Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Top Downloads
    • Annual Meeting Abstracts
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • Rights and Permissions
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • Log in
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Top Downloads
    • Annual Meeting Abstracts
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • Rights and Permissions
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

CD8+, αβ-TCR+, and γδ-TCR+ Cells in the Recipient Hematopoietic Environment Mediate Resistance to Engraftment of Allogeneic Donor Bone Marrow

Hong Xu, Beate G. Exner, Daniel E. Cramer, Michael K. Tanner, Yvonne M. Mueller and Suzanne T. Ildstad
J Immunol February 15, 2002, 168 (4) 1636-1643; DOI: https://doi.org/10.4049/jimmunol.168.4.1636
Hong Xu
Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Beate G. Exner
Department of General and Thoracic Surgery, University of Kiel, Kiel, Germany; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Daniel E. Cramer
Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael K. Tanner
Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yvonne M. Mueller
Department of Microbiology and Immunology, Medical College of Pennsylvania, Hahnemann University, Philadelphia, PA 19129
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Suzanne T. Ildstad
Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Historically, conditioning for engraftment of hematopoietic stem cells has been nonspecific. In the present study, we characterized which cells in the recipient hematopoietic microenvironment prevent allogeneic marrow engraftment. Mice defective in production of αβ-TCR+, γδ-TCR+, αβ- plus γδ-TCR+, CD8+, or CD4+ cells were transplanted with MHC-disparate allogeneic bone marrow. Conditioning with 500 cGy total body irradiation (TBI) plus a single dose of cyclophosphamide (CyP) on day +2 establishes chimerism in normal recipients. When mice were conditioned with 300 cGy TBI plus a single dose of CyP on day +2, all engrafted, except wild-type controls and those defective in production of CD4+ T cells. Mice lacking both αβ- and γδ-TCR+ cells engrafted without conditioning, suggesting that both αβ- and γδ-TCR T cells in the host play critical and nonredundant roles in preventing engraftment of allogeneic bone marrow. CD8 knockout (KO) mice engrafted without TBI, but only if they received CyP on day +2 relative to the marrow infusion, showing that a CD8− cell was targeted by the CyP conditioning. The CD8+ cell effector function is mechanistically different from that for conventional T cells, and independent of CD4+ T helper cells because CD4 KO mice require substantially higher levels of conditioning than the other KO phenotypes. These results suggest that a number of cell populations with different mechanisms of action mediate resistance to engraftment of allogeneic marrow. Targeting of specific recipient cellular populations may permit conditioning approaches to allow mixed chimerism with minimal morbidity and could potentially avoid the requirement for myelotoxic agents altogether.

The replacement of a failing organ with a functional one has become a clinical reality. However, organ transplantation currently relies upon the chronic use of nonspecific immunosuppressive agents to promote graft survival. The use of these agents is associated with opportunistic infection (1), an increased rate of malignancy (2), and end organ toxicities (3, 4), including nephrotoxicity and hepatotoxicity. Although these agents successfully prevent acute rejection, chronic rejection remains the primary cause of late graft loss (5). The five-year survival for renal allografts is 74% (6), cardiac transplants 68%, and pulmonary allografts 43% (7).

Mixed allogeneic chimerism has been suggested as a potential approach to induce tolerance to solid organ allografts (8). Mixed chimerism has the following advantages: 1) a relative resistance to graft-vs-host disease (GVHD)4 (3, 9); 2) superior immunocompetence (10, 11); 3) it can be established with partial myeloablative conditioning approaches (10, 12, 13); and 4) it prevents the development of chronic rejection in allografts (14).

Approaches to establish mixed chimerism with partial conditioning have been successfully pursued. Most conditioning approaches have used a combination of irradiation, other myelotoxic agents, and nonspecific immunosuppression (15, 16, 17, 18, 19, 20, 21, 22). We previously developed a total body irradiation (TBI)-based model to establish durable multilineage mixed chimerism in MHC plus minor Ag disparate recipients (12). Seven hundred centigray of TBI alone was required for 100% engraftment of MHC-disparate allogeneic bone marrow. The posttransplant administration of cyclophosphamide (CyP) (day +2) significantly reduced the minimum dose of TBI sufficient to establish mixed chimerism in 100% of recipients to 500 cGy (12). The addition of anti-lymphocyte globulin (ALG) (day −3) to the peritransplant-conditioning further reduced the minimum TBI dose to 200 cGy (23). We hypothesized that CyP and ALG removed activated alloreactive host cells from the repertoire. In the present study, we have characterized which host effector cells mediate alloresistance for engraftment of allogeneic bone marrow using mice genetically modified to lack production of specific cell types (knockout (KO) mice). We report for the first time that αβ-TCR+ as well as γδ-TCR+ T cells play critical and nonredundant roles in alloresistance to engraftment. Their mechanism of action was CyP, and to a slightly lesser extent, radiation-sensitive, because mice that did not produce αβ- and γδ-T cells did not require any conditioning, while mice lacking only γδ-T cells or only αβ-T cells required low-dose TBI plus CyP to engraft. In addition, CD8+ cells in the recipient exert a CD4-independent effect, as shown by requirement for significant conditioning for CD4 KO mice to engraft. A better understanding of the mechanism of engraftment and the role of host conditioning will enable development of cell-specific partial conditioning approaches associated with less toxicity than the nonspecific agents currently in use.

Materials and Methods

Mice

Transplant donor, B10.BR.SgSnJ (B10.BR; H-2k), and various recipient mice: C57BL/6J (B6; H2b), C57BL/6-TcrbtmlMom (TCR-β KO, as αβ-TCR T cell-deficient recipients), C57BL/6-TcrdtmlMom (TCR-δ KO, as γδ-T cell-deficient recipients), C57BL/6-TcrbtmlMom TcrdtmlMom (TCR-β/TCR-δ double KO, as αβ- and γδ-T cell deficient recipients), C57BL/6-Cd8Tm/mak (CD8 KO), C57BL/10-Cd4tml (CD4 KO), and C57BL/6J-CD4tm/mak (CD4 KO) were purchased from The Jackson Laboratory (Bar Harbor, ME). Mice were housed in a special pathogen-free barrier facility at the Institute for Cellular Therapeutics (Louisville, KY). Mice were cared for according to National Institutes of Health animal care guidelines.

Chimera preparation

Bone marrow was prepared from B10.BR donor mice as previously described (8). Briefly, B10.BR donor mice were euthanized and tibias and femurs were harvested. Bone marrow was expelled from the bones with Media 199 (Life Technologies, Grand Island, NY) containing 10 μg/ml Gentamicin (Life Technologies), referred to thereafter as chimera media (CM). The cells were filtered through sterile nylon mesh with 100 μm pores, centrifuged at 1000 rpm for 10 min at 4°C, and resuspended in CM. A cell count was performed, and the cells were diluted to a final concentration of 15 × 106 bone marrow cells per 1 ml of CM.

Recipient mice were treated with 0–300 cGy TBI from a cesium source (Gamma-cell 40, Nordion, Ontario, Canada). Animals were transplanted with 1 ml CM containing 15 × 106 B10.BR bone marrow cells via lateral tail vein injection within 4–6 h after irradiation. All animals in groups treated with CyP received a single i.p. injection of 200 mg/kg CyP (Sigma Aldrich, St. Louis, MO) 48 h after bone marrow transplantation (BMT). Each experiment was repeated at least three times.

Flow cytometric analysis

The level of hematopoietic chimerism was assessed monthly after BMT by flow cytometric analysis of PBL using mAb against MHC Ags of donor and host origin. Fifty microliters of whole blood obtained by tail-bleeding of the mice were incubated at room temperature for 8 min with lysing buffer (8.29 g of NH4Cl, 1.0 g of KHCO3, and 0.0372 g Na2EDTA in 1 liter H2O; prepared in our laboratory) to eliminate RBC. The leukocytes were then incubated with 10 μl diluted mAb for 30 min at 4°C in the dark. The appropriate dilution for the use of the mAb was determined in titration experiments before use. The cells were washed twice with 2 ml of FACS medium (0.36 g NaHCO3, 1.0 g NaN3, and 1.0 g BSA in 1 liter HBSS; prepared in our laboratory) and centrifuged at 1000 rpm for 10 min at 4°C. Finally, the cells were fixed with 1% paraformaldehyde in PBS (prepared in our laboratory). The analysis was conducted on a FACSCalibur (BD Biosciences, Mountain View, CA) with CellQuest software (BD Biosciences).

mAbs

Abs specific for MHC class I Ags of donor FITC-conjugated anti-H2Kk, (36-7-5, mouse IgG1) and recipient PE-conjugated anti-H2Kb (AF6-88.5, mouse IgG2a) origin were used to determine the percentage of donor cell chimerism in the recipient’s peripheral blood. Multilineage engraftment was assessed by staining with biotinylated anti-H2Kk (36-7-5, mouse IgG1), FITC-conjugated anti-H2Kb (AF6-88.5, mouse IgG2a) and PE-conjugated lineage markers 3 mo after BMT. The biotinylated Ab was counterstained with streptavidin-allophycocyanin. The following Abs were used as lineage markers: anti-GR-1 (RB6-8C5, rat IgG2b), anti-MAC-1 (M1/70, rat IgG2b), anti-CD4 (RM4-5, rat IgG2a), anti-CD8α (53-6.7, rat IgG2a), anti-B220 (RA3-6B2, rat IgG2a), anti-NK1.1 (PK136, mouse IgG2a), anti-TCR-β-chain (H57-597, hamster IgG), and anti-δ-TCR (GL3, hamster IgG). NK subpopulations were assessed by four-color staining in naive B6, TCR-β KO, TCR-δ KO, TCR-β/TCR-δ KO, and CD8 KO mice with FITC-conjugated anti-TCR-β/TCR-γδ, PerCP-conjugated anti-CD3e (145-2C11, hamster IgG), APC-conjugated anti-CD8a, and PE-conjugated NK subpopulation markers. The following Abs were used as NK subpopulation markers: anti-NK1.1, anti-5E6 (5E6, mouse IgG2a), anti-2B4 (2B4, mouse IgG2b), and anti-DX5 (DX5, rat IgM). Nonspecific background staining was controlled by using isotype control Abs directed against irrelevant Ags conjugated with the same color as the experimental Ab (i.e., anti-TNP mouse IgG2a Ags conjugated with PE served as isotype control for PE-conjugated anti-H2Kb mouse IgG2a). All mAb were obtained from BD PharMingen (San Diego, CA). Streptavidin-allophycocyanin was purchased from BD Biosciences.

Assessment of GVHD

The primary diagnosis of GVHD was based on previously described clinical criteria, which consist of diffuse erythema (particularly of the ear), hyperkeratosis of the foot pads, hair loss, weight loss, unkempt appearance, or diarrhea (24). At the time of sacrifice, sections of skin, tongue, liver, and small intestine were fixed in 10% buffered formalin, stained with H&E, and processed for light microscopy.

Skin grafting

Skin grafting was performed by a modification of the method of Billingham (25). Full-thickness tail-skin grafts were harvested from the tails of B10.BR and nonobese diabetic (NOD) (H2Kd) mice. Recipient mice were anesthetized with Nembutal (pentobarbital sodium injection; Abbott Laboratories, Abbott Park, IL), and full-thickness graft beds were prepared surgically in the lateral thoracic wall, while preserving the panniculus carnosum. The grafts were covered with a double layer of Vaseline gauze and a plaster cast. Casts were removed on day 7, and grafts were scored by daily inspection for the first month, then weekly thereafter for percentage of rejection. Rejection was defined as complete when no residual viable graft could be detected.

Statistical analysis

Statistical significance was determined with a Student’s one-way t test. The difference between groups was considered to be significant if p < 0.05.

Results

TCR-β, TCR-β/TCR-δ and CD8 KO mice require less TBI for engraftment

Conditioning with 500 cGy TBI followed by CyP treatment (day +2 relative to marrow infusion) is sufficient to establish durable chimerism in MHC-disparate as well as MHC- plus minor Ag-disparate recipients (12). The administration of CyP before transplantation is not sufficient to establish engraftment with 500 cGy TBI. We hypothesized that the administration of CyP on day +2 removes alloreactive host T-lymphocytes that have been activated by the marrow infusion. To evaluate which host cells play a role in engraftment, mice deficient in production of αβ-TCR+, γδ-TCR+, both αβ-TCR+ and γδ-TCR+, CD8+, and CD4+ cells were used as recipients. All are H-2b in MHC (B6). A total of 15 × 106 bone marrow cells from B10.BR donor mice were administered. One hundred percent of TCR-β/TCR-δ KO recipients engrafted after conditioning with 300 cGy TBI plus CyP (n = 8) (Fig. 1⇓A). The level of chimerism was 41.8 ± 1.2% 1 mo following BMT (Fig. 1⇓B). Similarly, 100% of TCR-β KO mice engrafted after conditioning with 300 cGy TBI plus CyP (n = 14; Fig. 1⇓A). The level of chimerism was similar to that for the TCR-β/TCR-δ KO recipients (42.5 ± 14%) (Fig. 1⇓B). The engraftment was durable and multilineage. Similarly, engraftment occurred in 100% of CD8 KO mice conditioned with 300 cGy TBI plus CyP (n = 16; Fig. 1⇓A). The level of chimerism was 48.7 ± 18.1% at 1 mo posttransplantation. At 3 mo, donor chimerism was 30.3 ± 8.4%, and remained stable for ≥6 mo. In striking contrast, only five of nine (56%) γδ-TCR KO recipients engrafted, and the level of chimerism was significantly lower in mice that engrafted (14.5 ± 4.3%) compared with the TCR-β KO (p < 0.005) and TCR-β/TCR-δ double KO mice (p < 0.005) (Fig. 1⇓B). CD4 KO mice conditioned in a similar fashion did not engraft (n = 20), suggesting that host CD8+ cells and αβ- and γδ-TCR+ T cells play a major role in alloresistance to engraftment, while CD4+ cells do not. Moreover, the effector cells in alloresistance did not require CD4+ cells, because CD4 KO mice require significant levels of conditioning for engraftment. As expected, B6 control mice did not engraft when conditioned, and transplanted in a similar fashion (n = 6; Fig. 1⇓A).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Characteristics of engraftment in KO mice conditioned with TBI plus CyP. B6 control mice and mice deficient in the production of γδ-TCR+ cells (TCR-δ KO), αβ-TCR+ cells (TCR-β KO), both αβ- and γδ-TCR+ cells (TCR-β/TCR-δ double KO), CD4 (CD4 KO), or CD8 (CD8 KO) were conditioned with 300 cGy TBI, transplanted with 15 × 106 bone marrow cells from B10. BR donors, and given 200 mg/kg CyP (i.p.) on day +2. This figure shows the frequency of engraftment (A) and level of chimerism in animals that engrafted (percentage of donor cells in PBL) 1 mo (B) and >3 mo after (C) as assessed by PBL typing. The results are the summary of three experiments.

Mice lacking αβ- and γδ-T cells do not require CyP conditioning for engraftment

It is hypothesized that CyP on day +2 relative to the marrow infusion removes alloreactive T lymphocytes that have been activated by alloantigen (17). To evaluate the contribution of CyP to the conditioning approach and further define which host cells were the target on the day +2 CyP infusion, TCR-β KO, TCR-δ KO, TCR-β/TCR-δ KO, and CD8 KO mice were conditioned with 300 cGy of TBI and transplanted with 15 × 106 B10.BR bone marrow cells (Fig. 2⇓). Neither the TCR-β KO (n = 6), the TCR-δ KO (n = 6), nor CD8 KO (n = 4) mice engrafted. A total of 100% of the TCR-β/TCR-δ KO mice engrafted (n = 6). The level of chimerism was 76.1 ± 10.4% 1 mo post-BMT and was durable for ≥6 mo. Therefore, these data suggest that recipient γδ-TCR+ T cells as well as αβ-TCR+ T cells contribute to alloresistance to engraftment, and support the hypothesis that alloreactive T cells are the primary target for the CyP conditioning in wild-type recipients.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Characteristics of engraftment in KO mice conditioned with TBI alone. To evaluate which cell subsets were sensitive to CyP in normal recipients, CD8 KO, TCR-β KO, TCR-δ KO, and TCR-β/TCR-δ KO mice were conditioned with 300 cGy TBI alone. Recipients were transplanted with 15 × 106 bone marrow cells from B10. BR donors on the same day with TBI. The figure shows the percentage of animals that engrafted 1 mo post-BMT. The results are the summary of three experiments.

TCR-β/TCR-δ KO mice engraft with minimal conditioning

A dose-titration of TBI was performed to determine the minimal conditioning required in mice deficient in production of αβ- and γδ-TCR+ T cells. A total of 100% of mice conditioned with 200 cGy (n = 6) or 100 cGy (n = 7) of TBI engrafted (Fig. 3⇓A). Eighty-six percent of TCR-β/TCR-δ double KO mice engrafted without any conditioning (n = 7) (Fig. 3⇓A). The level of chimerism was directly correlated with the degree of conditioning (Fig. 3⇓, B–D). The engraftment in all groups was durable for ≥5 mo.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

The influence of TBI dose on engraftment and the level of chimerism in TCR-β/TCR-δ double KO mice. TCR-β/TCR-δ KO mice were transplanted with 15 × 106 bone marrow cells from B10. BR donors after conditioning with TBI doses ranging from 0 to 300 cGy on day 0. Animals were typed by flow cytometric analysis monthly for up to 5 mo after BMT. n = number of animals in each experiment. The percentage of animals with engraftment at 1 mo (A), and the level of donor chimerism at 1 mo (B), 3 mo (C), and 5 mo (D) post-BMT are shown. The results are the summary of three experiments.

Interestingly, the level of chimerism after BMT was approximately twice as high in TCR-β/TCR-δ KO mice conditioned with 300 cGy of TBI alone (76.1 ± 10.4%) vs those conditioned with 300 cGy of TBI plus CyP (42 ± 1.2%). In the absence of the CyP-sensitive αβ- and γδ-TCR+ T cells, when CyP is not required for engraftment to occur, the impact of conditioning on the level of chimerism that results can be evaluated. Clearly, CyP itself somewhat impairs engraftment of the donor marrow. However, in the presence of host αβ- and γδ-TCR+ T cells, CyP is critical to overcoming the barrier for alloresistance because 0% of the TCR-β and TCR-δ single KO recipients engraft without CyP when conditioned with 300 cGy TBI.

Conditioning in CD8 KO mice requires CyP more than TBI

CD8 KO mice were transplanted with 15 × 106 bone marrow cells from B10.BR donors on day 0. A total of 300, 200, 100, or 0 cGy TBI were administered 4–6 h before the marrow infusion on day 0. A single dose of CyP was administered on day +2. Engraftment was present at 1 mo in 100% of recipients including those that received no TBI (Fig. 4⇓A). The level of chimerism was proportional to the dose of TBI (Fig. 4⇓B). Engraftment was durable ≥6 mo in all animals conditioned with 300 (n = 16), 200 (n = 6), and 100 (n = 6) cGy of TBI. The engraftment was multilineage for donor-derived T cells, B cells, NK cells, macrophages, and granulocytes. Half of the CD8 KO mice conditioned with only CyP (n = 6) lost their chimerism by 4 mo. CD8 KO mice did not engraft with 300 cGy TBI only, but 50% exhibited durable engraftment without TBI. If CyP were used, these data suggest that the CD8+ cell population mediating alloresistance is more sensitive to TBI than to CyP. Therefore, these data also support that a CyP-sensitive host cell in CD8 KO mice that is probably not a conventional αβ-TCR+ T cell also contributes to alloresistance.

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

TBI dose-titration in CD8 KO mice conditioned with posttransplant CyP. CD8 KO mice were conditioned with TBI doses ranging from 0 to 300 cGy on day 0 and transplanted with 15 × 106 allogeneic B10. BR bone marrow cells 4–6 h after. A total of 200 mg/kg CyP was administered 2 days after BMT. Chimerism was analyzed by flow cytometry 1 mo (B) and 6 mo (C) after BMT. The results are the summary of three experiments.

Multilineage chimerism occurs in chimeras

The pluripotent hematopoietic stem cell (HSC) produces at least 11 different lineages. To confirm that the engraftment in TCR-β/TCR-δ double KO recipients reflects engraftment of the pluripotent stem cell, animals were followed for ≥3 mo. The engraftment in TCR-β/TCR-δ double KO mice conditioned with 300 cGy irradiation and administration of CyP on day +2 was durable (Fig. 1⇑C) and multilineage. Three-color flow cytometric analysis showed the presence of multiple myeloid and lymphoid lineages of donor and host origin in all engrafted animals (Table I⇓, group A). The T cell lineages deficient in the KO animals were restored after transplant recipients, and were all of donor origin. Similarly, TCR-β/TCR-δ KO mice conditioned with 300 cGy TBI without CyP (Table I⇓; group B) as well as CD8 KO recipients conditioned with 200 cGy TBI plus CyP (Table I⇓; group C) were evaluated. In all groups, donor-derived T cells, B cells, NK cells, macrophages, and granulocytes were present, suggesting engraftment of the HSC itself rather than selected lineages.

View this table:
  • View inline
  • View popup
Table I.

Multilineage engraftment in TCR-β/TCR-δ KO and CD8 KO micea

Evidence for specific tolerance in vivo to donor-type skin grafts

Skin grafting was performed to assess donor-specific tolerance in vivo. Five unmanipulated TCR-β/TCR-δ KO mice (H2b) received full-thickness skin grafts of both B10/BR (H2k) and NOD (H2d) origin. All grafts survived >160 days, demonstrating that naive TCR-β/TCR-δ KO mice do not reject skin allografts. Both donor-specific (B10.BR) and MHC-disparate third-party (NOD) skin grafts were placed on the chimeric TCR-β/TCR-δ KO mice with different levels of donor chimerism (Table II⇓). Grafts were assessed daily for the first 4 wk and weekly thereafter for evidence of rejection. The single nonchimeric mouse accepted the donor-specific and third party skin grafts in a fashion similar to that observed in naive TCR-β/TCR-δ KO mice. In all other recipients, donor-specific allogeneic skin grafts were accepted by the mice with chimerism (range from 2.5 to 71.3%), while third-party skin grafts were promptly rejected. Therefore, these data demonstrate that TCR-β/TCR-δ KO recipients that engraft as chimeras exhibit donor-specific tolerance, but are immunocompetent to reject MHC-disparate third party allografts.

View this table:
  • View inline
  • View popup
Table II.

Donor-specific tolerange in mixed allogeneic chimerasa

Characterization of lymphoid populations in marrow and spleen of TCR-β KO, TCR-δ, TCR-β/TCR-δ KO, and CD8 KO mice

T cells, NK cells, and NK/T cells have all been implicated in alloresistance to engraftment. A number of NK subfamilies have been described, including 5E6, 2B4 (26), NK/T cells (27, 28), and CD8+ NK cells (27, 29). Marrow and splenocytes from CD8, TCR-β/TCR-δ, TCR-β, and TCR-δ KO mice were analyzed by four-color flow cytometry to enumerate which NK subfamilies might be absent (Fig. 5⇓). All KO mice produced NK1.1+ and 5E6+ cells in marrow and spleen at levels similar to wild-type B6 controls. Marrow from the TCR-β/TCR-δ KO mice contained a significantly lower number of NK/T cells than B6 mice (p < 0.0019). 2B4+ NK cells were also significantly reduced (p = 0.0005), and CD8+/NK cells were virtually absent (p = 0.0046). CD8 KO mice lack CD8+/NK cells (Fig. 5⇓) as well as CD8+ T cells, as expected (data not shown). It could be hypothesized that the NK/T subfamily present in CD8 KO mice but lacking in TCR-β/TCR-δ KO mice may represent the CyP-sensitive cell, and may explain why CyP is not required to achieve engraftment in TCR-β/TCR-δ KO mice.

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

NK subset expression in bone marrow and spleen of KO mice. NK subset expression was enumerated for bone marrow and spleen from TCR-β KO, TCR-δ, TCR-β/TCR-δ KO, CD8 KO mice, and the B6 control mice using four-color cytometry. NK1.1 (□), 5E6 (▦), NK/T (NK1.1 and TCR positive; ▨), CD8/NK1.1 (▪), and 2B4 (▤). Each bar represents the mean of three mice and their SDs.

Discussion

Mixed hematopoietic chimerism induces tolerance to solid organ and cellular allografts (8). Strategies to establish chimerism with partial myeloablation have the advantage that the mortality associated with ablative conditioning can be avoided. Conditioning of the recipient with a combination of cytoreductive plus immunosuppressive agents is required to achieve engraftment of MHC-disparate marrow (21, 30). Historically, cytoreduction has used a combination of nonspecific agents, such as irradiation and busulfan, which have a broad specificity and poorly defined mechanism of action. If those host components which regulate engraftment could be defined, more specific approaches to target only those host cells responsible for alloresistance to engraftment would be possible. Theoretically, chimerism could be achieved with minimal regimen-related morbidity.

Until recently, it was believed that full ablation of the host hematopoietic microenvironment was a prerequisite to achieve engraftment of allogeneic marrow. The discovery that multiple hematopoietic systems can coexist and function in concert in the form of mixed hematopoietic chimerism (8, 31) led to the recognition that partially ablative conditioning may be sufficient. The observation that engraftment of physiologic numbers of sygeneic bone marrow cells required low-dose conditioning (32), and that substantially more TBI was required for MHC-matched but minor Ag-disparate (32, 12), and MHC-disparate marrow (12) led to the hypothesis that more than one host mechanism is probably operational in engraftment of allogeneic marrow (12). Down et al. (32) hypothesized that the differences in radiation dose for syngeneic vs allogeneic engraftment may represent removal of specific host cell populations with differing radiosensitivities.

A number of studies have demonstrated that engraftment can be achieved using partial conditioning (22, 33, 34). To date, most protocols have used nonspecific cytoreductive immunosuppressive and/or mAb treatment. Two stages occur in the early events in engraftment: host factors that control whether the donor marrow can engraft (alloresistance), and alloreactive host cells that can reject grafts that initially engrafted. One of three outcomes can occur: 1) the graft may never take; 2) the graft may take and then be rejected or be only transient due to failure of survival of self-renewing HSC; and 3) durable engraftment may occur. We and others have shown that CD8+ cells play an important role in host alloresistance to engraftment (22, 35, 36). Conditioning of recipients with a combination of both anti-CD4 and anti-CD8 depleting and nondepleting mAb allows engraftment of MHC-congenic marrow (35). The addition of 600–850 cGy of TBI (13) or 300 cGy of TBI plus 700 cGy of thymic irradiation (20) is required if the donor and recipient are MHC-disparate. It was concluded from those studies that conventional thymic-derived αβ-TCR+ T cells were the primary effector cells being removed by this approach (20).

In our own model of partial conditioning to achieve stable mixed chimerism, the TBI dose could be reduced from 700 to 300 cGy TBI if ALG (day −3) plus a single dose of CyP were administrated on day +2 (12, 21). If the CyP was administrated before the TBI and BMT, engraftment did not occur. If ALG was given without CyP, engraftment occurred but was only transient. Moreover, if the ALG was replaced by in vivo pretreatment of the recipient with anti-CD8 mAb, a higher level of chimerism was consistently achieved. Anti-CD4 mAb pretreatment of the recipient did not replace the ALG. One caveat to preconditioning of normal recipients with mAb is that all putative effector cells may not be coated or depleted. The presence of immunocompetent residual CD8+ cells after mAb treatment has been demonstrated. Rosenberg et al. (37) demonstrated that CD8+ cells are the main effector cells in the rejection of MHC class I disparate skin grafts. Ichikawa et al. (38) reported that skin grafts with MHC class I disparities are rejected in CD8-depleted recipients. Rosenberg et al. (39) characterized the cell populations that mediated this rejection, and found it to be mediated by a small number of residual CD8+ cells, despite the fact that >99% of the CD8+ cells had been removed. To avoid the variable of residual effector cells in our studies, we used mice that were genetically deficient in the production of these cells. Thus, no residual cells are present in these animals.

T cells have been implicated as the primary effector cells in solid organ allograft rejection. Targeting αβ-TCR+ T cells significantly prolongs survival of skin grafts (40). Although the same effect could be achieved by targeting CD3+ T cells, animals prepared by depletion of αβ-TCR+ cells demonstrated superior immunocompetence (40). Similarly, TCR-β KO mice do not reject MHC-disparate cardiac allografts (41). BMT from normal B10.BR donors restores the immunocompetence to reject third-party cardiac allografts in TCR-β KO mice (41). In the present studies, we have demonstrated that TCR-β KO mice do not accept allogeneic bone marrow without conditioning.

The role of T cells in rejection of bone marrow grafts has not been fully defined. When Kernan et al. (42) characterized the cells present in recipients of HLA-mismatched bone marrow grafts at the time of rejection, they found that graft failure was associated with the emergence of donor-reactive T cells. Other groups report that CD2+, CD3+, and CD8+ T cells of recipient origin in the peripheral blood of bone marrow recipients effectively inhibit the proliferation and differentiation of donor bone marrow cells in vitro (43). CyP has been shown to deplete activated T cells (17, 21, 44). Therefore, it is likely that the engraftment-enhancing effect of CyP on day +2 is due to the effect on activated donor-reactive cells. The results of the experiments described here confirm a critical role for host αβ-TCR+ as well as γδ-TCR+ T cells in the resistance to engraftment of allogeneic bone marrow. Animals deficient in the production of both αβ- and γδ-TCR+ T cells are significantly enhanced in their ability to accept allogeneic bone marrow grafts compared with wild-type controls. If both host αβ- and γδ-TCR+ T cells are lacking, allogeneic marrow is accepted with little or no conditioning. Mice lacking αβ-TCR+ cells alone engraft only if CyP is administered 2 days after BMT after conditioning with 300 cGy TBI, suggesting that γδ-T cells also contribute in a significant fashion to alloresistance. Mice lacking γδ-TCR+ T cells exhibit enhanced engraftment, although to a lesser extent than those lacking αβ-TCR+ cells, as evidenced by the fact that only 56% engrafted after conditioning with 300 cGy TBI plus CyP. Taken together, these data confirm that host αβ-TCR+ T cells as well as γδ-TCR+ T cells exert a critical influence on engraftment of allogeneic marrow. This finding is supported by the fact that only mice deficient in production of αβ- plus γδ-TCR+ T cells (TCR-β/TCR-δ KO) reliably engraft with a low TBI dose alone or even no conditioning at all, confirming that both αβ- and γδ-TCR+ cells in the host contribute in a nonredundant and critical fashion in alloresistance to engraftment. These data also refute a role for conventional NK cells as the primary effector cells in allogeneic marrow graft rejection.

In the present studies, we defined a critical role for host CD8+ cells in regulating engraftment. Durable, multilineage engraftment occurred in all CD8 KO mice conditioned with CyP plus low dose TBI, and in 50% of CD8 KO mice conditioned with CyP alone. The level of chimerism was directly correlated with the dose of TBI. In striking contrast, none of the CD4 KO mice conditioned with as high as 300 cGy TBI plus CyP engrafted when transplanted in a similar fashion. These results demonstrate that TBI-sensitive CD8+ cells in the wild-type recipient hematopoietic microenvironment play a critical role in marrow rejection. Because CD8 KO mice produce γδ-TCR+ T cells and CD4+ αβ-TCR+ T cells, it could be hypothesized that in addition to γδ-TCR+ T cells a separate CD8+ cell in normal recipients contributes to alloresistance. The fact that TCR-δ KO mice required TBI plus CyP to engraft while CD8 KO mice engraft with CyP conditioning alone supports this hypothesis. Moreover, a CD8− cell in the CD8 KO recipient that is CyP-sensitive also contributes to alloresistance, because TCR-β/TCR-δ KO mice engraft with no conditioning while TCR-β KO mice require TBI plus CyP. However, the fact that NK/T cells are present in the marrow of CD8 KO mice, although not present in TCR-β/TCR-δ KO mice, could support a role for a subfamily of NK cells contributing to alloresistance as well. If the sole effector cell were CD8+/TCR+ T cells, it would be expected for the CD8 KO mice to have similar conditioning requirements to the TCR-β KO mice.

NK cells have been implicated as playing a major role in marrow rejection (24, 45). Several subfamilies of NK cells have been described, including 5E6, 2B4, and DX5. The 5E6+ NK cells comprise 50% of NK cells and have been demonstrated to influence engraftment and hematopoiesis (46, 47). The fact that the NK1.1+ and 5E6 NK subsets are present in TCR-δ KO, TCR-β KO, TCR-β/TCR-δ, and CD8 KO mice at levels similar to that for B6 control mice suggests that these subfamilies probably do not play a major role in graft rejection. The fact that mice which lack αβ- and/or γδ-TCR+ T cells engraft with less conditioning strongly supports a critical role for conventional T cells rather than NK cells in alloresistance. Moreover, the fact that TCR-β KO mice have no NK/T cells and still require conditioning makes it unlikely that NK/T cells contribute significantly to alloresistance to engraftment.

It is of note that CD4 KO mice did not engraft when conditioned with 300 cGy TBI plus CyP, although CD8 KO, TCR-β KO, and TCR-δ KO mice did. The classic pathway to initiate cytotoxicity mediated by CD8+ T cells requires the help of CD4+ cells (48). However, pathways of CD4+ cell-independent initiation of cytotoxicity have been described. Purified CD8+ cells can mount cytolytic responses without CD4 mediated help in vitro (49, 50) and in vivo (51). Another CD4-independent CD8-mediated mechanism of cytotoxicity is an NK-like mechanism of alloreactivity (52). A number of groups have described an overlap between T cells and NK cells. Dennert et al. (53) have suggested that CD3+ NK1.1+ cells can develop into CD8+ cytotoxic T cells during acute rejection of allogeneic bone marrow grafts. Although T cell-mediated cytotoxicity usually requires activation and takes ∼7–8 days to generate a cytotoxic response (54), rejection via NK cells occurs within 4–5 days (54). However, the early events of alloreactivity for T cell activation take only hours after exposure to Ag (55, 56). Our data demonstrate a critical role for a CD4-independent CD8-mediated mechanism that mediates resistance to engraftment in recipients of allogeneic bone marrow. Although this could be due to T cells or NK/T cells, the fact that αβ-TCR+ T cells play a significant role in alloresistance to engraftment and that TCR-β/TCR-δ KO mice produce NK cells strongly supports a role for conventional T cells.

CyP suppresses cell-mediated immunity and induces quantitative and qualitative changes in the lymphocyte repertoire (57). The administration of CyP results in leukopenia by depletion of mononuclear cell populations. At the same time, CyP can mediate a marked decrease in the cellular cytotoxic function of the remaining cells (57). We previously confirmed that the administration of CyP in the preparative regimen enhances allogeneic engraftment if it is administered 2 days after low dose TBI and bone marrow infusion. A similar effect does not occur if the CyP is administered before marrow infusion in this model (21). It was hypothesized that the mechanism for this effect involves elimination of alloreactive T cells from the recipient during the early stages of priming. In the present studies, the fact that mice lacking αβ- and γδ-TCR T cells engraft with very low doses of TBI alone suggests that these cell types are two major targets removed by CyP in wild-type recipients.

The level of engraftment in TCR-β/TCR-δ double KO mice was higher in animals conditioned with 300 cGy TBI alone as compared with animals conditioned with 300 cGy TBI and CyP. We hypothesize that this is due to the fact that proliferation of donor reactive T cell clones is triggered within 2 days after BMT, rendering these cells an optimal target for CyP. At the same time, recipient-reactive T cell clones of donor origin that are proliferating against host alloantigens will be depleted by CyP as well. Thus, it would be expected for the level of engraftment to be higher in TCR-β/TCR-δ double KO mice when CyP is not administered. Recipient-reactive donor T cells will not be depleted in animals conditioned without CyP. This could theoretically increase the level of donor chimerism, because T cells enhance engraftment through graft-vs-host reactivity. However, when αβ- and γδ-TCR+ T cells are present in the host, CyP is essential to neutralize their alloreactivity. Alternatively, there may be a CyP-sensitive mechanism which inhibits a residual rejection mechanism in αβ- and γδ-T cells.

The fact that CD8 KO mice do not require TBI for conditioning suggests that a TBI-sensitive population of CD8+ host marrow cells also contributes to alloresistance. The target of the TBI could be CD8+ T cells, or alternatively, a different cell type. TBI does not completely eliminate conventional T cells from the recipient microenvironment, even at high doses. Davenport et al. (58) described that CD8+ T cells of host origin capable of rejecting MHC mismatched donor bone marrow persist even in ablated mice (950 cGy). The importance of this phenomenon has been shown clinically in patients with graft rejection where donor-reactive T cell clones present before conditioning re-emerged after BMT (59). In the present studies, we observed that CyP is required to prevent rejection of allogeneic bone marrow grafts in mice conditioned with 300 cGy TBI, unless the animals lack both αβ- and γδ-TCR+ T cells, in which case the requirement for CyP but not TBI is eliminated to achieve significant levels of engraftment. These data therefore confirm that donor-reactive T cells in the recipient hematopoietic environment are not completely removed by low dose irradiation.

A low dose of TBI is required for conditioning if physiologic numbers of syngeneic bone marrow cells are administrated (32). It is important to note that CD8 KO mice engraft without TBI, but do not engraft if CyP is omitted from the conditioning. The level of engraftment is proportional to the irradiation dose and in this way resembles the characteristics of syngeneic engraftment. Therefore, our data confirm that host CD8+ cells as well as αβ-T cells and γδ-T cells each play a mechanistically different role in engraftment of MHC-disparate marrow.

Our data suggest that more than one cell type mediate the rejection of fully MHC and minor Ag disparate bone marrow grafts, showing a critical role for recipient αβ-TCR+ and γδ-TCR+ T cells, but also implicating an additional population CD8+ cells in the resistance to allogeneic bone marrow grafts. It is likely that the different cell types mediate rejection by different mechanisms. Interestingly, CD8+ host cells reject allogeneic bone marrow in the complete absence of CD4+ cells, suggesting a CD4 independent mechanism. Targeting αβ- and γδ-TCR+ T cells, as well as CD8+ cells in the recipient may allow a specific approach to the development of cell-specific conditioning strategies to establish mixed chimerism with less toxicity. If so, mixed chimerism could be more readily applied for tolerance induction, in gene therapy and treatment of nonmalignant diseases, autoimmune diseases, and hematological disorders, such as sickle cell disease and thalassemia.

Acknowledgments

We thank H. Leighton Grimes, Geoffrey P. Herzig, Thomas C. Mitchell, and Paula M. Chilton for their careful review and helpful advice during the manuscript preparation, Mukunda B. Ray for the histopathologic analyses, Theresa Perry for technical support, Carolyn DeLautre for her superb administrative assistance in preparation of the manuscript, and the outstanding animal care team at the University of Louisville.

Footnotes

  • ↵1 This work was supported in part by grants from the National Institutes of Health, DK 43901-09 (to S.T.I.), the Deutsche Forschungsgemeinschaft, EX 11/1-1 (to B.G.E.), the Jewish Hospital Foundation, and the Commonwealth of Kentucky Research Challenge Trust Fund.

  • ↵2 H.X. and B.G.E. contributed equally in this study.

  • ↵3 Address correspondence and reprint requests to: Dr. Suzanne T. Ildstad, University of Louisville, 570 South Preston Street, Suite 404, Louisville, KY 40202-1760. E-mail address: stilds01{at}gwise.louisville.edu

  • ↵4 Abbreviations used in this paper: GVHD, graft-vs-host disease; TBI, total body irradiation; CyP, cyclophosphamide; ALG, antilymphocyte globuln; KO, knockout; BMT, bone marrow transplantation; NOD, nonobese diabetic; HSC, hematopoietic stem cell; CM, chimeria media.

  • Received July 13, 2001.
  • Accepted November 28, 2001.
  • Copyright © 2002 by The American Association of Immunologists

References

  1. ↵
    Farges, O., F. Saliba, H. Farhamant, D. Samuel, A. Bismuth, M. Reynes, H. Bismuth. 1996. Incidence of rejection and infection after liver transplantation as a function of the primary disease: possible influence of alcohol and polyclonal immunoglobulins. Hepatology 23: 240
    OpenUrlCrossRefPubMed
  2. ↵
    Penn, I.. 1994. Malignancy. Surg. Clin. North Am. 74: 1247
    OpenUrlPubMed
  3. ↵
    Platz, K. P., A. R. Mueller, G. Blumhardt, S. Bachmann, W. O. Bechstein, A. Kahl, P. Neuhaus. 1994. Nephrotoxicity after orthotopic liver transplantation in cyclosporin A and FK 506-treated patients. Transpl. Int. 7: (Suppl. 1):S52
    OpenUrlCrossRefPubMed
  4. ↵
    Vercauteren, S. B., J. L. Bosmans, M. M. Elseviers, G. A. Verpooten, M. E. De Broe. 1998. A meta-analysis and morphological review of cyclosporine-induced nephrotoxicity in auto-immune diseases. Kidney Int. 54: 536
    OpenUrlCrossRefPubMed
  5. ↵
    Nagano, H., N. L. Tilney. 1997. Chronic allograft failure: the clinical problem. Am. J. Med. Sci. 313: 305
    OpenUrlCrossRefPubMed
  6. ↵
    Terasaki, P. I., J. M. Cecka, D. W. Gjertson, Y. Cho, S. Takemoto, M. Cohn. 1992. A Ten-Year Prediction for Kidney Transplant Survival 501 UCLA Tissue Typing Laboratory, Los Angeles.
  7. ↵
    Opelz, G.. 1999. Effect of immunosuppressive therapy on graft half-life projections: the collaborative transplant study. Transplant Proc. 31: 31S
    OpenUrl
  8. ↵
    Ildstad, S. T., D. H. Sachs. 1984. Reconstitution with syngeneic plus allogeneic or xenogeneic bone marrow leads to specific acceptance of allografts or xenografts. Nature 307: 168
    OpenUrlCrossRefPubMed
  9. ↵
    Van Leeuwen, J. M., M. D. Van Tol, A. M. Joosten, J. T. Wijnen, P. M. Verweij, P. M. Khan, J. M. Vossen. 1994. Persistence of host-type hematopoiesis after allogeneic bone marrow transplantation for leukemia is significantly related to the recipient’s age and/or the conditioning regimen, but it is not associated with an increased risk of relapse. Blood 83: 3059
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Ruedi, E., M. Sykes, S. T. Ildstad, C. H. Chester, A. Althage, H. Hengartner, D. H. Sachs, R. M. Zinkernagel. 1989. Antiviral T cell competence and restriction specificity of mixed allogeneic (P1 + P2→P1) irradiation chimeras. Cell. Immunol. 121: 185
    OpenUrlCrossRefPubMed
  11. ↵
    Ildstad, S. T., S. M. Wren, J. A. Bluestone, S. A. Barbieri, D. H. Sachs. 1985. Characterization of mixed allogeneic chimeras: immunocompetence, in vitro reactivity and genetic specificity of tolerance. J. Exp. Med. 162: 231
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Colson, Y. L., S. M. Wren, M. J. Schuchert, K. D. Patrene, P. C. Johnson, S. S. Boggs, S. T. Ildstad. 1995. A nonlethal conditioning approach to achieve durable multilineage mixed chimerism and tolerance across major, minor, and hematopoietic histocompatibility barriers. J. Immunol. 155: 4179
    OpenUrlAbstract
  13. ↵
    Cobbold, S. P., G. Martin, H. Waldmann. 1986. Monoclonal antibodies for the prevention of graft-versus-host disease and marrow graft rejection: the depletion of T-cell subsets in vitro and in vivo. Transplantation 42: 239
    OpenUrlCrossRefPubMed
  14. ↵
    Gammie, J. S., S. Li, A. J. Demetris, A. Zeevi, S. T. Ildstad, S. M. Pham. 1998. Tacrolimus-based partial conditioning produces stable mixed lymphohematopoietic chimerism and tolerance for cardiac allografts. Circulation 98: II163
    OpenUrlPubMed
  15. ↵
    Wood, M. L., A. P. Monaco, J. J. Gozzo, A. Liegeois. 1971. Use of homozygous allogeneic bone marrow for induction of tolerance with antilymphocyte serum: dose and timing. Transplant. Proc. 3: 676
    OpenUrlPubMed
  16. ↵
    McCarthy, S. A., I. J. Griffith, P. Gambel, L. H. Francescutti, T. G. Wegman. 1985. Characterization of host lymphoid cells in antibody-facilitated bone marrow chimeras. Transplantation 40: 12
    OpenUrlPubMed
  17. ↵
    Mayumi, H., R. A. Good. 1988. The necessity of both allogeneic antigens and stem cells for cyclophosphamide-induced skin allograft tolerance in mice. Transplant. Proc. 20: 139
    OpenUrlPubMed
  18. ↵
    Cobbold, S. P., G. Martin, S. Qin, H. Waldmann. 1986. Monoclonal antibodies to promote marrow engraftment and tissue graft tolerance. Nature 323: 164
    OpenUrlCrossRefPubMed
  19. ↵
    Sykes, M., G. L. Szot, K. A. Swenson, D. A. Pearson. 1997. Induction of high levels of allogeneic hematopoietic reconstitution and donor-specific tolerance without myelosuppressive conditioning. Nat. Med. 3: 783
    OpenUrlCrossRefPubMed
  20. ↵
    Sharabi, Y., D. H. Sachs. 1989. Mixed chimerism and permanent specific transplantation tolerance induced by a nonlethal preparative regimen. J. Exp. Med. 169: 493
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Colson, Y. L., H. Li, S. S. Boggs, K. D. Patrene, P. C. Johnson, S. T. Ildstad. 1996. Durable mixed allogeneic chimerism and tolerance by a nonlethal radiation-based cytoreductive approach. J. Immunol. 157: 2820
    OpenUrlAbstract
  22. ↵
    Exner, B. G., Y. L. Colson, H. Li, S. T. Ildstad. 1997. In vivo depletion of host CD4+ and CD8+ cells permits engraftment of bone marrow stem cells and tolerance induction with minimal conditioning. Surgery 122: 221
    OpenUrlCrossRefPubMed
  23. ↵
    Li, H., C. L. Kaufman, S. S. Boggs, P. C. Johnson, K. D. Patrene, S. T. Ildstad. 1996. Mixed allogeneic chimerism induced by a sublethal approach prevents autoimmune diabetes and reverses insulitis in non-obese diabetic (NOD) mice. J. Immunol. 156: 380
    OpenUrlAbstract
  24. ↵
    Cudkowicz, G., M. Bennett. 1971. Peculiar immunobiology of bone marrow allografts. I. Graft rejection by irradiated responder mice. J. Exp. Med. 134: 83
    OpenUrlAbstract
  25. ↵
    Billingham, R. E.. 1961. Free Skin Grafting in Mammals 1 The Wistar Institute Press, Philadephia.
  26. ↵
    Garni-Wagner, B. A., A. Purohit, P. A. Mathew, M. Bennett, V. Kumar. 1993. A novel function-associated molecule related to non-MHC-restricted cytotoxicity mediated by activated natural killer cells and T cells. J. Immunol. 151: 60
    OpenUrlAbstract
  27. ↵
    Sykes, M.. 1990. Unusual T cell populations in adult murine bone marrow: prevalence of CD3+CD4−CD8− and αβ TCR+NK1.1+ cells. J. Immunol. 145: 3209
    OpenUrlAbstract
  28. ↵
    Kaufman, C. L., W. H. Chambers, S. T. Ildstad. 1993. Coexpression of NKR-P1 and αβ-TCR on lymphoid cells in fully xenogeneic (ratrarrow]mouse) chimeras and syngeneically reconstituted (A→A) rats. J. Immunol. 151: 6002
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Kaufman, C. L., Y. L. Colson, S. M. Wren, S. L. Watkins, R. L. Simmons, S. T. Ildstad. 1994. Phenotypic characterization of a novel bone-marrow derived cell that facilitates engraftment of allogeneic bone marrow stem cells. Blood 84: 2436
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Gammie, J. S., S. Li, Y. L. Colson, A. J. Demetris, M. Neipp, S. T. Ildstad, S. M. Pham. 1998. A partial conditioning strategy for achieving mixed chimerism in the rat: tacrolimus and anti-lymphocyte serum substantially reduce the minimum radiation dose for engraftment. Exp. Hematol. 26: 927
    OpenUrlPubMed
  31. ↵
    Slavin, S., S. Strober, Z. Fukes, H. S. Kaplan. 1977. Induction of specific tissue transplantation tolerance using fractionated total lymphoid irradiation in adult mice: long-term survival of allogeneic bone marrow and skin grafts. J. Exp. Med. 146: 34
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Down, J. D., N. J. Tarbell, H. D. Thames, P. M. Mauch. 1991. Syngeneic and allogeneic bone marrow engraftment after total body irradiation: dependence on dose, dose rate, and fractionation. Blood 77: 661
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Jankowski, R. A., S. T. Ildstad. 1997. Chimerism and tolerance: from freemartin cattle and neonatal mice to humans. Hum. Immunol. 52: 155
    OpenUrlCrossRefPubMed
  34. ↵
    Sykes, M., F. Preffer, S. McAfee, S. L. Saidman, D. Weymouth, D. M. Andrews, C. Colby, R. Sackstein, D. H. Sachs, T. R. Spitzer. 1999. Mixed lymphohaemopoietic chimerism and graft-versus-lymphoma effects after non-myeloablative therapy and HLA-mismatched bone-marrow transplantation. Lancet 353: 1755
    OpenUrlCrossRefPubMed
  35. ↵
    Cobbold, S. P., A. Jayasuriya, A. Nash, T. D. Prospero, H. Waldman. 1984. Therapy with monoclonal antibodies by elimination of T-cell subsets in vivo. Nature 312: 548
    OpenUrlCrossRefPubMed
  36. ↵
    Sykes, M., K. A. Hoyles, M. L. Romick, D. H. Sachs. 1990. In vivo and in vitro analysis of bone marrow-derived CD3+, CD4−, CD8−, NK1.1+ cell lines. Cell. Immunol. 129: 478
    OpenUrlCrossRefPubMed
  37. ↵
    Rosenberg, A. S., T. Mizuochi, A. Singer. 1986. Analysis of T-cell subsets in rejection of κβ mutant skin allografts differing at class I MHC. Nature 322: 829
    OpenUrlCrossRefPubMed
  38. ↵
    Ichikawa, T., E. Nakayama, A. Uenaka, M. Monden, J. Okamura, T. Mori. 1987. Effect of Lyt-2 mAb on murine skin graft survivals. Transplant. Proc. 19: 579
    OpenUrlPubMed
  39. ↵
    Rosenberg, A. S., T. I. Munitz, T. G. Maniero, A. Singer. 1991. Cellular basis of skin allograft rejection across a class I major histocompatibility barrier in mice depleted of CD8+ T cells in vivo. J. Exp. Med. 173: 1463
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Eto, M., Y. Yoshikai, Y. Nishimura, K. Hiromatsu, T. Maeda, K. Nomoto, Y. Y. Kong, R. T. Kubo, J. Kumazawa, K. Nomoto. 1994. Inhibition of allograft rejection by anti-T-cell receptor-αβ monoclonal antibodies preserving resistance to bacterial infection. Immunology 81: 198
    OpenUrlPubMed
  41. ↵
    Exner, B. G., X. Que, Y. M. Mueller, M. A. Domenick, M. Neipp, S. T. Ildstad. 1999. αβ TCR+ T cells play a nonredundant role in the rejection of heart allografts in mice. Surgery 126: 121
    OpenUrlPubMed
  42. ↵
    Kernan, N. A., N. Flomenberg, B. Dupont, R. J. O’Reilly. 1987. Graft rejection in recipients of T-cell-depleted HLA-nonidentical marrow transplants for leukemia: identification of host-derived antidonor allocytotoxic T lymphocytes. Transplantation 43: 842
    OpenUrlCrossRefPubMed
  43. ↵
    Bierer, B. E., S. G. Emerson, J. Antin, R. Maziarz, J. M. Rappeport, B. R. Smith, S. J. Burakoff. 1988. Regulation of cytotoxic T lymphocyte-mediated graft rejection following bone marrow transplantation. Transplantation 46: 835
    OpenUrlPubMed
  44. ↵
    Tripp, R. A., S. Hou, A. McMickle, J. Houston, P. C. Doherty. 1995. Recruitment and proliferation of CD8+ T cells in respiratory virus infections. J. Immunol. 154: 6013
    OpenUrlAbstract
  45. ↵
    Bennett, M.. 1987. Biology and genetics of hybrid resistance. Adv. Immunol. 41: 333
    OpenUrlCrossRefPubMed
  46. ↵
    Sentman, C. L., V. Kumar, M. Bennett. 1991. Rejection of bone marrow cell allografts by natural killer cell subsets: 5E6+ cell specificity for Hh-1 determinant 2 shared by H-2d and H-2f. Eur. J. Immunol. 21: 2821
    OpenUrlCrossRefPubMed
  47. ↵
    Sentman, C. L., J. J. Hackett, V. Kumar, M. Bennett. 1989. Identification of a subset of murine natural killer cells that mediates rejection of Hh-Id but not Hh-1b bone marrow grafts. J. Exp. Med. 170: 191
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Cantor, H., E. A. Boyse. 1975. Functional subclasses of T lymphocytes bearing different Ly antigens. II. Cooperation between subclasses of Ly+ cells in the generation of killer activity. J. Exp. Med. 141: 1390
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Singer, A., A. M. Kruisbeek, P. M. Andrysiak. 1984. T cell-accessory cell interactions that initiate allospecific cytotoxic T lymphocyte responses: existence of both Ia-restricted and Ia-unrestricted cellular interaction pathways. J. Immunol. 132: 2199
    OpenUrlAbstract/FREE Full Text
  50. ↵
    Sprent, J., M. Schaefer. 1985. Properties of purified T cell subsets. I. In vitro responses to class I vs. class II H-2 alloantigens. J. Exp. Med. 162: 2068
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Sprent, J., M. Schaefer, D. Lo, R. Korngold. 1986. Properties of purified T cell subsets. II. In vivo responses to class I vs. class II H-2 differences. J. Exp. Med. 163: 998
    OpenUrlAbstract/FREE Full Text
  52. ↵
    Davenport, C., A. Haile, V. Kumar, M. Bennett. 1995. Hybrid and allogeneic resistance to T cell grafts mediated by murine NK and CD8+ T cells. J. Immunol. 154: 2568
    OpenUrlAbstract
  53. ↵
    Dennert, G., C. Knobloch, S. Sugawara, B. Yankelevich. 1990. Evidence for differentiation of NK1+ cells into cytotoxic T cells during acute rejection of allogeneic bone marrow grafts. Immunogenetics 31: 161
    OpenUrlPubMed
  54. ↵
    Murphy, W. J., V. Kumar, M. Bennett. 1987. Acute rejection of murine bone marrow allografts by natural killer cells and T cells: differences in kinetics and target antigen recognized. J. Exp. Med. 166: 1499
    OpenUrlAbstract/FREE Full Text
  55. ↵
    Cebrian, M., E. Yague, M. Rincon, M. Lopez-Botet, M. O. De Landazuri, F. Sanchez-Madrid. 1988. Triggering of T cell proliferation through AIM, an activation inducer molecule expressed on activated human lymphocytes. J. Exp. Med. 168: 1637
    OpenUrl
  56. ↵
    Testi, R., J. H. Phillps, L. L. Lanier. 1989. Leu23 induction as an early marker of functional CD3/T cell antigen receptor triggering: requirement for receptor cross-linking, prolonged elevation of intracellular calcium and stimulation of protein kinase. J. Immunol. 142: 1860
    OpenUrl
  57. ↵
    Hunninghake, G. W., A. S. Fauci. 1976. Quantitative and qualitative effects of cyclophosphamide administration on circulating polymorphonuclear leucocytes. Immunology 31: 139
    OpenUrlPubMed
  58. ↵
    Davenport, C., V. Kumar, M. Bennett. 1995. Rapid Rejection of H2k and H2k/b bone marrow cell grafts by CD8+ T cells and NK cells in irradiated mice. J. Immunol. 155: 3742
    OpenUrlAbstract
  59. ↵
    Voogt, P. J., W. E. Fibbe, W. A. Marijt, E. Goulmy, W. F. Veenhof, M. Hamilton, A. Brand, F. E. Zwann, R. Willemze, J. J. van Rood. 1990. Rejection of bone-marrow graft by recipient-derived cytotoxic T lymphocytes against minor histocompatibility antigens. Lancet 335: 131
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top

In this issue

The Journal of Immunology: 168 (4)
The Journal of Immunology
Vol. 168, Issue 4
15 Feb 2002
  • Table of Contents
  • About the Cover
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
CD8+, αβ-TCR+, and γδ-TCR+ Cells in the Recipient Hematopoietic Environment Mediate Resistance to Engraftment of Allogeneic Donor Bone Marrow
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
Citation Tools
CD8+, αβ-TCR+, and γδ-TCR+ Cells in the Recipient Hematopoietic Environment Mediate Resistance to Engraftment of Allogeneic Donor Bone Marrow
Hong Xu, Beate G. Exner, Daniel E. Cramer, Michael K. Tanner, Yvonne M. Mueller, Suzanne T. Ildstad
The Journal of Immunology February 15, 2002, 168 (4) 1636-1643; DOI: 10.4049/jimmunol.168.4.1636

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
CD8+, αβ-TCR+, and γδ-TCR+ Cells in the Recipient Hematopoietic Environment Mediate Resistance to Engraftment of Allogeneic Donor Bone Marrow
Hong Xu, Beate G. Exner, Daniel E. Cramer, Michael K. Tanner, Yvonne M. Mueller, Suzanne T. Ildstad
The Journal of Immunology February 15, 2002, 168 (4) 1636-1643; DOI: 10.4049/jimmunol.168.4.1636
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2019 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606