There is growing interest in using human umbilical cord blood (CB) for allogeneic bone marrow transplantation (BMT), particularly in children. Thus, CB has been identified as a rich source of hematopoietic progenitors of the erythroid, myeloid, and B-cell lineages. Whether CB blood cells engrafting in the BM space also comprise T-cell progenitors capable of trafficking to the thymus and reconstituting a functional thymopoiesis in young recipients is presently unknown. Here, we show that CB progenitors, engrafted in the BM of immunodeficient mice, sustain human thymopoiesis by generating circulating T-cell progenitors capable of homing to and developing within a human thymic graft. Surprisingly, development of CB stem cells in this in vivo model extended to elements of the endothelial cell lineage, which contributed to the revascularization of transplants and wound healing. These results demonstrate that human CB stem cell transplantation can reconstitute thymic-dependent T-cell lymphopoiesis and show a novel role of CB-derived hematopoietic stem cells in angiogenesis.

TRANSPLANTATION of hematopoietic stem cells is currently the treatment for reconstitution of the immune system in immunodeficiency diseases1,2 or after myeloablative chemotherapy.3,4 Clinical experience5,6 and animal models7-11 indicate that both the proliferative and differentiative capacity of the transfused stem cells are critical for restoration of host immune competence, including a functional mature B- and T-cell repertoire.

Human umbilical cord blood (CB) has been recently identified as a rich source of multipotent hematopoietic progenitors.12-15 As compared with adult bone marrow (BM), CB progenitors have been shown to have a higher proliferative and self-renewal potential,16-19 suggesting a higher capacity for reconstitution of hematopoiesis. Colony-forming unit (CFU) assays demonstrated that CB hematopoietic stem cells differentiate along granulocyte, erythrocyte, monocyte, and megakaryocyte lineages.20-23 Transfusion of human CB in immunodeficient severe combined immunodeficiency (SCID) mice demonstrated repopulation of the BM with clonogenic progenitors, which support development of erythroid, myeloid, and B-cell lineages.24-26 Although CB is a mixture of primitive and more committed hematopoietic progenitors, only the most primitive CD34+CD38 cell population allows for long-term multilineage engraftment in nonobese diabetic (NOD)/SCID mice.27 Whether CD34+CD38 CB stem cells can also reconstitute T-cell lymphopoiesis in vivo is presently unknown. Understanding the T-cell developmental potential of CB progenitors is important to determine whether transplanted patients will recover T-cell–dependent primary antibody responses and immunity against environmental pathogens.

Human CB CD34+ cells injected into transplanted fetal thymus or fetal thymic organ culture can give rise to mature thymocytes.28-30 Although these studies suggest the presence of T-cell progenitors, they do not establish whether such progenitors exist in the CB as terminally committed cells or more primitive elements. Moreover, these studies cannot address the question of whether T-cell progenitors traffic from the BM to the thymic compartment. To address these issues, we developed an in vivo model of continuous human T-cell lymphopoiesis. In this model, the BM compartment of NOD/SCID mice is reconstituted with T- and B-cell–depleted human CB, followed by transplantation of human fetal thymus under the kidney capsule. Implicit to this model is the requirement of primitive human CB progenitors to engraft in the BM compartment and sustain T-cell lymphopoiesis by regulated homing of their progeny to the thymus. Furthermore, a functional vascular connection between the mouse tissue and the human thymic graft must develop to mediate progenitor homing and the exit of mature T cells to the periphery. Therefore, we extended our studies to investigate the development of this critical vascular interface at the site of thymic transplants.

Here we report that CB-derived hematopoietic stem cells, engrafted in the BM of NOD/SCID mice, generated T-cell progenitors capable of homing to and developing within a human thymic graft transplanted at a distant site. Human thymopoiesis led to the exit of mature T cells to peripheral lymphoid compartments. These results demonstrate that human CB stem cell transplantation supports thymic T-cell development. Surprisingly, BM reconstitution with CB also led to engraftment of CB-derived progenitors of the endothelial lineage, which were recruited to form new blood vessels extending from the thymic implant into the surrounding mouse tissue. This finding shows that human CB may also represent a unique reservoir of endothelial progenitors and suggests a novel mechanism of neovascularization involving the participation of circulating endothelial stem cells.

Animals.

NOD/LtSz-SCID mice (NOD/SCID) were obtained from our colony (original founders a kind gift of Dr L. Shultz, Bar Harbor Laboratories, Bar Harbor, ME). The colony was derived by cesarean section under specific pathogen-free conditions. Mice were maintained on an irradiated sterile diet and autoclaved acidified water.

Human CB and thymic tissue.

Sterile human CB was obtained from normal deliveries at the Mary Birch Hospital for Women (San Diego, CA). Samples were processed within 24 hours. Mononuclear cells were first separated on a Ficoll gradient. CB cells were depleted of mature T and B cells by negative selection using an immunomagnetic separation technique. Cells were with anti-CD3 monoclonal antibody (MoAb) (OKT3; American Type Culture Collection [ATCC], Bethesda, MD) and anti-CD19 MoAb (HD37, Dako, Carpinteria, CA) for 30 minutes at 4°C followed sequentially by a biotin-conjugated goat anti-mouse IgG (Caltag, Burlingame, CA), fluorescein isothiocyanate (FITC)-conjugated streptavidin (GIBCO, Gaithersburg, MD) and biotin-magnetic beads (Miltenyi Biotec, Sunnyvale, CA). The cells were applied to a VarioMacs magnetic column (Miltenyi Biotec, a kind gift of Dr Kevin Mills) and nonadherent cells collected. This method consistently yielded a cell purity > 95% as determined by double-color immunostaining using an R-phycoerythrin (RPE)-conjugated anti-human T-cell receptor (TCR)α/β (BMA031; Immunotech, Westbrook, ME) and an FITC-conjugated anti-CD20 MoAbs (B-Ly1; Dako). Cell preparations contained less than 5% CD20-positive contaminants, but no detectable TCRα/β-positive cells. The proportion of CD34+ progenitors in each CB sample was finally assessed with an FITC-conjugated anti-CD45 MoAb (Becton Dickinson, Bedford, MA) and a biotin-conjugated anti-CD34 MoAb (QBEnd10, Immunotech) followed by Cy-Chrome-streptavidin (Pharmingen, San Diego, CA). CD34+ cells represented 2% to 5% of total CB cells after T- and B-cell depletion.

Fetal thymic tissue (18 to 24 weeks of gestation) was obtained from ABR (Alameda, CA). To deplete the tissue of endogenous thymocytes, the thymi were minced in ≈1 mm × 1 mm pieces and cultured in Iscove medium containing 10% fetal calf serum (FCS), penicillin/streptomycin, and 1.35 mmol/L deoxyguanosine (dGuo) (Sigma, St Louis, MO) for 5 to 6 days.

Generation of human BM and thymus-NOD/SCID chimeras.

NOD/SCID mice (6 to 8 weeks old) were sublethally irradiated (300 cGy) and injected in the tail vein with T- and B-cell–depleted CB cells. The number of injected cells was adjusted to deliver 100 to 200 × 103 CD34+ progenitors per mouse. After 4 to 6 weeks, the mice were transplanted under the kidney capsule with the human thymic fragments (2 to 3 pieces/mouse). At 2 and 4 months postthymic transplant, the mice were euthanized. Bone marrow cells were recovered by flushing femurs and tibia with RPMI-10% FCS. The thymic implants and spleens were excised and either embedded for histologic analysis or cells were prepared by straining through a 100-μm stainless steel mesh. Peripheral blood lymphocytes (PBLs) were obtained by cardiac puncture and separated from red blood cells on a Ficoll-Paque gradient (Pharmacia, Uppsala, Sweden).

Flow cytometry and cell sorting.

BM, thymic, splenic, and PBL cells were incubated for 15 minutes at 4°C in the presence of polyclonal mouse IgG (50 μg mL−1) and anti-mouse Fc MoAb (2.4G2; Pharmingen). Lymphocytes were then stained using the following antibodies: FITC anti-mouse H2kd (SF1-1.1, Pharmingen); RPE anti-human HLA-A,B,C (G46-2.6; Pharmingen); an FITC anti-human CD45 MoAb (2D1; Becton Dickinson), an RPE anti-human CD4 MoAb (13B8.2; Immunotech); a Cy-Chrome anti-human CD8 (RPA-T8; Pharmingen), or an FITC anti-human CD8 (B9.11; Immunotech); an RPE anti-human TCR TCRα/β (BMA031; Immunotech) and FITC, RPE, or CyChrome species and isotype matching control antibodies. In preliminary experiments, only minimal cross-reactivity to mouse determinants was observed for the MoAbs, whereas the anti-human CD45 and HLA-A,B,C MoAbs showed no cross-reactivity. Therefore, these antibodies were used to gate for human lymphocytes. The samples were analyzed with a FACScan (Becton Dickinson). In some experiments, single- and double-positive thymocytes were stained with FITC anti-human CD8 and RPE anti-human CD4 MoAbs and purified by cell sorting (FACStar Plus; Becton Dickinson).

Spleen and thymic graft histology.

Ten-micrometer cryostat tissue sections were fixed in phosphate-buffered saline (PBS) 2% paraformaldehyde (PFA) for 10 minutes at room temperature. Sections were permeabilized in PBS/0.1% Triton-X 100 for 5 minutes, blocked first in PBS/50 mmol/L glycine and then in PBS/10% goat or donkey serum for 1 to 2 hours. For immunohistochemistry, sections were treated for 5 minutes with a 0.05% H2O2 solution for saturation of the endogenous peroxidase activity and then probed with a human-specific anti-CD45 MoAb (T29/33; Dako) followed by a biotinylated human-adsorbed goat anti-mouse IgG Fab2 (Caltag) and Peroxidase-streptavidin (GIBCO).

For the detection of human lymphoid and endothelial cells, thymic sections were probed with anti-CD45 MoAb and Lissamine-Rhodamine (LSRC) anti-mouse IgG (Jackson Labs, West Grove, PA) or a combination of a mouse anti-CD34 MoAb (8G12; Becton Dickinson) and a goat anti–PECAM-1 (platelet endothelial adhesion molecule-1) (polyclonal IgG M-20; Santa Cruz Biotechnology, Santa Cruz, CA) followed by an LSRC anti-mouse IgG Fab2 and an FITC anti-goat IgG Fab2 (Jackson Labs). In some experiments, an anti-human–specific von Willebrand Factor (vWF) MoAb (Takara, Shiga, Japan; clone vW1-2) was used. The anti–HLA-A2 antibody (clone MI2.1) was the kind gift of Dr H. Kaneshima (Systemix, Palo Alto, CA). The secondary reagents were preadsorbed to eliminate cross-species reactivity. In some experiments, NOD/SCID mice reconstituted with T- and B-depleted CB cells or human umbilical vein endothelial cells (HUVECs) labeled with the vital dye DiI (Molecular Probes, Eugene, OR) were used as hosts for thymic transplants. In these mice (n = 4), blood vessels developed at the interface with the thymic transplants and comprising endothelial cells of CB origin (ie, DiI+) were quantified by morphometric analysis. For this purpose, a total of 160 optical fields (324.8 μm × 216.5 μm) were scored for the presence of PECAM-1+ vascular profiles containing DiI+ cells.

For simultaneous detection of human CD45+ and apoptotic cells, air-dried thymic sections were fixed in PBS 2% PFA for 10 minutes at room temperature. Sections were incubated with digoxigenin-labeled deoxyuridine triphosphate (dUTP) and terminal deoxynucleotidyl transferase (TdT) enzyme (Oncor, Gaithersburg, MD) for 1 hour at 37°C. After blocking the enzymatic reaction, the sections were incubated with an FITC sheep antidigoxigenin antiserum (Oncor) for 30 minutes at room temperature. The sections were blocked overnight in PBS/10% donkey serum and sequentially probed with the anti-CD45 MoAb and an LSRC donkey anti-mouse IgG preabsorbed with sheep IgG (Jackson Labs). Sections were analyzed on a Zeiss Axiovert microscope with a scanning laser confocal attachment (MRC 1024; BioRad, Hercules, CA).

Polymerase chain reaction (PCR) of Y chromosome–specific DNA.

To identify male BM-derived T cells in the thymic grafts, total or sorted thymocytes were lysed in 10 mmol/L Tris-HCl, 1.5 mmol/L MgCl2, 50 mmol/L KCl, 0.5% Tween 20, 100 μg/mL−1 Proteinase K (Boehringer Mannheim, Indianapolis, IN), pH 8.3, for 45 minutes at 56°C. After inactivation of Proteinase K by heating the samples at 95°C for 10 minutes, an aliquot of each sample (DNA from 10,000 cells) was used for PCR amplification of human Y chromosome sequences. The Y chromosome–specific probes were: forward 5′-TGGGCTGGAATGGAAAGGAATCGAAAC-3′ and reverse 5′-TCCATTCGATTCCATTTTTTTCGAGAA-3′.31 PCR was performed for 25 cycles at 95°C for 1 minute, 65°C for 1 minute, and 72°C for 1 minute, and a 10-minute final extension at 72°C. PCR products were separated on 1.2% agarose gels and visualized by ethidium bromide.

To investigate the presence of male BM-derived stromal cells at the site of thymic engraftment, sections were first stained with an anti-human CD34 MoAb to localize human vessels. Based on this localization, samples were then scraped from consecutive sections using a 30 G needle under microscopic visualization.32 Samples were collected at 3 sites: within the thymic graft, at the interface of the transplant with the mouse kidney, and within the kidney parenchyma distant from the graft. The scraped tissue was placed in 50 μL of lysing buffer, DNA was extracted, and used as template for PCR amplification of human Y chromosome-specific sequences.

Fluorescence in situ hybridization (FISH).

To simultaneously identify cells bearing human Y chromosome and CD4, CD8, CD34, or PECAM-1 in tissue sections, we adapted the protocol described by Gerritsen et al33 for 2-color FISH. Briefly, 10-μm cryostat sections were fixed in acetone for 7 minutes at −20°C. The sections were blocked for 1 hour at room temperature in PBS/10% donkey serum and then probed sequentially with anti-CD4/CD8 MoAbs (OKT4, OKT8), or anti-CD34 MoAb (8G12) followed by LSRC or CY5 donkey anti-mouse IgG absorbed to sheep IgG. In some experiments, sections were probed with anti–PECAM-1 followed by a biotin donkey anti-goat IgG and Cy5 streptavidin. In this case, free binding sites of the secondary reagent were blocked with sheep IgGs (50 μg/mL−1). Sections were fixed for 10 minutes at room temperature in PBS/1% PFA and dehydrated through graded ethanol solutions (70%, 80%, and 100%). Thirty microliters of hybridization mixture (Hybrisol V, Oncor) containing the digoxigenin-labeled Y chromosome-specific DYZ1 and DYZ2 probes (Oncor) was then applied to the sections. The slides were sealed with a glass coverslip, heated to 80°C for 10 minutes, and hybridized for 16 hours in a humidified chamber at 37°C. After washing in 1X sodium citrate buffer (SSC) for 5 minutes at 72°C, the sections were incubated in the presence of FITC sheep antidigoxigenin for 30 minutes at room temperature. Sections were washed 3 times and analyzed by confocal microscopy.

Engraftment of CB-derived BM progenitors sustains thymopoiesis in thymic grafts transplanted at a distant site.

To study the capability of CB-derived BM progenitors to sustain human thymopoiesis in vivo, NOD/SCID chimeras were generated by reconstitution of the murine BM compartment with human CB depleted of mature T and B cells, followed by transplantation of human fetal thymus under the kidney capsule. Control mice received only thymic transplants. We refer to these two groups of animals as CB/Thy-NOD/SCID and Thy-NOD/SCID chimeras, respectively. At 4 and 6 months post-CB reconstitution, the BM and thymocytes from the thymic transplants were harvested and analyzed by flow cytometry for the presence of human cells expressing the leukocyte common antigen CD45.

Figure 1a shows human CD45+cells detected in the BM and thymic grafts of CB/Thy-NOD/SCID chimeras at 4 months post-CB reconstitution. As many as 80% and 95% of the lymphoid cells populating the BM and thymic grafts, respectively, were human. At 6 months, a decrease in the proportion of human cells was observed in the BM compartment (Fig 1b, %CD45+ cells: 7.7 ± 2.8 at 6 months v 51.3 ± 6.5 at 4 months, mean ± standard error of mean [SEM], n = 11). However, overall, the percentage of human CD45+ cells in the thymic transplants remained unchanged (Fig 1b; %CD45+ cells: 55.8 ± 13 at 6 months v 57.3 ± 10.4 at 4 months, mean ± SEM, n = 11). Indeed, by 6 months, the thymic grafts had grown considerably from the original size (eg, ≈2 mm3) at the time of implantation to ≈250 to 500 mm3. Moreover, the comparison of thymic cell numbers at 4 and 6 months (Fig 1c and d) demonstrates an increased number of human cells. In contrast to the significant growth of thymic tissue in CB/Thy-NOD/SCID chimeras, the grafts of control Thy-NOD/SCIDs displayed significantly lower cellularity (CD45+ cells: 3.1 ± 0.8 × 106v 12.1 ± 4.3 × 106; mean ± SEM; n = 11, P = .02).

Fig. 1.

Engraftment of human CD45+ lymphoid cells in the BM compartment and thymic grafts of CB/Thy-NOD/SCID mice. Each bar represents the proportion (a and b) or absolute numbers (c and d) of human CD45+ cells detected by FACS analysis of cell suspensions from the BM (white bars) or thymic grafts (gray bars) of individual mice at 4 months (a and c) and 6 months (b and d) post-CB reconstitution. Data in (a and c) were generated in 4 different experiments and data in (b and d) in 6 different experiments using different CB donors in each experiment.

Fig. 1.

Engraftment of human CD45+ lymphoid cells in the BM compartment and thymic grafts of CB/Thy-NOD/SCID mice. Each bar represents the proportion (a and b) or absolute numbers (c and d) of human CD45+ cells detected by FACS analysis of cell suspensions from the BM (white bars) or thymic grafts (gray bars) of individual mice at 4 months (a and c) and 6 months (b and d) post-CB reconstitution. Data in (a and c) were generated in 4 different experiments and data in (b and d) in 6 different experiments using different CB donors in each experiment.

Close modal

Three-color immunostaining for human CD45, CD4, and CD8 showed that the thymic grafts 4 months postthymic transplant (eg, 6 months post-CB reconstitution) comprised thymocytes at both double- and single-positive stages of maturation (Fig2a through d). Notably, most thymocytes in the grafts of CB/Thy-NOD/SCIDs were human (Fig 2a). Overall, they comprised a significantly higher percentage of CD4 single-positive cells than thymocytes from Thy-NOD/SCIDs (Fig 2b, d, and e; %CD4+: 17.9 ± 3.3 v 6.8 ± 1.6, mean ± SEM, P < .01, n = 11). Conversely, in the absence of human BM, fewer human CD45+ cells were detected in the thymic grafts and the distribution of thymic subsets was abnormally skewed toward a CD8 single-positive phenotype (Fig 2c through e).

Fig. 2.

T-cell development in the thymic grafts of NOD/SCID-hu chimeras. Three-color flow cytometric analysis of thymocytes harvested from thymic grafts of a CB/Thy-NOD/SCID at 6 months post-CB reconstitution (a and b) and a control Thy-NOD/SCID mouse (c and d) stained for human CD45, CD4, and CD8. The staining specific for CD45 (a and c) identifies a population of human CD45+ lymphoid cells (Region 1, R1). Gating on R1 shows that human CD45+ cells from the grafts of CB/Thy-NOD/SCIDs comprise both CD4/CD8 double- and single-positive subsets (b), whereas human CD45+ cells from Thy-NOD/SCID mice harbor mainly CD4 and CD8 single-positive thymocytes (d). The vertical lines within the histograms (a and c) mark the upper limit of the fluorescence intensity corresponding to the isotype-control antibody. In the dot plots (b and d), quadrants were set to comprise the staining given by control antibodies in the left lower quadrant. Histograms and dot plots represent the analysis of 10,000 events. (e) Distribution of human thymic subsets in the grafts of individual CB/Thy-NOD/SCID (▪) and Thy-NOD/SCIDs (◊) at 6 months post-CB reconstitution. Horizontal lines in each column mark mean values. Thymocytes from CB/Thy-NOD/SCID mice comprise a significantly higher percentage of CD4 single-positive cells than thymocytes from Thy-NOD/SCID (%CD4+ = 17.9 ± 3.3 v 6.8 ± 1.6, mean ± SEM, P < .01).

Fig. 2.

T-cell development in the thymic grafts of NOD/SCID-hu chimeras. Three-color flow cytometric analysis of thymocytes harvested from thymic grafts of a CB/Thy-NOD/SCID at 6 months post-CB reconstitution (a and b) and a control Thy-NOD/SCID mouse (c and d) stained for human CD45, CD4, and CD8. The staining specific for CD45 (a and c) identifies a population of human CD45+ lymphoid cells (Region 1, R1). Gating on R1 shows that human CD45+ cells from the grafts of CB/Thy-NOD/SCIDs comprise both CD4/CD8 double- and single-positive subsets (b), whereas human CD45+ cells from Thy-NOD/SCID mice harbor mainly CD4 and CD8 single-positive thymocytes (d). The vertical lines within the histograms (a and c) mark the upper limit of the fluorescence intensity corresponding to the isotype-control antibody. In the dot plots (b and d), quadrants were set to comprise the staining given by control antibodies in the left lower quadrant. Histograms and dot plots represent the analysis of 10,000 events. (e) Distribution of human thymic subsets in the grafts of individual CB/Thy-NOD/SCID (▪) and Thy-NOD/SCIDs (◊) at 6 months post-CB reconstitution. Horizontal lines in each column mark mean values. Thymocytes from CB/Thy-NOD/SCID mice comprise a significantly higher percentage of CD4 single-positive cells than thymocytes from Thy-NOD/SCID (%CD4+ = 17.9 ± 3.3 v 6.8 ± 1.6, mean ± SEM, P < .01).

Close modal

Further phenotypic analysis confirmed that human CD4 and CD8 single-positive thymocytes of either CB reconstituted or control mice expressed high levels of TCR α/β, HLA class I, and CD69 (not shown). This phenotype is consistent with that of terminally differentiated thymocytes, further evidence for a full thymic T-cell development program. However, thymocytes from Thy-NOD/SCID mice demonstrated a significantly lower viability as determined by light scatter analysis (eg, % viable TCRhigh cells = 59 ± 7.7 in Thy-NOD/SCID v 81 ± 4.8 in CB/Thy-NOD/SCID mice [n = 6], mean ± SEM, P < .05) suggesting that development in these control grafts is incomplete and many cells undergo cell death. Finally, flow cytometry of lymphocytes from the NOD/SCID mouse thymi showed the presence of less than 2% human CD45+cells, indicating that human T cells developed preferentially in the human thymic microenvironment.

To study the morphology of thymic grafts, sections from human fetal thymi before transplantation and after engraftment in vivo were stained for human CD45 and/or mouse major histocompatibility complex (MHC) class I antigens. In agreement with previous reports,34human fetal thymi cultured in the presence of deoxyguanosine were composed mainly of an epithelial network, almost completely devoid of human CD45+ cells (not shown). After engraftment in mice reconstituted with human BM, the thymic transplants displayed a defined cortex and medulla densely repopulated by human CD45+ cells (Fig 3a). Few mouse cells were observed within the grafts (Fig 3b, red fluorescence, arrow). In contrast, grafts from Thy-NOD/SCIDs showed a profoundly perturbed architecture, with no clear demarcation of cortical and medullary regions (Fig 3d). Consistent with the lower cellularity of these transplants, the grafts showed large areas depleted of lymphocytes or harboring only dispersed human CD45+ cells. Clusters of mouse cells were often observed within these grafts (Fig3e, red fluorescence).

Fig. 3.

Histological analysis of thymic transplants. Cryostat sections from thymic grafts of CB/Thy-NOD/SCID mice (a, b, c, g, and h) and control Thy-NOD/SCID mice (d through f) stained by a peroxidase detection method with an anti-human CD45 MoAb (a and d, dark brown color), by 2-color immunofluorescence with an anti-human CD45 MoAb (b and e, green fluorescence), and an anti-mouse MHC class I MoAb (b and e, red fluorescence) or by a TUNEL assay and propidium iodide (c and f, green and red fluorescence, respectively). (g and h) Background staining of isotype control antibodies for the immunohistochemistry and immunofluorescence techniques, respectively. The preservation of cortical and medullary regions (a, cx, and m) in the grafts of CB/Thy-NOD/SCID mice contrast with the perturbed tissue architecture of the grafts from control Thy-NOD/SCID mice (d). Clusters of mouse cells (e, arrows) and numerous apoptotic cells organized in rosette-like clusters (f) can be frequently observed in the grafts of Thy-NOD/SCID mice. In contrast, rare mouse cells (b, arrow) and sparse apoptotic events are observed in the grafts of CB/Thy-NOD/SCID mice (c). Bar in a, d, and g, 254 μm; Bar in b, c, e, and h, 63.5 μm.

Fig. 3.

Histological analysis of thymic transplants. Cryostat sections from thymic grafts of CB/Thy-NOD/SCID mice (a, b, c, g, and h) and control Thy-NOD/SCID mice (d through f) stained by a peroxidase detection method with an anti-human CD45 MoAb (a and d, dark brown color), by 2-color immunofluorescence with an anti-human CD45 MoAb (b and e, green fluorescence), and an anti-mouse MHC class I MoAb (b and e, red fluorescence) or by a TUNEL assay and propidium iodide (c and f, green and red fluorescence, respectively). (g and h) Background staining of isotype control antibodies for the immunohistochemistry and immunofluorescence techniques, respectively. The preservation of cortical and medullary regions (a, cx, and m) in the grafts of CB/Thy-NOD/SCID mice contrast with the perturbed tissue architecture of the grafts from control Thy-NOD/SCID mice (d). Clusters of mouse cells (e, arrows) and numerous apoptotic cells organized in rosette-like clusters (f) can be frequently observed in the grafts of Thy-NOD/SCID mice. In contrast, rare mouse cells (b, arrow) and sparse apoptotic events are observed in the grafts of CB/Thy-NOD/SCID mice (c). Bar in a, d, and g, 254 μm; Bar in b, c, e, and h, 63.5 μm.

Close modal

To investigate the occurrence of apoptosis in the developing thymic grafts, sections of thymic transplants were stained using a TdT-mediated dUTP nick-end labeling (TUNEL) assay. Thymic transplants from CB/Thy-NOD/SCID chimeras showed few apoptotic cells, mostly scattered in the cortex (Fig 3c, green fluorescence). In contrast, thymic grafts from the control Thy-NOD/SCIDs showed numerous apoptotic cells arranged in rosette-like clusters throughout the sections (Fig3f, green fluorescence) or grouped in foci of more than 50 cells around Hassals corpuscles (not shown). A rosette pattern of apoptotic cells has previously been described in pediatric thymi.35Two-color immunofluorescence to detect fragmented DNA and human CD45 demonstrated that the apoptotic cells were human (not shown). Morphometric analysis on more than 30 microscopic fields from sections cut at 100-μm intervals demonstrated that grafts from control Thy-NOD/SCIDs harbored a higher number of apoptotic events than those from mice reconstituted with human CB (number apoptotic cells per field = 0.99 ± 0.2 in Thy-NOD/SCIDs, n = 3 v 0.3 ± 0.1 in CB/Thy-NOD/SCIDs, n = 3, mean ± standard deviation [SD], P = .02; size of each field = 249 × 217 μm). This result supports the hypothesis that a high number of thymocytes developing in the grafts of the Thy-NOD/SCID mice die in the thymus and is consistent with the increased proportion of nonviable thymocytes shown by our fluorescence-activated cell sorting (FACS) analysis.

Taken together, the data indicate that on reconstitution of a human BM hematopoietic compartment by CB cells, human thymic grafts transplanted at a distant site maintain their thymopoietic capability. The data suggest that CB-derived stem cells generated thymocyte progenitors capable of homing to and developing within the human thymic grafts.

Detection of BM-derived progenitors in the thymic grafts.

The fact that some human CD45+ cells were detected in thymic grafts of control Thy-NOD/SCID mice indicated that some lymphoid cells of thymic donor origin had survived the dGuo treatment and may have contributed to the cellularity of the thymic grafts in the CB/Thy-NOD/SCIDs. To unequivocally demonstrate the presence of CB donor-derived thymocyte progenitors in the grafts of CB/Thy-NOD/SCIDs, chimeras were generated with male human CB cells and transplantation of female fetal thymi. At 4 months postthymic transplant, the CD4/CD8 double- and single-positive subsets from the thymic grafts were sorted and used for PCR amplification of Y chromosome DNA-specific sequences. These experiments showed that thymic transplants from CB/Thy-NOD/SCID mice harbored immature double-positive as well as mature single-positive thymocytes of CB origin (Fig 4). Thus, CB-derived thymocyte progenitors indeed home to the thymic grafts and develop through intermediates to the most mature single-positive stages. Parallel experiments using NOD/SCID mice transplanted with CB and thymi mismatched for HLA-A2, confirmed that as many as 95% of thymocytes repopulating the thymic transplants were of CB origin, as determined by FACS analysis for CD3, HLA-A2, CD4, and CD8.

Fig. 4.

Detection of CB-derived thymocytes in the thymic grafts of CB/Thy-NOD/SCID mice. CB/Thy-NOD/SCID chimeras were generated by BM reconstitution with male human CB followed by transplantation of female thymi. Control Thy-NOD/SCID received human female thymi only. At 6 months post-CB reconstitution, the CD4/CD8 double and single-positive thymic subsets were purified by cell sorting. DNA was extracted and assessed for the presence of Y chromosome–specific sequences by PCR. The resulting products resolved on a 1.2% agarose gel are shown. A strong band with the predicted size of 150 bp is detected in the BM, total thymocytes (Total Thys), as well as double and single-positive thymic subsets of CB/Thy-NOD/SCID mice (SCID no. 1 and no. 2), indicating the colonization of the grafts by BM-derived thymocyte progenitors of CB origin. DNA from total thymocytes of Thy-NOD/SCID mice (SCID controls) is negative. Each lane corresponds to the analysis of 10,000 cells. Control PCR using DNA extracted from male (♂) and female (♀) CB cells are shown on the left. The far left lane represents a 100-bp DNA ladder. The lane on the far right shows a control using Y chromosome–specific primers and no template DNA.

Fig. 4.

Detection of CB-derived thymocytes in the thymic grafts of CB/Thy-NOD/SCID mice. CB/Thy-NOD/SCID chimeras were generated by BM reconstitution with male human CB followed by transplantation of female thymi. Control Thy-NOD/SCID received human female thymi only. At 6 months post-CB reconstitution, the CD4/CD8 double and single-positive thymic subsets were purified by cell sorting. DNA was extracted and assessed for the presence of Y chromosome–specific sequences by PCR. The resulting products resolved on a 1.2% agarose gel are shown. A strong band with the predicted size of 150 bp is detected in the BM, total thymocytes (Total Thys), as well as double and single-positive thymic subsets of CB/Thy-NOD/SCID mice (SCID no. 1 and no. 2), indicating the colonization of the grafts by BM-derived thymocyte progenitors of CB origin. DNA from total thymocytes of Thy-NOD/SCID mice (SCID controls) is negative. Each lane corresponds to the analysis of 10,000 cells. Control PCR using DNA extracted from male (♂) and female (♀) CB cells are shown on the left. The far left lane represents a 100-bp DNA ladder. The lane on the far right shows a control using Y chromosome–specific primers and no template DNA.

Close modal
Circulating T cells are present in NOD/SCID chimeras reconstituted with human CB and thymus.

To assess whether reconstitution of human thymopoiesis in the NOD/SCID chimeras led to repopulation of the periphery by mature T cells, spleens and peripheral blood were screened for the presence of human CD45+ TCR α/β+ T cells by immunohistochemistry and/or flow cytometry. Human CD45+cells filling the periarteriolar T-cell–dependent areas of the spleen were detected in mice reconstituted with human CB (Fig 5a), but not in control mice (Fig 5b). Flow cytometry of splenocytes demonstrated that human CD45+ cells represented 7.34% ± 1.6% (mean ± SEM, n = 5) of the total lymphoid cells populating the spleen of CB/Thy-NOD/SCIDs. Immunofluorescence of either splenocytes or PBL for human CD45 and TCR α/β or human CD45, CD4, and CD8 further demonstrated that the majority of circulating human lymphocytes in these mice were, in fact, T cells with mature CD4 and CD8 phenotypes (Fig 5c; %TCR α/β+ cells within the CD45 subset = 62.3 ± 7.8, n = 9). In contrast, less than 0.5% human T cells were detected in the spleen or peripheral blood of control Thy-NOD/SCID mice.

Fig. 5.

Circulating mature T cells are present in the periphery of CB/Thy-NOD/SCID chimeras. Splenic sections from a CB/Thy-NOD/SCID mouse at 6 months post-CB reconstitution (a) and a control Thy-NOD/SCID mouse (b) stained with an anti-human–specific CD45 MoAb using a peroxidase detection method. Human CD45+ cells occupying the periarteriolar areas are observed in the spleen of CB/Thy-NOD/SCID mouse (a). The brown staining detected in the spleen of the control Thy-NOD/SCID mouse (b) corresponds to the background levels of staining obtained with the isotype control antibody (not shown). The staining is representative of 3 CB/Thy-NOD/SCID and 4 Thy-NOD/SCID chimeras generated in independent experiments. Percentage of human TCR /β+ lymphocytes (c) in the periphery of individual CB/Thy-NOD/SCIDs as detected by flow cytometry of splenocytes or PBL. The proportion of CD4+ (closed bars) and CD8+ (open bars) lymphocytes within the human TCR /β+ population is represented. The NOD/SCID chimeras are numerated as in Fig 1 to allow direct comparison of levels of reconstitution in the BM and thymic compartments. (d) Confocal microscopy of a splenic section from a CB/Thy-NOD/SCID mouse reconstituted with male CB stained by 2-color FISH with human Y chromosome-specific probes (green fluorescence) and anti-CD4 and CD8 MoAbs (red fluorescence). This staining is representative of 3 CB/Thy-NOD/SCID chimeras generated in independent experiments. The inset in (d) shows a magnified detail of Y chromosome-positive cells identified in the spleen. Bar in (a and b) = 125 μm; bar in d = 63.5 μm.

Fig. 5.

Circulating mature T cells are present in the periphery of CB/Thy-NOD/SCID chimeras. Splenic sections from a CB/Thy-NOD/SCID mouse at 6 months post-CB reconstitution (a) and a control Thy-NOD/SCID mouse (b) stained with an anti-human–specific CD45 MoAb using a peroxidase detection method. Human CD45+ cells occupying the periarteriolar areas are observed in the spleen of CB/Thy-NOD/SCID mouse (a). The brown staining detected in the spleen of the control Thy-NOD/SCID mouse (b) corresponds to the background levels of staining obtained with the isotype control antibody (not shown). The staining is representative of 3 CB/Thy-NOD/SCID and 4 Thy-NOD/SCID chimeras generated in independent experiments. Percentage of human TCR /β+ lymphocytes (c) in the periphery of individual CB/Thy-NOD/SCIDs as detected by flow cytometry of splenocytes or PBL. The proportion of CD4+ (closed bars) and CD8+ (open bars) lymphocytes within the human TCR /β+ population is represented. The NOD/SCID chimeras are numerated as in Fig 1 to allow direct comparison of levels of reconstitution in the BM and thymic compartments. (d) Confocal microscopy of a splenic section from a CB/Thy-NOD/SCID mouse reconstituted with male CB stained by 2-color FISH with human Y chromosome-specific probes (green fluorescence) and anti-CD4 and CD8 MoAbs (red fluorescence). This staining is representative of 3 CB/Thy-NOD/SCID chimeras generated in independent experiments. The inset in (d) shows a magnified detail of Y chromosome-positive cells identified in the spleen. Bar in (a and b) = 125 μm; bar in d = 63.5 μm.

Close modal

To directly demonstrate that circulating T cells were BM-derived and therefore of CB origin, splenic sections from the chimeras reconstituted with male CB and female thymi were analyzed by FISH for the presence of cells expressing the human Y chromosome and the T-cell markers CD4 and CD8 (Fig 5d). Numerous cells coexpressing male Y chromosome (green fluorescence) and CD4/CD8 markers (red fluorescence) were observed. Notably, not all of the cells expressing the CD4/CD8 markers displayed a positive signal for the Y chromosome. This result may reflect a limitation of this technique to detect chromosomes on a single focal plane imposed by the tissue sectioning. Similar results were obtained in 2 other chimeras. Unlike SCID mice reconstituted with T- and B-cell–depleted CB followed by transplantation with thymic tissue, we never detected circulating mature T cells in SCID mice without thymic transplants, regardless of whether they were reconstituted with either whole CB (n = 50)36 or purified CD34+ cells (n = 20) (B.E.T., unpublished observations). Altogether, these results indicate that colonization of the thymic grafts by CB-derived thymocyte progenitors generated a mature T-cell progeny capable of exit and repopulation of peripheral immune compartments.

Identification of human endothelial cells at the site of the thymic implant.

Revascularization plays an important role in the engraftment of transplants. In the case of thymic grafts, the newly formed vasculature is also a primary component regulating the homing of progenitors and possibly release of mature T cells from this organ.37Because these processes of lymphocyte trafficking may be restricted by species-specific adhesive interactions on the endothelium,38 we next investigated the extent to which host and/or donor endothelium contributed to the revascularization of the thymic transplants. For this purpose, sections were stained with an anti-human CD34 MoAb and an anti–PECAM-1 polyclonal antibody. In experiments testing the species specificity of these antibodies, we found that the anti–PECAM-1 antibody cross-reacts with human and mouse endothelium, whereas the anti-CD34 MoAb is human-specific. Figure 6 shows a series of microscopic fields from a thymic graft of a CB/Thy-NOD/SCID chimera. Numerous human blood vessels identified by the coexpression of CD34 and PECAM-1 (Fig 6a and b; coexpression marked by yellow) were observed in the subcapsular and interlobular connective tissue (Fig6a, arrows). Many human vessels were also identified infiltrating the mouse kidney parenchyma directly adjacent to the graft (Fig 6b). To determine the distance human vessels could be detected within the surrounding mouse tissue, we analyzed a series of consecutive microscopic fields (Fig 6d). Human vessels were detectable within a range of ≈700 μm from the edge of the thymic graft. Thymic transplants of control Thy-NOD/SCID mice demonstrated a similar human vascular network, indicating that these vessels may form from endothelial elements contained within the transplant. This human vascular component provides a mechanism by which human marrow-derived T-cell progenitors could be effectively targeted to human thymic grafts at a distant site in our xenogeneic model.

Fig. 6.

Human blood vessels contribute to the vascularization of the thymic implant. Confocal microscopy of cryostat sections from the thymic graft of a CB/Thy-NOD/SCID mouse stained by 2-color immunofluorescence with an anti-human–specific CD34 MoAb (a, b, and d, red fluorescence) and an anti–PECAM-1 polyclonal Ab (a, b, and d, green fluorescence) or with isotype-matched control antibodies (c). Colocalization of the 2 markers (yellow) identifies numerous human vascular structures within the thymic parenchyma, as well as in the subcapsular region of the graft (a, arrows). Human vessels are also seen to penetrate the mouse kidney at the interface with the thymic graft (b, interface marked by dots). (d) Composite of 4 consecutive microscopic fields acquired from a section stained as above showing numerous human vessels infiltrating the mouse kidney at the interface with the thymic graft. Bar in a, b, and c = 42 μm; bar in d = 32 μm.

Fig. 6.

Human blood vessels contribute to the vascularization of the thymic implant. Confocal microscopy of cryostat sections from the thymic graft of a CB/Thy-NOD/SCID mouse stained by 2-color immunofluorescence with an anti-human–specific CD34 MoAb (a, b, and d, red fluorescence) and an anti–PECAM-1 polyclonal Ab (a, b, and d, green fluorescence) or with isotype-matched control antibodies (c). Colocalization of the 2 markers (yellow) identifies numerous human vascular structures within the thymic parenchyma, as well as in the subcapsular region of the graft (a, arrows). Human vessels are also seen to penetrate the mouse kidney at the interface with the thymic graft (b, interface marked by dots). (d) Composite of 4 consecutive microscopic fields acquired from a section stained as above showing numerous human vessels infiltrating the mouse kidney at the interface with the thymic graft. Bar in a, b, and c = 42 μm; bar in d = 32 μm.

Close modal
CB-derived progenitors of the endothelial cell lineage contribute to the vascularization of thymic transplants and wound healing.

Previous studies have shown that an adherent fraction of human CB cells can be induced to differentiate in vitro into endothelial cells.39 This observation suggested that human CB may contain circulating endothelial cells or endothelial progenitors. To test whether CB-derived cells of the endothelial lineage had engrafted in our NOD/SCID chimeras and participated in the revascularization of the thymic transplants, frozen sections from female thymi engrafted into mice reconstituted with male CB were screened for the presence of male cells at the interface of the mouse kidney with the thymic grafts. This anatomically discrete region contained human CD34+blood vessels (Fig 6), but lacked detectable human CD45+lymphoid cells as assessed by confocal microscopy (not shown). The presence of human Y chromosome–positive cells at this site was first investigated by a PCR technique, which allows amplification of gene sequences from tissue sections.31 Tissue samples from thymic graft/kidney interfaces were dissected from frozen sections under microscopic visualization. DNA was extracted and used for PCR of human Y chromosome–specific sequence (Fig 7). DNA from regions within the thymic grafts containing thymocytes of BM origin (eg, male) served as positive controls. In addition, to exclude the possibility of detecting Y chromosome–positive cells of marrow origin circulating in the mouse blood stream, DNA from the mouse kidney distant from the grafts was also used. Figure 7a documents the tissue sampling sites from a representative tissue section. Human Y chromosome DNA was detected at the interface of the thymic grafts with the mouse kidney, as well as within the thymic grafts, but not within the mouse kidney at sites distant from the graft (Fig 7b). The data indicate that CB-derived cells are resident at the interface of the mouse kidney with the thymic implant in a region in which no CD45+ human cells (eg, leukocytes) are detected.

Fig. 7.

Detection of CB-derived endothelial cells at the site of thymic implant. (a) Cryostat section from a human female thymic transplant engrafted into a NOD/SCID mouse reconstituted with male CB, stained by hematoxylin/eosin showing the sites of tissue sampling used for DNA preparation. PCR of these DNA samples for human male Y chromosome–specific sequences (b) shows the presence of Y chromosome in tissue within the graft, as well as in the mouse kidney, at the interface with the graft, but not in the mouse kidney distant from the thymic transplant. The DNA products amplified from triplicate samples of the indicated anatomical regions obtained from 3 consecutive sections are shown. The product from a positive control PCR using male CB cells (♂) is also shown on the left. The far left lane represents a 100-bp DNA ladder. The lane on the far right shows a control PCR using Y chromosome–specific primers and no template DNA. (c through f) Confocal microscopy of female thymic grafts from CB/Thy-NOD/SCID mice reconstituted with male CB stained by 2-color FISH using human Y chromosome–specific probes (green fluorescence) and a human specific anti-CD34 MoAb (c and d, red fluorescence) or an anti–PECAM-1 polyclonal antibody (e, red fluorescence). (f) Background fluorescence obtained using the FITC-conjugated antidigoxigenin antibody and isotype-matched control antibodies for the red fluorescence. CD34+ cells bearing the male Y chromosome are detected at the interface of the grafts with the mouse kidney (c, arrowheads), and in the pericapsular connective tissue (d, arrowheads). The star in (c) indicates the thymic graft. Y chromosome–bearing cells comprised within vascular structures, unequivocally identified by the PECAM-1 staining (e, red fluorescence, arrowheads), as well as other nonendothelial stromal elements (e, arrows) are also observed at the interface of the grafts with the mouse kidney. Bar in c, d, and f, 63.5 μm; bar in e, 31 μm.

Fig. 7.

Detection of CB-derived endothelial cells at the site of thymic implant. (a) Cryostat section from a human female thymic transplant engrafted into a NOD/SCID mouse reconstituted with male CB, stained by hematoxylin/eosin showing the sites of tissue sampling used for DNA preparation. PCR of these DNA samples for human male Y chromosome–specific sequences (b) shows the presence of Y chromosome in tissue within the graft, as well as in the mouse kidney, at the interface with the graft, but not in the mouse kidney distant from the thymic transplant. The DNA products amplified from triplicate samples of the indicated anatomical regions obtained from 3 consecutive sections are shown. The product from a positive control PCR using male CB cells (♂) is also shown on the left. The far left lane represents a 100-bp DNA ladder. The lane on the far right shows a control PCR using Y chromosome–specific primers and no template DNA. (c through f) Confocal microscopy of female thymic grafts from CB/Thy-NOD/SCID mice reconstituted with male CB stained by 2-color FISH using human Y chromosome–specific probes (green fluorescence) and a human specific anti-CD34 MoAb (c and d, red fluorescence) or an anti–PECAM-1 polyclonal antibody (e, red fluorescence). (f) Background fluorescence obtained using the FITC-conjugated antidigoxigenin antibody and isotype-matched control antibodies for the red fluorescence. CD34+ cells bearing the male Y chromosome are detected at the interface of the grafts with the mouse kidney (c, arrowheads), and in the pericapsular connective tissue (d, arrowheads). The star in (c) indicates the thymic graft. Y chromosome–bearing cells comprised within vascular structures, unequivocally identified by the PECAM-1 staining (e, red fluorescence, arrowheads), as well as other nonendothelial stromal elements (e, arrows) are also observed at the interface of the grafts with the mouse kidney. Bar in c, d, and f, 63.5 μm; bar in e, 31 μm.

Close modal

To determine whether these cells at the graft interface comprised endothelial cells, we used the in situ fluorescence hybridization assay described above to identify cells coexpressing the endothelial markers CD34, PECAM-1, or vWF and human Y chromosome. In 3 of 5 grafts, this analysis showed the presence of stromal elements coexpressing CD34 or PECAM-1 and human Y chromosome (Fig 7c through e) or human vWF and human Y chromosome (Fig 8a and b). CD34+ cells bearing the Y chromosome were identified at the interface of the grafts with the mouse kidney (Fig 8a, arrows) and at a subcapsular location (Fig 8b). Figure 7e shows the interface of a graft with the mouse kidney stained for PECAM-1 (red fluorescence) and Y chromosome (green fluorescence). Cells coexpressing the 2 markers can be observed within a blood vessel (arrowheads), indicating the presence of CB-derived endothelial cells. Other Y chromosome+ cells negative for PECAM-1 (arrows) are also evident in this region. They appear as large fibroblast-like cells by light microscopy. Y chromosome+ endothelial or stromal cells were not observed within the thymic parenchyma (not shown). The presence of blood vessels of cord origin was further demonstrated by studying SCID chimeras transplanted with CB and thymic tissue mismatched for HLA-A2. Figure 8c and d shows microscopic fields at the thymic-kidney interface of 1 of such chimeras stained with an anti-human–specific vWF antibody (red fluorescence) and an anti–HLA-A2 specific MoAb (green fluorescence) identifying cells of thymic origin. As can be observed, blood vessels of both CB origin (eg, positive for vWF, but negative for HLA-A2, arrowheads) and thymic origin (eg, positive for both vWF and HLA-A2, arrows) are present at this site.

Fig. 8.

Detection of CB-derived blood vessels at the site of thymic implant in SCID CB/Thy chimeras mismatched for sex or HLA-A2. (a and b) Confocal microscopy of a thymic graft from a CB/Thy SCID chimera transplanted with CB and thymus mismatched by sex. Human blood vessels, here identified by the expression of vWF (red), are shown to comprise Y chromosome-positive cells of CB origin, as detected by FISH (green, arrowheads). (c and d) Confocal microscopy of a thymic graft from a CB/Thy SCID chimera transplanted with CB and thymus mismatched for the expression of HLA-A2. The transplanted thymic tissue was from an HLA-A2+ donor, whereas CB cells were from an HLA-A2 donor. Human blood vessels of thymic origin (arrows) are identified by the coexpression of vWF (red) and HLA-A2 (green). In addition, a number of human vWF+ blood vessels lacking expression of HLA-A2 are observed (arrowheads), indicating the participation of CB cells to the revascularization of the graft. Bar in b = 42.3 μm; bar in a, c, and d = 63.5 μm.

Fig. 8.

Detection of CB-derived blood vessels at the site of thymic implant in SCID CB/Thy chimeras mismatched for sex or HLA-A2. (a and b) Confocal microscopy of a thymic graft from a CB/Thy SCID chimera transplanted with CB and thymus mismatched by sex. Human blood vessels, here identified by the expression of vWF (red), are shown to comprise Y chromosome-positive cells of CB origin, as detected by FISH (green, arrowheads). (c and d) Confocal microscopy of a thymic graft from a CB/Thy SCID chimera transplanted with CB and thymus mismatched for the expression of HLA-A2. The transplanted thymic tissue was from an HLA-A2+ donor, whereas CB cells were from an HLA-A2 donor. Human blood vessels of thymic origin (arrows) are identified by the coexpression of vWF (red) and HLA-A2 (green). In addition, a number of human vWF+ blood vessels lacking expression of HLA-A2 are observed (arrowheads), indicating the participation of CB cells to the revascularization of the graft. Bar in b = 42.3 μm; bar in a, c, and d = 63.5 μm.

Close modal

These results indicate that unlike the human vessels described in the control Thy-NOD/SCIDs, which must be of thymic graft origin, recruitment of CB-derived cells of the endothelial lineage occurred in the CB/Thy-NOD/SCIDs. Quantitative histology using T-and B-depleted CB cells labeled with a fluorescent vital dye indicated that 40% to 60% of PECAM-1+ vascular profiles identified at the interface with the thymic grafts comprised cells of CB origin. We considered whether the formation of CB-derived vessels could be attributed to mature endothelial cells, which might contaminate our CB preparations. Thus, T- and B-depleted CB samples (n = 3) were analyzed by flow cytometry for cells expressing the mature endothelial markers PECAM-1, E-selectin, and vWF. These experiments showed that nonmyeloid (eg, CD11b) PECAM-1+ cells, possibly comprising mature endothelial cells, represented less than 0.5% of T- and B-depleted CB. Purification of this subset by positive selection on a magnetic column demonstrated that only 5.7% of CD11b/PECAM-1+ cells expressed vWF (ie, 0.02% of T- and B-cell–depleted CB cells) and could therefore be unequivocally identified as mature endothelial cells. Thus, at most, ≈3,000 vWF+ cells were injected per mouse in our reconstitution experiments. Further experiments in which as many as 2 × 106 HUVEC labeled with a vital dye were injected in our NOD/SCID model demonstrated that mature endothelial cells do not efficiently contribute to the development of blood vessels at the site of thymic transplantation (manuscript in preparation). Taken together, these experiments indicate that CB-derived endothelial cells present in the vessels between the mouse kidney and the transplants were derived from circulating progenitors trafficking into this site of surgical injury and subsequent healing.

In these studies, we demonstrate that CB hematopoietic progenitors, engrafted in the BM compartment of immunodeficient hosts, sustain human T-cell lymphopoiesis by generating circulating progenitors capable of homing to and developing within a human thymus. Furthermore, we provide evidence that the in vivo developmental program of CB stem cells is not limited to blood cell lineages, but also extends to the generation of endothelial cells, as well as stromal elements, which contribute to the revascularization of transplants and wound healing. This result discloses a novel role of CB-derived stem cells in angiogenesis.

Immunodeficient SCID and NOD/SCID mice are valuable in vivo models to study human hematopoiesis.40-43 In particular, studies have shown that intravenous injection of human BM or whole CB in these mice results in the engraftment of primitive hematopoietic stem cells of myeloid, erythroid, and B-cell lymphoid lineages.24-27 We show that injection of CB rigorously depleted of mature T and B cells consistently results in high levels of engraftment of human cells in the BM compartment. This observation indicates that mature T and/or B cells from CB are not required for initial engraftment of human stem cells.44,45 

Bone marrow reconstitution was demonstrated up to 6 months post-CB injection, indicating long-term in vivo self-renewal of the engrafted stem cells. However, by 6 months, exhaustion of the human BM component was evident in most of our chimeras. Several factors may explain this observation. First, it may reflect a limitation of the murine environment to support human hematopoiesis. Second, this result may indicate a propensity of primitive CB stem cells to proliferate at high rate in vitro,15,18 but for a limited number of cycles in vivo. Third, the purification procedure may deplete the CB of the most primitive progenitors or this population does not efficiently home to the BM compartment. Late graft failure after initial engraftment has been associated with T-cell–depleted allogeneic BM transplantation (BMT) in human patients.46 To date, only 1 report has investigated the repopulation capability of purified human CD34+ progenitors in NOD/SCID mice.26 While engraftment of CD34+ progenitors could be demonstrated up to 4 months postreconstitution, further experiments testing for the persistence of primitive hematopoietic progenitors by serial BM transplants showed much lower levels of BM engraftment in secondary as compared with primary recipients after 6 months (eg, 6% to 10% human cells in secondary recipients v 51% in primary recipients). These results suggest a decline in the frequency and/or engrafting capability of primitive progenitors after reconstitution with purified hematopoietic progenitors from CB.

A key result in our experiments is that engraftment of CB-derived BM progenitors supported human T lymphopoiesis in animals transplanted with human thymic grafts. In the presence of BM input, the thymic transplants increased in cellularity, preserved a relatively normal architecture, and supported thymocyte survival and development. Moreover, human thymopoiesis led to the production of mature T cells of CB origin that populated the periphery. These results represent the first evidence that CB stem cells comprise progenitors of the T-cell lineage capable of supporting continuous thymopoiesis in vivo from circulating elements. Recent studies have demonstrated that primitive hematopoietic progenitors from CB (eg, CD34+CD38) are required for long-term engraftment of human cells in the NOD/SCID BM.27 In contrast, more committed CD34+CD38+ progenitors appear not to engraft, as they are not detected 4 weeks after transplantation.27 This suggests that circulating thymic progenitors, which colonize the thymic grafts in our model, arise from primitive progenitors in the BM. The requirement of a thymic tissue for the generation of mature circulating T cells in our chimeras is consistent with ours as well other previous reports showing a lack of T-cell lymphopoiesis in SCID mice transplanted with purified CD34+ CB cells,36,47 BM, or fetal liver hemopoietic progenitors.48 Our observation has important implications for the use of CB in clinical transplantation. For example, it indicates that CB stem cells may successfully reconstitute pediatric recipients with naive thymus-derived T cells and reestablish a normal T-cell repertoire after ablative radiotherapy or chemotherapy. In light of our results and other transplant models,48,49CB reconstitution in combination with thymus transplantation may also be envisaged as a strategy to regenerate thymic-derived T-cell lymphopoiesis in aging, human immunodeficiency virus (HIV)-induced immunodeficiency, or autoimmune diseases.

T-cell lymphopoiesis in our model was dependent on the successful trafficking of circulating BM-derived progenitors to the human thymic graft. The contribution of human vessels to the revascularization of the thymic grafts in our chimeras may have played an important role in this process. Indeed, evidence that human hematopoietic cells can only establish weak adhesive interaction with the mouse endothelium has been previously reported.38 In contrast with the successful progenitor cell homing observed in our model, previous in vivo studies of human thymopoiesis using fragments of human fetal liver and fetal thymus showed that progenitors from the fetal liver grafts transplanted under the kidney capsule of SCID mice fail to reconstitute thymopoiesis in thymic tissue engrafted under the capsule of the opposite kidney.40 These data indicated a defect of progenitor homing in that model.

We show that CB-derived cells contribute to the generation of new blood vessels at sites of graft implantation and wound healing. This result provides evidence for the existence of a previously unrecognized source of endothelial progenitors within human CB. That such cells have characteristics of progenitors is supported by their ability to engraft and survive in vivo, while retaining the ability to home and develop into blood vessels at sites of wound healing. These data provide the first documentation that the developmental potential of CB stem cells in vivo is not restricted to hematopoietic lineages. The coexistence of hematopoietic and endothelial progenitors in CB may not be fortuitous. Much evidence supports a developmental and/or functional relationship between endothelial cells and hematopoietic precursors during embryogenesis. Thus, formation of blood vessels progresses simultaneously with hematopoiesis in the blood islands of the yolk sac; the center of these islands form primitive blood cells and the outer cell layers develop into endothelial cells.49,50 Peptides secreted by the developing thymic epithelium such as thymosin α1 may promote differentiation of immature thymocyte precursors51 and function as angiogenic factors for endothelial cells.52 The umbilical cord itself may represent a unique site of hematopoiesis and endothelial cell development. Hence, in the mouse embryo, the endothelium of the proximal portion of umbilical arteries display an unusual rounded morphology with clusters of CD34+ cells bound to the luminal surface.53 This pattern is reminiscent of that described for the paraaortic hematopoietic sites in the avian embryo.54 It suggests that the umbilical cord may comprise sites in which endothelial and hematopoietic progenitors coexist and be released into the embryo’s circulation.

The formation of new vessels from circulating progenitors contrasts with the current view that neovascularization of tissues in adult life occurs only by angiogenesis—the local proliferation of preexisting endothelial cells.55 The evidence we present for a role of circulating progenitors in neovascularization rather implies mechanisms of endothelial differentiation similar to those occurring during vasculogenesis, the process by which preendothelial cells condense to form new vascular channels in the embryo.55 Interestingly, a similar mechanism of vessel formation from circulating endothelial progenitors was recently shown in a model of limb ischemia. In this study, immunodeficient mice, injected with human CD34+cells purified from peripheral blood of adults, develop new human blood vessels in the ischemic tissue.56 The development of BM-derived endothelial cells has been also documented in dogs transplanted with allogeneic BM.57 Thus, the presence of circulating endothelial progenitors and their unique mode of vessel formation appears not to be limited to fetal life. Indeed, our studies suggest that reparative/inflammatory processes such as those occurring at sites of surgical injury and wound healing may provide stimuli for the mobilization, homing, and/or differentiation of such endothelial progenitors. Thus, the physiologic role of stem cells documented to circulate in the CB of newborns and peripheral blood of adults may not be restricted to hematopoiesis, but also extend to healing processes such as those following birth trauma, accidental tissue injury, and surgery.

We thank the medical staff of the Mary Birch Hospital for the procurement of the CB and Dr Alberto Hayek for instructing us on murine microsurgery. We also thank Drs Zaverio Ruggeri and Thomas Edgington for critical reading of this manuscript. This is TSRI manuscript 11495-MEM.

Supported in part by Grants No. RO1 DK49886-01 (to B.E.T.) and R01 AI42384-01 (to D.R.S.) from the National Institutes of Health (NIH). The National Center for Microscopy and Imaging Research is supported by NIH Grant No. RR-04050 (to M.H.E.). L.C. and V.C. were supported by Career Development Awards from The Juvenile Diabetes Foundation International. L.C. was also supported in part by a grant from The Scripps Clinic and Research Foundation, Department of Academic Affairs.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Friedrich
W
Goldman
SF
Ebell
W
Blutters-Sawatzki
R
Gaedich
G
Raghavachar
A
Peter
HH
Belohradsky
B
Kreth
W
Kubanech
B
Kleihauer
E
Severe combined immunodeficiency: Treatment by bone marrow transplantation in 15 infants using HLA haploidentical donors European survey 1968-1985.
Eur J Pediatr
144
1985
125
2
Parkman
R
The application of bone marrow transplantation to the treatment of genetic diseases.
Science
232
1986
1373
3
Korbling
MB
Dorken
AD
Ho
A
Pezzutto
W
Hunstein
Fliedner
TM
Autologous transplantation of blood-derived haemopoietic stem cells after myeloablative therapy in a patient with Burkitt’s lymphoma.
Blood
67
1986
529
4
Korbling
M
Przepiorka
D
Huh
YO
Engel
H
van Besien
K
Giralt
S
Andersson
A
Kleine
HD
Seong
D
Deisseroth
AB
Anreeff
M
Champlin
R
Allogeneic blood stem cell transplantation for refractory leukemia and lymphoma: Potential advantage of blood over marrow allografts.
Blood
85
1995
1659
5
Harrison
DE
Astle
CM
Loss of stem cell repopulating ability upon transplantation: Effects of donor age cell number and tranplant procedure.
J Exp Med
156
1982
1767
6
Christensen
RD
Circulating pluripotent hematopoietic progenitor cells in neonates.
J Pediatr
110
1987
622
7
Micklem
HS
Anderson
N
Ross
E
Limited potential of circulating haemopoietic stem cells.
Nature
256
1975
41
8
Molineux
G
Pojda
Z
Hampson
IN
Lord
BI
Dexter
TM
Transplantation potential of peripheral blood stem cells induced by granulocyte colony-stimulating factor.
Blood
76
1990
2153
9
Lubin
I
Faktorowich
Y
Lapidot
T
Gan
Y
Eshhar
Z
Gazit
E
Levite
M
Reisner
Y
Engraftment and development of human T and B cells in mice after bone marrow transplanattion.
Science
252
1991
427
10
Rebel
VI
Miller
CL
Thornbury
GR
Dragowska
WH
Eaves
CJ
Lansdorp
PM
A comparison of long-term repopulating hematopoietic stem cells in fetal liver and adult bone marrow from the mouse.
Exp Hematol
24
1996
638
11
Zhong
R
Astle
CM
Harrison
DE
Distinct developmental patterns of short-term and long-term functioning lymphoid and myeloid precursors defined by competitive limiting dilution analysis in vivo.
J Immunol
157
1996
138
12
Wagner
JE
Rosenthal
J
Sweetman
R
Shu
XO
Davies
SM
Ramsay
NKC
McGlave
PB
Sender
L
Cairo
MS
Successful transplantation of HLA-matched and HLA-mismatched umbilical cord blood from unrelated donors: Analysis of engraftment and acute graft-versus-host disease.
Blood
88
1996
795
13
Cairo
MS
Wagner
JE
Placental and/or umbilical cord blood: An alternative source of hematopoietic stem cells for transplantation.
Blood
90
1997
4665
14
Broxmeyer
HE
Douglas
GW
Hangoc
G
Cooper
S
Bard
J
English
D
Arny
M
Thomas
L
Boyse
EA
Human umbilical cord blood as a potential source of transplantable hematopietic stem/progenitor cells.
Proc Natl Acad Sci USA
86
1989
3828
15
Broxmeyer
HE
Hangoc
G
Cooper
S
Ribeiro
R
Graves
V
Yoder
M
Wagner
J
Vhadan-Raj
S
Benninger
L
Rubinstein
P
Broun
ER
Growth characteristics and expansion of human umbilical cord blood and estimation of its potential for transplantation in adults.
Proc Natl Acad Sci USA
89
1992
4109
16
Leary
AG
Ogawa
M
Blast cell colony assay for umbilical cord blood and adult bone marrow progenitors.
Blood
69
1987
953
17
Hows
JM
Bradley
BA
Marsh
JCW
Luft
T
Coutinho
L
Testa
NG
Dexter
TM
Growth of human umbilical cord blood in long term haemopoietic cultures.
Lancet
340
1992
73
18
Lu
L
Xiao
M
Shen
R
Grigsby
S
Broxmeyer
HE
Enrichment characterization and responsivness of single primitive CD34+ human umbilical cord blood hematopoietic progenitors with high proliferative and replating potential.
Blood
81
1993
41
19
Wagner
JE
Broxmeyer
HE
Byrd
RL
Zehnbauer
B
Schmeckpeper
B
Shah
N
Griffin
C
Emanuel
PD
Zuckerman
KS
Cooper
S
Carow
C
Bias
W
Santos
GW
Transplantation of umbilical cord blood after myeloablative therapy: Analysis of engraftment.
Blood
79
1992
1874
20
Emerson
SG
Sieff
CA
Wang
EA
Wong
GG
Clark
SC
Nathan
DG
Purification of fetal hematopoietic progenitors and demonstration of recombinant multipotential colony-stimulating activity.
J Clin Invest
76
1985
1286
21
Haneline
LS
Marshall
KP
Clapp
DW
The highest concentration of primitive hematopoietic progenitor cells in cord blood is found in extremely premature infants.
Pediatr Res
39
1996
820
22
Christensen
RD
Harper
TE
Rothstein
G
Granulocyte-macrophage progenitor cells in term and preterm neonates.
J Pediatr
109
1986
1047
23
Olson
T
Levine
R
Mazur
E
Wright
D
Salvado
A
Megakaryocytes and megakaryocyte progenitors in human cord blood.
Am J Pediatr Hematol Oncol
14
1992
241
24
Vormoor
J
Lapidot
T
Pflumio
F
Risdon
G
Patterson
B
Broxmeyer
HE
Dick
JE
Immature human cord blood progenitors engraft and proliferate to high levels in severe combined immunodeficient mice.
Blood
83
1994
2489
25
Pflumio
F
Izac
B
Katz
A
Shultz
LD
Vainchenker
W
Coulombel
L
Phenotype and function of human hematopoietic cells engrafting immunodeficient CB17-severe combined immunodeficient mice and nonobese diabetic-severe combined immunodeficiency mice after transplantation of human cord blood mononuclear cells.
Blood
88
1996
3731
26
Hogan
CJ
Shpall
EJ
McNulty
O
McNiece
I
Dick
JE
Shultz
LD
Keller
G
Engraftment and development of human CD34+-enriched cells from umbilical cord blood in NOD/LtSz-scid/scid mice.
Blood
90
1997
85
27
Larochelle
A
Vormoor
J
Hanenberg
H
Wang
JCY
Bhatia
M
Lapidot
T
Moritz
T
Murdoch
B
Xiao
XL
Kato
I
Williams
DA
Dick
JE
Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: Implications for gene therapy.
Nature Med
2
1996
1329
28
Yeoman
D
Gress
RE
Bare
CV
Leary
AG
Boyse
EA
Bard
J
Shultz
LD
Harris
DT
DeLuca
D
Human bone marrow and umbilical cord blood cells generate CD4+ and CD8+ single-positive T cells in murine fetal thymus organ culture.
Proc Natl Acad Sci USA
90
1993
10778
29
Verhasselt
B
De Smedt
M
Verhelst
R
Naessens
E
Plum
J
Retrovirally transduced CD34+ human cord blood cells generate T cells expressing high levels of the retroviral encoded green fluorescent protein marker in vitro.
Blood
91
1998
431
30
DiGiusto
DL
Lee
R
Moon
J
Moss
K
O’Toole
T
Voyotovich
A
Webster
D
Mule
JJ
Hematopoietic potential of cryopreserved and ex vivo manipulated umbilical cord blood progenitors cells evaluated in vitro and in vivo
Blood
87
1996
1261
31
Carow
CE
Harrington
MA
Broxmeyer
HE
Detection of human myeloid progenitor cells in a murine background.
Exp Hematol
21
1993
66
32
Hiller
T
Snell
L
Watson
PH
Microdissection RT-PCR analysis of gene expression in pathologically defined frozen tissue sections.
Biotecniques
21
1996
38
33
Gerritsen
WR
Jagiello
CA
Bourhis
J
Detection of chimerism in subpopulations of cells by fluorescence in situ hybridization and immunofluorescent staining of cell surface antigens.
Bone Marrow Transplant
13
1994
441
34
Jenkinson
EJ
Franchi
LL
Kingston
R
Owen
JJT
Effect of deoxyguanosine on lymphopoiesis in the developing thymus rudiment in vitro: Application in the production of chimeric thymus rudiments.
Eur J Immunol
12
1982
583
35
Le
PT
Maecker
HT
Cook
JE
In situ detection and characterization of apopototic thymocytes in human thymus expression of Bcl-2 in vivo does not prevent apoptosis.
J Immunol
154
1995
4371
36
Smith
KA
Conners
K
Crisa
L
Salomon
DR
Torbett
BE
Primitive human hematopoietic progenitors can be mobilized from the bone marrow of human-NOD/SCID mice.
Blood
90
1997
96a
37
Dunon
D
Imhof
BA
Mechanisms of thymus homing.
Blood
81
1993
1
38
Butcher
E
Scollay
R
Weissman
I
Evidence of continuous evolutionary change in structures mediating adherence of lymphocytes to specialized venules.
Nature
280
1979
496
39
Sutkowski
N
Kuo
M
Amenta
PS
Dougherty
JP
Ron
Y
A peripheral blood-derived monolayer supports long-term cultures of human CD4+ and CD8+ T lymphocytes.
Blood
85
1995
3213
40
McCune
JM
Namikawa
R
Kaneshima
H
Shultz
LD
Lieberman
M
Weissman
IL
The SCID-hu mouse murine model for the analysis of human hematolymphoid differentiation and function.
Science
241
1988
1632
41
Shultz
LD
Schweitzer
PA
Christianson
SW
Gott
B
Shweitzeir
IB
Tennent
B
McKenna
S
Mobraaten
L
Rajan
TV
Greiner
DL
Leiter
EH
Multiple defects in innate and adaptive immunologic function in NOD/LtSz-Scid mice.
J Immunol
154
1995
180
42
Larochelle
A
Vormoor
J
Lapidot
T
Sher
G
Furukawa
T
Li
Q
Shultz
LD
Olivieri
NF
Stamatoyannopoulos
G
Dick
JE
Engraftment of immune-deficient mice with primitive hematopoietic cells from β-thalassemia and sickle cell anemia patients: Implications for evaluating human gene therapy protocols.
Hum Mol Genet
4
1995
163
43
Lowry
PA
Shultz
LD
Greiner
DL
Hesselton
RM
Kittler
LW
Tiarks
CY
Rao
S
Reilly
J
Leif
JH
Ramshaw
H
Stewart
FM
Quesenberry
PJ
Improved engraftment of human cord blood stem cells in NOD/LtSz-Scid/Scid mice after irradiation or multiple-day injections into un-irradiated recipients.
Biol Blood Marrow Transplant
2
1996
15
44
Kaufman
CL
Colson
YL
Wren
SM
Watkins
S
Simmons
RL
Ildstad
ST
Phenotypic characterization of a novel bone marrow derived cell that facilitates engraftment of allogeneic bone marrow stem cells.
Blood
84
1994
2436
45
Wang
B
El-Badri
NS
Good
C
Good
R
Purified hematopoietic stem cells without facilitating cells can repopulate fully allogeneic recipients across entire major histocompatibility complex transplantation barrier in mice.
Proc Natl Acad Sci USA
94
1997
14632
46
Martin
PJ
Hansen
JA
Buckner
CD
Sanders
JE
Deeg
HG
Stewart
P
Appelbaum
FR
Clift
R
Feger
A
Witherspoon
RP
Effects of in vitro depletion of T cells in HLA-identical allogeneic marrow grafts.
Blood
66
1985
664
47
Hogan
CJ
Shpall
EJ
McNulty
O
McNiece
I
Dick
JE
Shultz
LD
Keller
G
Engraftment and development of human CD34+-enriched cells from umbilical cord blood in NOD/LtSz-scid/scid mice.
Blood
90
1997
85
48
Yurasov
S
Kollman
TR
Kim
A
Raker
CA
Hachamovitch
M
Wong-Staal
F
Goldstein
H
Severe combined immnodeficiency mice engrafted with human T cells B cells and myeloid cells after transplantation with human fetal bone marrow or liver cells and implanted with human fetal thymus: A model for studying human gene therapy.
Blood
89
1997
1800
49
Lee
L
Gritsch
A
Sergio
JJ
Arn
JS
Glaser
RM
Sablinski
T
Sachs
DH
Sykes
M
Specfic tolerance across a discordant xenogeneic transplantation barrier.
Proc Natl Acad Sci USA
91
1994
10864
50
Moore
M
Metcalf
D
Ontogeny of the hematopoietic system: Yolk sac origin of in vivo and in vitro colony forming cells in the developing mouse embryo.
Br J Haematol
18
1970
279
51
Hadden
EM
Malec
P
Sosa
M
Hadden
JW
Mixed interleukins and thymosin fraction V synergistically induce T lymphocyte development in hydrocortisone-treated aged mice.
Cell Immunol
144
1992
228
52
Malinda
MK
Sidhu
GS
Banaudha
KK
Gaddipati
JP
Maheshwari
RK
Goldstein
AL
Kleinman
HK
Thymosin-α1 stimulates endothelial cell migration angiogenesis and wound healing.
J Immunol
160
1998
1001
53
Wood
HB
May
G
Healy
L
Enver
T
Morris-Kay
GM
CD34 expression patterns during early mouse development are related to modes of blood vessel formation and reveal additional sites of hematopoiesis.
Blood
90
1997
2300
54
Dieterlen-Lievre
F
Martin
C
Diffuse intra-embryonic haemopoiesis in normal and chimeric avain development.
Dev Biol
88
1981
180
55
Risau
W
Vasculogenesis angiogenesis and endothelial cell differentiation during embryonic development
The Development of the Vascular System
Feinberg
RN
Sherer
GH
Auerbach
R
1991
58
Karger
Basel, Switzerland
56
Asahara
T
Murohara
T
Sullivan
A
Silver
M
van der Zee
R
Li
T
Witzenbichler
B
Schatterman
G
Isner
JM
Isolation of putative progenitor endothelial cells for angiogenesis.
Science
275
1997
964
57
Shi
Q
Rafii
S
Wu
MH-D
Wijelath
ES
Yu
C
Ishida
A
Fujita
Y
Kothari
S
Mohle
R
Sauvage
LR
Moore
MAS
Storb
RF
Hammond
WP
Evidence for circulating bone marrow-derived endothelial cells.
Blood
92
1998
362

Author notes

Address reprint requests to Daniel R. Salomon, Department of Molecular and Experimental Medicine, MEM55, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037; e-mail: dsalomon@scripps.edu.

Sign in via your Institution