution via peripheral expansion from a limited inocula. These findings contrast with the experiments using T-cell inocula constitutively expressing the bcl-2 transgene where effects were only seen with recall responses. Thus, we postulated that the ability of IL-7 to restore immune competence in this setting involves other effects on peripherally expanding T cells beyond a modulation of programmed cell death via up-regulation of bcl-2. In addition, we could not exclude the possibility that IL-7 may also enhance other cell populations critical to the generation of immune responses.

Fig. 5.

IL-7 potently restores both recall and primary responses to HY-disparate skin grafts in T-cell–depleted hosts receiving suboptimal T cell inocula.

(A) TXY/TCD mice were grafted with male tail skin and injected with 1 × 106 primed LN cells via tail vein 24 hours after grafting. Then rhIL-7 was injected intraperitoneally at a dose of 5 μg/d beginning on the day of primed cell injection and continuing for 28 days. Animals receiving rhIL-7 and 1 × 106 primed LN cells, shown in previous experiments to be a suboptimal inocula (▪, n = 8), rapidly rejected HY-disparate skin grafts at nearly the same rate as thymus-bearing animals (▴, dashed line, n = 9,P = .2) and significantly faster than animals injected with vehicle only plus primed LN cells (●, n = 9,P = .0002) or rhIL-7 without primed LN cells (♦, n = 6, P = .019). Note the lack of effect in thymus-bearing animals injected with rhIL-7 (▾, dashed line, n = 5) compared to vehicle (▴, dashed line, n = 5). (B) TXY/TCD mice were injected with 1 × 106 naive LN cells and enriched male dendritic cells. The rhIL-7 at 5 μg/d intraperitoneally was initiated on the day of cell injection and continued for 28 days. Male skin grafting was performed on day 21. Similar to recall responses, administration of rhIL-7 in the primary model led to rapid rejection of HY-disparate skin grafts in TCD mice (●, n = 8), whereas mice receiving the same inocula and injected with vehicle were completely unable to reject these grafts (▪, n = 7,P = .0002). Again, there was no difference between thymus-bearing mice injected with rhIL-7 (▴, dashed line, n = 6) or vehicle (♦, dashed line, n = 6).

Fig. 5.

IL-7 potently restores both recall and primary responses to HY-disparate skin grafts in T-cell–depleted hosts receiving suboptimal T cell inocula.

(A) TXY/TCD mice were grafted with male tail skin and injected with 1 × 106 primed LN cells via tail vein 24 hours after grafting. Then rhIL-7 was injected intraperitoneally at a dose of 5 μg/d beginning on the day of primed cell injection and continuing for 28 days. Animals receiving rhIL-7 and 1 × 106 primed LN cells, shown in previous experiments to be a suboptimal inocula (▪, n = 8), rapidly rejected HY-disparate skin grafts at nearly the same rate as thymus-bearing animals (▴, dashed line, n = 9,P = .2) and significantly faster than animals injected with vehicle only plus primed LN cells (●, n = 9,P = .0002) or rhIL-7 without primed LN cells (♦, n = 6, P = .019). Note the lack of effect in thymus-bearing animals injected with rhIL-7 (▾, dashed line, n = 5) compared to vehicle (▴, dashed line, n = 5). (B) TXY/TCD mice were injected with 1 × 106 naive LN cells and enriched male dendritic cells. The rhIL-7 at 5 μg/d intraperitoneally was initiated on the day of cell injection and continued for 28 days. Male skin grafting was performed on day 21. Similar to recall responses, administration of rhIL-7 in the primary model led to rapid rejection of HY-disparate skin grafts in TCD mice (●, n = 8), whereas mice receiving the same inocula and injected with vehicle were completely unable to reject these grafts (▪, n = 7,P = .0002). Again, there was no difference between thymus-bearing mice injected with rhIL-7 (▴, dashed line, n = 6) or vehicle (♦, dashed line, n = 6).

Close modal

Sensitization with IL-7Rα−/− antigen-presenting cells partially abrogates the effect of IL-7 on naive T-cell responses

To dissect possible effects of IL-7 on antigen-presenting cells (APCs) from those on expanding T cells, mice were injected with a suboptimal naive T-cell inocula as before, along with a sensitizing population of enriched dendritic cells from IL7Rα−/−males. Thus, the peripherally expanding T cells would still respond to IL-7, and the APCs used for sensitization would be unresponsive due to a lack of a functional receptor. Although the dendritic cell yield per animal was lower from the IL-7Rα−/− mice than from IL-7Rα+/+ animals, the phenotype was identical (Figure6). Further, as seen in Figure7, IL-7Rα−/− APCs are able to induce HY graft rejection in animals receiving a sufficient LN inocula (25 × 106) at essentially the same rate as thymus-bearing animals, thus establishing functionality of these APCs. However, when an insufficient LN inocula was sensitized with enriched IL-7Rα−/− dendritic cells, administration of rhIL-7 resulted in significantly delayed rejection relative to mice receiving enriched dendritic cells capable of responding to IL-7 (P = .006). Therefore, a partial loss of the effect of IL-7 on restoration of naive responses was seen, demonstrating that the effects of IL-7 in this model are not limited to peripherally expanding T cells but also include modulation of APCs.

Fig. 6.

Identical phenotype of enriched dendritic cells from normal and IL-7Rα−/− males.

Dendritic cells were enriched from male splenocytes following plate adherence and incubation with IL-4 and GM-CSF as described in “Materials and methods.” Before injection, portions of the cells were analyzed by flow cytometry. Staining with FITC-conjugated antibodies expressed by dendritic cells were analyzed against a cocktail of lineage-specific antibodies conjugated to phycoerythrin (PE). The dashed circle in each graph delineates the dendritic cells. Staining for PE in the indicated cells represents intermediate labeling with anti-CD11b with no expression of B220 or CD3 (data not shown), a finding consistent with the observations of other investigators.65 The phenotype of dendritic cells from normal (top panels) and IL-7Rα−/−mice (bottom panels), as characterized by this set of markers, was identical. MHC indicates major histocompatibility complex.

Fig. 6.

Identical phenotype of enriched dendritic cells from normal and IL-7Rα−/− males.

Dendritic cells were enriched from male splenocytes following plate adherence and incubation with IL-4 and GM-CSF as described in “Materials and methods.” Before injection, portions of the cells were analyzed by flow cytometry. Staining with FITC-conjugated antibodies expressed by dendritic cells were analyzed against a cocktail of lineage-specific antibodies conjugated to phycoerythrin (PE). The dashed circle in each graph delineates the dendritic cells. Staining for PE in the indicated cells represents intermediate labeling with anti-CD11b with no expression of B220 or CD3 (data not shown), a finding consistent with the observations of other investigators.65 The phenotype of dendritic cells from normal (top panels) and IL-7Rα−/−mice (bottom panels), as characterized by this set of markers, was identical. MHC indicates major histocompatibility complex.

Close modal
Fig. 7.

Lack of IL-7 signaling in APCs leads to a partial abrogation of the effect of IL-7 in restoring primary responses to HY skin grafts.

TXY/TCD mice were injected with naive LN cells and 1 × 105 enriched dendritic cells from the spleens of male IL-7Rα−/− mice or normal, IL-7Rα+/+males. The rhIL-7 at 5 μg/d or vehicle was initiated on the day of cell injections and continued for 28 days. Male skin grafting was performed on day 21. IL-7Rα−/− APCs are functionally able to induce HY graft rejection in animals receiving a sufficient T-cell inocula (25 × 106) with vehicle (▪, n = 5) at a rate analogous to thymus-bearing animals receiving enriched IL-7Rα+/+ dendritic cells (▾, n = 5). Mice receiving an insufficient LN inocula (1 × 106) with vehicle are completely unable to reject these grafts (♦, n = 6). Mice injected with a suboptimal T-cell inocula, sensitized with IL-7R−/− APCs and given rhIL-7 (●, n = 11) rejected HY-disparate skin grafts but significantly slower than mice receiving IL-7Rα+/+ dendritic cells (▴, n = 10,P = .006).

Fig. 7.

Lack of IL-7 signaling in APCs leads to a partial abrogation of the effect of IL-7 in restoring primary responses to HY skin grafts.

TXY/TCD mice were injected with naive LN cells and 1 × 105 enriched dendritic cells from the spleens of male IL-7Rα−/− mice or normal, IL-7Rα+/+males. The rhIL-7 at 5 μg/d or vehicle was initiated on the day of cell injections and continued for 28 days. Male skin grafting was performed on day 21. IL-7Rα−/− APCs are functionally able to induce HY graft rejection in animals receiving a sufficient T-cell inocula (25 × 106) with vehicle (▪, n = 5) at a rate analogous to thymus-bearing animals receiving enriched IL-7Rα+/+ dendritic cells (▾, n = 5). Mice receiving an insufficient LN inocula (1 × 106) with vehicle are completely unable to reject these grafts (♦, n = 6). Mice injected with a suboptimal T-cell inocula, sensitized with IL-7R−/− APCs and given rhIL-7 (●, n = 11) rejected HY-disparate skin grafts but significantly slower than mice receiving IL-7Rα+/+ dendritic cells (▴, n = 10,P = .006).

Close modal

Depletion of T cells is a hallmark of a number of disease processes and is an anticipated side effect of therapeutic modalities such as cancer chemotherapy and stem cell transplantation. Although much has been learned about the pathways that lead to T-cell immune reconstitution, including the substantial degree to which thymic-independent pathways can contribute to this process, it remains unclear to what extent thymic-independent pathways can restore host immune competence. In this report, we describe a model that illustrates functional deficits in immune responses to nominal antigen after thymic-independent T-cell regeneration. Using this model, we have shown that immune competence can be restored in the absence of thymic pathways by modulation of the process of peripheral expansion by increasing T-cell inocula, by inhibiting programmed cell death, and by cytokine administration.

A number of interesting findings emerge from these experiments. First, in this murine model, the number of T cells required to restore host immune competence (25 × 106 LN cells or approximately 20 × 106 T cells) is approximately equal to 10% of the total T-cell number estimated for an intact mouse.52 Although the number required would be likely to vary depending on the antigen and the precursor frequency of the antigen-specific T cells within the inocula, these results illustrate the degree of redundancy that exists within the immune system. What remains unclear is whether this number is relative and must be increased proportionally to restore immune competence in larger hosts (such as humans) or whether this represents an absolute number of T-cell receptor specificities required for response to diverse environmental antigens. Second, the finding that restoration of immune responses can occur in the absence of newly developed T-cell receptor specificities via thymic pathways emphasizes that the deficits in T-cell receptor repertoire that result from peripheral expansion are relative rather than absolute. Thus, by improving the efficiency of peripheral expansion by inhibiting cell death and, potentially, by increasing APC capacity, substantially lower numbers of antigen-specific T cells may be adequate for successful induction of immune responses. Third, the data show a clear role for programmed cell death in limiting immune responses after thymic-independent peripheral expansion. Although this is suggested by the finding of increased annexin V staining on peripherally expanded T cells manipulated ex vivo, it is more definitively shown by the increased number of T cells derived from hbcl-2 transgenic inocula and, more importantly, by restoration of immune responses to nominal antigen by constitutive bcl-2 transgene expression. Thus, peripherally expanding T cells that would otherwise be lost to apoptotic cell death are rescued and, therefore, contribute to the reconstitution of the T-cell pool. However, restoration of immune responses by inhibition of apoptosis appears to be confined to the setting of recall responses indicating important differences in the requirements for induction of responses to recall and naive antigens after thymic-independent T-cell reconstitution. This phenomenon may partially explain the restoration of recall responses and protection from opportunistic infections, which has been observed during immune reconstitution of HIV-infected patients53-56 and cannot be clearly attributed to restoration of T-cell repertoire via thymic-dependent pathways.57 

The widely accepted model for T-cell death holds that there are 2 primary pathways that ultimately end in apoptosis.58,59The active process requires T-cell receptor stimulation, is mediated by Fas ligand and other tumor necrosis factor (TNF) family molecules, occurs at effective cytokine concentrations, and is inefficiently inhibited by bcl-2. The passive pathway, such as occurs with insufficient cytokine support, is strongly inhibited by bcl-2. Based on this model, our results present a potential paradox. The degree of activation of peripherally expanded T cells and the requirement for antigen would suggest that the limitations in immune responses observed in our experiments are the result of active apoptosis of peripherally expanding T cells. These models would predict that bcl-2 would be unable to restore immune responses in this setting. A potential explanation for this finding may be that there is some degree of passive cell death occurring during peripheral expansion due to competition for limiting growth factors. Alternatively, it has been reported that up-regulation of bcl-2 family members can lead to inhibition of Fas-mediated cell death in some situations.60 61 Whether inhibition of the processes involved in active cell death would have a more potent effect on immune responses in these models is currently under study.

Interleukin-7 is a cytokine with extremely diverse and potent effects on T- and B- lineage cells. It is uniquely suited to restore immune competence after TCD because of its effects on multiple pathways of T-cell regeneration. The effects of IL-7 on thymopoiesis have been well characterized. Emerging data suggest that IL-7 may also have effects on extrathymic T-cell differentiation (K. Weinberg, written communication, 2000) potentially providing a partial explanation for the restoration of responses to HY antigen observed in some animals when rhIL-7 is administered without primed T cells. In addition, IL-7 has been shown to induce proliferation of naive T cells without altering the phenotype.62 However, the effects of IL-7 on mature T-cell populations in vivo are less well characterized and the ability for this cytokine to enhance immune reconstitution via thymic-independent peripheral expansion has not been reported previously. In this report we show that IL-7 potently modulates the peripheral expansion pathway to T-cell regeneration. This action appears to be multifactorial and due to effects on expanding T cells as well as effects on APC populations. Modulation of APCs by IL-7 has not been reported and the mechanisms of this effect are currently being investigated in more detail (K. Komschlies, in preparation). Importantly, we have found IL-7 to be unique among the T-cell–active cytokines tested in inducing peripheral expansion.63 This finding raises the possibility that endogenously produced IL-7 could also play a role in regulating this process during T-cell regeneration and thus contribute to T-cell homeostasis. Indeed, it is well known that TCD leads to enhanced expansion of mature T cells and preliminary results from our laboratory (Fry et al, manuscript submitted) in patients with HIV and studies by others64 have shown elevated serum IL-7 levels in patients with TCD.

Interleukin-7 may have potential application as a biologic modifier in a number of clinical situations. First, after stem cell transplantation, prolonged lymphodepletion results in susceptibility to infection that contributes to transplant-related morbidity, especially in allogeneic transplants. Administration of IL-7 during this period would be predicted to lead to more rapid lymphocyte recovery, improved responses to foreign antigens, and a reduction in mortality from infections in the posttransplant period. Although there are clearly inherent differences between our model, which uses antibody depletion of T cells, and a stem cell transplantation model that uses cytotoxic conditioning regimens, there are many similarities including the reliance on peripheral expansion and the contribution of residual T cells to immune reconstitution. Thus, these findings are likely to be relevant to clinical transplantation. Second, although there has been success in substantially reducing viral load in patients with HIV infection, a proportion of patients fail to show complete immunologic recovery and discontinuation of antiretroviral agents results in eventual rebound in the viral load. Therefore, one of the current challenges in the treatment of HIV lies in restoring immune responses to the virus. IL-7, with its ability to restore both recall and naive responses after TCD, has the potential to be an excellent adjuvant in this setting. Whether activation of resting T cells will contribute to infection of new cells is not known. Lastly, immunotherapeutic approaches to cancer treatment will probably be most effective in the eradication of minimal residual disease remaining after standard cytotoxic therapy. However, as indicated by these findings, the TCD that results will likely provide a major barrier to the success of these approaches. Immune adjuvants such as IL-7 might vastly improve the results of cancer immunotherapy trials that are attempted in the setting of TCD.

We would like to thank Dr Scott Durum for his careful review of this manuscript.

The contents of this publication do not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government. This project was funded in whole or part with funds from the National Cancer Institute, National Institutes of Health, under contract no. NO1-CO-56000. By acceptance of this article, the publisher or recipient acknowledges right of the U.S. Government to retain a nonexclusive, royalty-free license in and to any copyright covering the article.

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 U.S.C. section 1734.

1
Mackall
CL
Hakim
FT
Gress
RE
Restoration of T-cell homeostasis after T-cell depletion.
Semin Immunol.
9
1997
339
346
2
Mackall
CL
Fleisher
TA
Brown
MR
et al
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy [see comments].
N Engl J Med.
332
1995
143
149
3
Roux
E
Helg
C
Dumont-Girard
F
Chapuis
B
Jeannet
M
Roosnek
E
Analysis of T-cell repopulation after allogeneic bone marrow transplantation: significant differences between recipients of T-cell depleted and unmanipulated grafts.
Blood.
87
1996
3984
3992
4
Soiffer
RJ
Bosserman
L
Murray
C
Cochran
K
Daley
J
Ritz
J
Reconstitution of T-cell function after CD6-depleted allogeneic bone marrow transplantation.
Blood.
75
1990
2076
2084
5
Storek
J
Witherspoon
RP
Storb
R
T cell reconstitution after bone marrow transplantation into adult patients does not resemble T cell development in early life.
Bone Marrow Transplant.
16
1995
413
425
6
de Gast
GC
Verdonck
LF
Middeldorp
JM
et al
Recovery of T cell subsets after autologous bone marrow transplantation is mainly due to proliferation of mature T cells in the graft.
Blood.
66
1985
428
431
7
Walker
RE
Carter
CS
Muul
L
et al
Peripheral expansion of pre-existing mature T cells is an important means of CD4+ T-cell regeneration HIV-infected adults.
Nat Med.
4
1998
852
856
8
Weinberg
K
Annett
G
Kashyap
A
Lenarsky
C
Forman
SJ
Parkman
R
The effect of thymic function on immunocompetence following bone marrow transplantation.
Biol Blood Marrow Transplant.
1
1995
18
23
9
Miller
RA
Stutman
O
T cell repopulation from functionally restricted splenic progenitors: 10,000-fold expansion documented by using limiting dilution analyses.
J Immunol.
133
1984
2925
2932
10
Rocha
B
Dautigny
N
Pereira
P
Peripheral T lymphocytes: expansion potential and homeostatic regulation of pool sizes and CD4/CD8 ratios in vivo.
Eur J Immunol.
19
1989
905
911
11
Bell
EB
Sparshott
SM
Drayson
MT
Hunt
SV
The origin of T cells in permanently reconstituted old athymic nude rats. Analysis using chromosome or allotype markers.
Immunology.
68
1989
547
556
12
Bell
EB
Sparshott
SM
Drayson
MT
Ford
WL
The stable and permanent expansion of functional T lymphocytes in athymic nude rats after a single injection of mature T cells.
J Immunol.
139
1987
1379
1384
13
Mackall
CL
Granger
L
Sheard
MA
Cepeda
R
Gress
RE
T-cell regeneration after bone marrow transplantation: differential CD45 isoform expression on thymic-derived versus thymic-independent progeny.
Blood.
82
1993
2585
2594
14
Rocha
B
von Boehmer
H
Peripheral selection of the T cell repertoire.
Science.
251
1991
1225
1228
15
Mackall
CL
Bare
CV
Granger
LA
Sharrow
SO
Titus
JA
Gress
RE
Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing.
J Immunol.
156
1996
4609
4616
16
Jendro
MC
Ganten
T
Matteson
EL
Weyand
CM
Goronzy
JJ
Emergence of oligoclonal T cell populations following therapeutic T cell depletion in rheumatoid arthritis.
Arthritis Rheum.
38
1995
1242
1251
17
Gorski
J
Yassai
M
Zhu
X
et al
Circulating T cell repertoire complexity in normal individuals and bone marrow recipients analyzed by CDR3 size spectratyping. Correlation with immune status.
J Immunol.
152
1994
5109
5119
18
Mackall
CL
Hakim
FT
Gress
RE
T-cell regeneration: all repertoires are not created equal.
Immunol Today.
18
1997
245
251
19
Gea-Banacloche
JC
Weiskopf
EE
Hallahan
C
et al
Progression of human immunodeficiency virus disease is associated with increasing disruptions within the CD4+ T cell receptor repertoire.
J Infect Dis.
177
1998
579
585
20
Hakim
FT
Cepeda
R
Kaimei
S
et al
Constraints on CD4 recovery postchemotherapy in adults: thymic insufficiency and apoptotic decline of expanded peripheral CD4 cells.
Blood.
90
1997
3789
3798
21
Lenardo
M
Chan
KM
Hornung
F
et al
Mature T lymphocyte apoptosis—immune regulation in a dynamic and unpredictable antigenic environment.
Annu Rev Immunol.
17
1999
221253
22
Boise
LH
Thompson
CB
Hierarchical control of lymphocyte survival.
Science.
274
1996
67
68
23
Sarin
A
Clerici
M
Blatt
SP
Hendrix
CW
Shearer
GM
Henkart
PA
Inhibition of activation-induced programmed cell death and restoration of defective immune responses of HIV+ donors by cysteine protease inhibitors.
J Immunol.
153
1994
862
872
24
Clerici
M
Sarin
A
Berzofsky
JA
et al
Antigen-stimulated apoptotic T-cell death in HIV infection is selective for CD4+ T cells, modulated by cytokines and effected by lymphotoxin.
AIDS.
10
1996
603
611
25
Meyaard
L
Otto
SA
Jonker
RR
Mijnster
MJ
Keet
RP
Miedema
F
Programmed death of T cells in HIV-1 infection.
Science.
257
1992
217
219
26
Groux
H
Torpier
G
Monte
D
Mouton
Y
Capron
A
Ameisen
JC
Activation-induced death by apoptosis in CD4+ T cells from human immunodeficiency virus-infected asymptomatic individuals.
J Exp Med.
175
1992
331
340
27
Gougeon
ML
Lecoeur
H
Dulioust
A
et al
Programmed cell death in peripheral lymphocytes from HIV-infected persons: increased susceptibility to apoptosis of CD4 and CD8 T cells correlates with lymphocyte activation and with disease progression.
J Immunol.
156
1996
3509
3520
28
Finkel
TH
Tudor-Williams
G
Banda
NK
et al
Apoptosis occurs predominantly in bystander cells and not in productively infected cells of HIV- and SIV-infected lymph nodes [see comments].
Nat Med.
1
1995
129
134
29
Muro-Cacho
CA
Pantaleo
G
Fauci
AS
Analysis of apoptosis in lymph nodes of HIV-infected persons. Intensity of apoptosis correlates with the general state of activation of the lymphoid tissue and not with stage of disease or viral burden.
J Immunol.
154
1995
5555
5566
30
Zhang
ZQ
Notermans
DW
Sedgewick
G
et al
Kinetics of CD4+ T cell repopulation of lymphoid tissues after treatment of HIV-1 infection.
Proc Natl Acad Sci U S A.
95
1998
1154
1159
31
Brugnoni
D
Airo
P
Pennacchio
M
et al
Immune reconstitution after bone marrow transplantation for combined immunodeficiencies: down-modulation of Bcl-2 and high expression of CD95/Fas account for increased susceptibility to spontaneous and activation-induced lymphocyte cell death.
Bone Marrow Transplant.
23
1999
451
457
32
Komschlies
KL
Grzegorzewski
KJ
Wiltrout
RH
Diverse immunological and hematological effects of interleukin 7: implications for clinical application.
J Leukoc Biol.
58
1995
623
633
33
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.
181
1995
1519
1526
34
Peschon
JJ
Morrissey
PJ
Grabstein
KH
et al
Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice.
J Exp Med.
180
1994
1955
1960
35
Akashi
K
Kondo
M
von Freeden-Jeffry
U
Murray
R
Weissman
IL
Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice.
Cell.
89
1997
1033
1041
36
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.
89
1997
1011
1019
37
Abdul-Hai
A
Or
R
Slavin
S
et al
Stimulation of immune reconstitution by interleukin-7 after syngeneic bone marrow transplantation in mice [published erratum appears in Exp Hematol 1996 Nov;24(13):1540].
Exp Hematol.
24
1996
1416
1422
38
Abdul-Hai
A
Ben-Yehuda
A
Weiss
L
et al
Interleukin-7-enhanced cytotoxic T lymphocyte activity after viral infection in marrow transplanted mice.
Bone Marrow Transplant.
19
1997
539
543
39
Bolotin
E
Smogorzewska
M
Smith
S
Widmer
M
Weinberg
K
Enhancement of thymopoiesis after bone marrow transplant by in vivo interleukin-7.
Blood.
88
1996
1887
1894
40
Morrissey
PJ
Conlon
P
Braddy
S
Williams
DE
Namen
AE
Mochizuki
DY
Administration of IL-7 to mice with cyclophosphamide-induced lymphopenia accelerates lymphocyte repopulation.
J Immunol.
146
1991
1547
1552
41
Boerman
OC
Gregorio
TA
Grzegorzewski
KJ
et al
Recombinant human IL-7 administration in mice affects colony-forming units-spleen and lymphoid precursor cell localization and accelerates engraftment of bone marrow transplants.
J Leukoc Biol.
58
1995
151
158
42
Vella
AT
Dow
S
Potter
TA
Kappler
J
Marrack
P
Cytokine-induced survival of activated T cells in vitro and in vivo.
Proc Natl Acad Sci U S A.
95
1998
3810
3815
43
Vella
A
Teague
TK
Ihle
J
Kappler
J
Marrack
P
Interleukin 4 (IL-4) or IL-7 prevents the death of resting T cells: stat6 is probably not required for the effect of IL-4.
J Exp Med.
186
1997
325
330
44
Seki
H
Iwai
K
Kanegane
H
et al
Differential protective action of cytokines on radiation-induced apoptosis of peripheral lymphocyte subpopulations.
Cell Immunol.
163
1995
30
36
45
Hernandez-Caselles
T
Martinez-Esparza
M
Sancho
D
Rubio
G
Aparicio
P
Interleukin-7 rescues human activated T lymphocytes from apoptosis induced by glucocorticosteroids and regulates bcl-2 and CD25 expression.
Hum Immunol.
43
1995
181
189
46
Hassan
J
Reen
DJ
IL-7 promotes the survival and maturation but not differentiation of human post-thymic CD4+ T cells.
Eur J Immunol.
28
1998
3057
3065
47
Boise
LH
Minn
AJ
June
CH
Lindsten
T
Thompson
CB
Growth factors can enhance lymphocyte survival without committing the cell to undergo cell division.
Proc Natl Acad Sci U S A.
92
1995
5491
5495
48
Strasser
A
Harris
AW
Vaux
DL
et al
Abnormalities of the immune system induced by dysregulated bcl-2 expression in transgenic mice.
Curr Top Microbiol Immunol.
166
1990
175
181
49
Billingham
R
Medawar
PB
The technique of free skin grafting in mammals.
J Exp Biol.
28
1951
385
50
Strasser
A
Harris
AW
Cory
S
bcl-2 transgene inhibits T cell death and perturbs thymic self- censorship.
Cell.
67
1991
889
899
51
Sentman
CL
Shutter
JR
Hockenbery
D
Kanagawa
O
Korsmeyer
SJ
bcl-2 inhibits multiple forms of apoptosis but not negative selection in thymocytes.
Cell.
67
1991
879
888
52
Jerne
NK
Idiotypic networks and other preconceived ideas.
Immunol Rev.
79
1984
5
24
53
Connick
E
Lederman
MM
Kotzin
BL
et al
Immune reconstitution in the first year of potent antiretroviral therapy and its relationship to virologic response.
J Infect Dis.
181
2000
358
363
54
Lederman
MM
Connick
E
Landay
A
et al
Immunologic responses associated with 12 weeks of combination antiretroviral therapy consisting of zidovudine, lamivudine, and ritonavir: results of AIDS Clinical Trials Group Protocol 315.
J Infect Dis.
178
1998
70
79
55
Rinaldo
CR
Jr
Liebmann
JM
Huang
XL
et al
Prolonged suppression of human immunodeficiency virus type 1 (HIV-1) viremia in persons with advanced disease results in enhancement of CD4 T cell reactivity to microbial antigens but not to HIV-1 antigens.
J Infect Dis.
179
1999
329
336
56
Pontesilli
O
Kerkhof-Garde
S
Notermans
DW
et al
Functional T cell reconstitution and human immunodeficiency virus-1- specific cell-mediated immunity during highly active antiretroviral therapy.
J Infect Dis.
180
1999
76
86
57
Connors
M
Kovacs
JA
Krevat
S
et al
HIV infection induces changes in CD4+ T-cell phenotype and depletions within the CD4+ T-cell repertoire that are not immediately restored by antiviral or immune-based therapies [see comments].
Nat Med.
3
1997
533
540
58
Strasser
A
Harris
AW
Huang
DC
Krammer
PH
Cory
S
Bcl-2 and Fas/APO-1 regulate distinct pathways to lymphocyte apoptosis.
Embo J.
14
1995
6136
147
59
Van Parijs
L
Biuckians
A
Abbas
AK
Functional roles of Fas and Bcl-2-regulated apoptosis of T lymphocytes.
J Immunol.
160
1998
2065
2071
60
Zhang
X
Li
L
Choe
J
et al
Up-regulation of Bcl-xL expression protects CD40-activated human B cells from Fas-mediated apoptosis.
Cell Immunol.
173
1996
149
154
61
Boise
LH
Thompson
CB
Bcl-x(L) can inhibit apoptosis in cells that have undergone Fas-induced protease activation.
Proc Natl Acad Sci U S A.
94
1997
3759
3764
62
Soares
MV
Borthwick
NJ
Maini
MK
Janossy
G
Salmon
M
Akbar
AN
IL-7-dependent extrathymic expansion of CD45RA+ T cells enables preservation of a naive repertoire.
J Immunol.
161
1998
5909
5917
63
Mackall CL, Fry TJ, Bare C, Morgan P, Galbraith A, Gress RE. IL-7 increases both thymic-dependent and thymic-independent pathways after bone marrow transplantation. Blood. In press.
64
Bolotin
E
Annett
G
Parkman
R
Weinberg
K
Serum levels of IL-7 in bone marrow transplant recipients: relationship to clinical characteristics and lymphocyte count.
Bone Marrow Transplant.
23
1999
783
788
65
Metlay
JP
Witmer-Pack
MD
Agger
R
Crowley
MT
Lawless
D
Steinman
RM
The distinct leukocyte integrins of mouse spleen dendritic cells as identified with new hamster monoclonal antibodies.
J Exp Med.
171
1990
1753
1771

Author notes

Terry J. Fry, Bldg 10, Rm 13N240, MSC 1928, 10 Center Dr, Bethesda, MD, 20892-1928; e-mail: tf60y@nih.gov.

Sign in via your Institution