Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Immunol ; 178(11): 7473-84, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17513799

ABSTRACT

Delayed immune reconstitution in adult recipients of allogeneic hemopoietic stem cell transplantations (HSCT) is related to age-induced thymic atrophy. Overcoming this paucity of T cell function is a major goal of clinical research but in the context of allogeneic transplants, any strategy must not exacerbate graft-vs-host disease (GVHD) yet ideally retain graft-vs-tumor (GVT) effects. We have shown sex steroid ablation reverses thymic atrophy and enhances T cell recovery in aged animals and in congenic bone marrow (BM) transplant but the latter does not have the complications of allogeneic T cell reactivity. We have examined whether sex steroid ablation promoted hemopoietic and T cell recovery following allogeneic HSCT and whether this benefit was negated by enhanced GVHD. BM and thymic cell numbers were significantly increased at 14 and 28 days after HSCT in castrated mice compared with sham-castrated controls. In the thymus, the numbers of donor-derived thymocytes and dendritic cells were significantly increased after HSCT and castration; donor-derived BM precursors and developing B cells were also significantly increased. Importantly, despite restoring T cell function, sex steroid inhibition did not exacerbate the development of GVHD or ameliorate GVT activity. Finally, IL-7 treatment in combination with castration had an additive effect on thymic cellularity following HSCT. These results indicate that sex steroid ablation can profoundly enhance thymic and hemopoietic recovery following allogeneic HSCT without increasing GVHD and maintaining GVT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Orchiectomy , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/transplantation , Bone Marrow Transplantation/immunology , Combined Modality Therapy , Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Interleukin-7/administration & dosage , Interleukin-7/deficiency , Interleukin-7/genetics , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Spleen/immunology , Spleen/transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Thymus Gland/immunology , Thymus Gland/transplantation , Transplantation, Homologous/immunology
2.
Nat Med ; 12(9): 1039-47, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936725

ABSTRACT

Immunoincompetence after allogeneic hematopoietic stem cell transplantation (HSCT) affects in particular the T-cell lineage and is associated with an increased risk for infections, graft failure and malignant relapse. To generate large numbers of T-cell precursors for adoptive therapy, we cultured mouse hematopoietic stem cells (HSCs) in vitro on OP9 mouse stromal cells expressing the Notch-1 ligand Delta-like-1 (OP9-DL1). We infused these cells, together with T-cell-depleted mouse bone marrow or purified HSCs, into lethally irradiated allogeneic recipients and determined their effect on T-cell reconstitution after transplantation. Recipients of OP9-DL1-derived T-cell precursors showed increased thymic cellularity and substantially improved donor T-cell chimerism (versus recipients of bone marrow or HSCs only). OP9-DL1-derived T-cell precursors gave rise to host-tolerant CD4+ and CD8+ populations with normal T-cell antigen receptor repertoires, cytokine secretion and proliferative responses to antigen. Administration of OP9-DL1-derived T-cell precursors increased resistance to infection with Listeria monocytogenes and mediated significant graft-versus-tumor (GVT) activity but not graft-versus-host disease (GVHD). We conclude that the adoptive transfer of OP9-DL1-derived T-cell precursors markedly enhances T-cell reconstitution after transplantation, resulting in GVT activity without GVHD.


Subject(s)
Adoptive Transfer , Hematopoietic Stem Cell Transplantation , Stem Cells/physiology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Animals , Coculture Techniques , Fibroblast Growth Factor 7/pharmacology , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Listeriosis/immunology , Lymphocyte Depletion , Mice , Regeneration , T-Lymphocytes/drug effects
3.
Blood ; 107(4): 1703-11, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16291587

ABSTRACT

The alpha4beta7 integrin plays a central role in the homing of T cells to the gut. We hypothesized that absence of the beta7 subunit would result in a reduction of intestinal graft-versus-host disease (GVHD) and an improvement in overall GVHD morbidity and mortality in recipients of hematopoietic stem cell transplantation (HSCT). Analysis of alloreactive beta7-/- T cells showed intact activation, proliferation, cytokine production, and cytotoxicity. However, recipients of beta7-/- donor T cells in murine HSCT models experienced less GVHD morbidity and mortality than recipients of wild-type (WT) T cells, associated with a decrease in donor T-cell infiltration of the liver and intestine and with an overall significant decrease in hepatic and intestinal GVHD. In graft-versus-tumor (GVT) experiments, we demonstrated intact or even enhanced GVT activity of beta7-/- donor T cells. In conclusion, beta7-/- donor T cells caused less GVHD morbidity and mortality than WT donor T cells because of selectively decreased T-cell infiltration of the liver and intestines. Our data suggest that strategies to target the beta7 integrin have the clinical potential to alleviate or prevent GVHD while sparing or potentiating GVT activity.


Subject(s)
Graft vs Host Disease/prevention & control , Integrin beta Chains/genetics , Mastocytoma/immunology , Mastocytoma/therapy , T-Lymphocytes/immunology , Animals , Cytotoxicity, Immunologic , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Deletion , Graft vs Host Disease/immunology , Immunity, Mucosal , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
4.
Blood ; 107(6): 2453-60, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16304055

ABSTRACT

Keratinocyte growth factor (KGF) is a member of the fibroblast growth factor family that mediates epithelial cell proliferation and differentiation in a variety of tissues, including the thymus. We studied the role of KGF in T-cell development with KGF-/- mice and demonstrated that thymic cellularity and the distribution of thymocyte subsets among KGF-/-, wildtype (WT), and KGF+/- mice were similar. However, KGF-/- mice are more vulnerable to sublethal irradiation (450 cGy), and a significant decrease was found in thymic cellularity after irradiation. Defective thymopoiesis and peripheral T-cell reconstitution were found in KGF-/- recipients of syngeneic or allogeneic bone marrow transplant, but using KGF-/- mice as a donor did not affect T-cell development after transplantation. Despite causing an early developmental block in the thymus, administration of KGF to young and old mice enhanced thymopoiesis. Exogenous KGF also accelerated thymic recovery after irradiation, cyclophosphamide, and dexamethasone treatment. Finally, we found that administering KGF before bone marrow transplantation (BMT) resulted in enhanced thymopoiesis and peripheral T-cell numbers in middle-aged recipients of an allogeneic BM transplant. We conclude that KGF plays a critical role in postnatal thymic regeneration and may be useful in treating immune deficiency conditions.


Subject(s)
Fibroblast Growth Factor 7/pharmacology , Regeneration/drug effects , Thymus Gland/physiology , Age Factors , Animals , Bone Marrow Transplantation , Lymphopoiesis/drug effects , Mice , Mice, Knockout , T-Lymphocytes/cytology , Thymus Gland/cytology
5.
Blood ; 106(9): 3322-30, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16037386

ABSTRACT

Graft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic stem cell transplantation (HSCT). Migration of donor-derived T cells into GVHD target organs plays a critical role in the development of GVHD and chemokines and their receptors are important molecules involved in this process. Here, we demonstrate in murine bone marrow transplantation models that the expression of the inflammatory CC chemokine receptor 2 (CCR2) on donor-derived CD8+ T cells is relevant for the control of CD8+ T-cell migration and development of GVHD. Recipients of CCR2-deficient (CCR2-/-) CD8+ T cells developed less damage of gut and liver than recipients of wild-type CD8+ T cells, which correlated with a reduction in overall GVHD morbidity and mortality. Assessment of donor CD8+ T-cell target organ infiltration revealed that CCR2-/- CD8+ T cells have an intrinsic migratory defect to the gut and liver. Other causes for the reduction in GVHD could be excluded, as alloreactive proliferation, activation, IFN-gamma production and cytotoxicity of CCR2-/- CD8+ T cells were intact. Interestingly, the graft-versus-tumor effect mediated by CCR2-/- CD8+ T cells was preserved, which suggests that interference with T-cell migration by blockade of CCR2 signaling can separate GVHD from GVT activity.


Subject(s)
CD8 Antigens/immunology , Graft vs Host Disease/immunology , Receptors, Chemokine/metabolism , Animals , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Movement , Cell Proliferation , Female , Interferon-gamma/biosynthesis , Intestinal Mucosa/metabolism , Intestines/cytology , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , Pancytopenia/metabolism , Receptors, CCR2 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Survival Rate , Thymus Gland/metabolism
6.
Blood ; 106(9): 3285-92, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-15956289

ABSTRACT

Inducible costimulator (ICOS) is expressed on activated and memory T cells and is involved in the regulation of cytokine production. We studied the role of ICOS on alloreactive T cells in graft versus host disease (GVHD) and determined that ICOS expression was up-regulated on alloreactive T cells in recipients of an allogeneic hematopoietic stem cell transplantation (allo-HSCT) with GVHD. We compared ICOS-/- T cells with wild-type (WT) T cells in 2 GVHD models. In both models, recipients of ICOS-/- T cells demonstrated significantly less GVHD morbidity and mortality, which was associated with less intestinal and hepatic GVHD but increased cutaneous GVHD. In addition, recipients of ICOS-/- donor T cells displayed a slight decrease in graft versus leukemia (GVL) activity. Further analysis of alloreactive ICOS-/- T cells showed no defect in activation, proliferation, cytotoxicity, and target organ infiltration. Recipients of ICOS-/- T cells had decreased serum levels of interferon-gamma (IFN-gamma), while interleukin-4 (IL-4) and IL-10 levels were increased, suggesting that alloreactive ICOS-/- T cells are skewed toward T helper-2 (Th2) differentiation. These data suggest a novel role for ICOS in the regulation of Th1/Th2 development of activated T cells. In conclusion, alloreactive ICOS-/- donor T cells induce less GVHD due to a Th2 immune deviation while GVL activity is slightly diminished.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Graft vs Host Disease/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Th2 Cells/immunology , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Differentiation , Cell Line , Cell Proliferation , Female , Hematopoietic Stem Cell Transplantation , Inducible T-Cell Co-Stimulator Protein , Mice , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism , Up-Regulation
7.
Blood ; 105(2): 865-73, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15280205

ABSTRACT

Interleukin-15 (IL-15) is a gamma-common cytokine that plays an important role in the development, survival, and proliferation of natural killer (NK), NK T, and CD8+ T-cells. We administered IL-15 to recipients of an allogeneic bone marrow transplantation (allo BMT) to determine its effects on immune reconstitution. Posttransplantation IL-15 administration significantly increased donor-derived CD8+ T (mostly CD122(+)CD44(+)CD8+ T-cells), NK, and NK T-cells at day +28 in young and old recipients of allo BMT. This was associated with enhanced T-cell and NK-cell function. IL-15 stimulated homeostatic proliferation of donor CD8+ T-cells in recipients of carboxyfluorescein diacetate succinimidyl ester-labeled donor T-cell infusions. Posttransplantation IL-15 administration also resulted in a decrease in apoptotic CD8+ T-cells, an increase in Bcl-2-expressing CD8+ T-cells, and an increase in the fraction of Ki67+ proliferative NK and CD8+ T-cells in recipients of allo BMT. IL-15 did not exacerbate graft-versus-host disease (GVHD) in recipients of T-cell-depleted BMT but could aggravate GVHD in some cases in recipients of a T-cell-repleted BMT. Finally, we found that IL-15 administration could enhance graft-versus-leukemia activity. In conclusion, IL-15 can be administered safely to recipients of a T-cell-depleted allo BMT to enhance CD8+ T, NK, and NK T-cell reconstitution.


Subject(s)
Bone Marrow Transplantation/immunology , Interleukin-15/pharmacology , Recovery of Function/immunology , Animals , Apoptosis/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Division/drug effects , Female , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Hyaluronan Receptors/metabolism , Immunologic Memory , Killer Cells, Natural/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Receptors, Interleukin-2/metabolism , Recovery of Function/drug effects , Spleen/cytology , Spleen/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Transplantation, Homologous
8.
J Exp Med ; 200(2): 149-57, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15249593

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor family-related gene (GITR) is a member of the tumor necrosis factor receptor (TNFR) family that is expressed at low levels on unstimulated T cells, B cells, and macrophages. Upon activation, CD4(+) and CD8(+) T cells up-regulate GITR expression, whereas immunoregulatory T cells constitutively express high levels of GITR. Here, we show that GITR may regulate alloreactive responses during graft-versus-host disease (GVHD) after allogeneic bone marrow transplantation (BMT). Using a BMT model with major histocompatibility complex class I and class II disparity, we demonstrate that GITR stimulation in vitro and in vivo enhances alloreactive CD8(+)CD25(-) T cell proliferation, whereas it decreases alloreactive CD4(+)CD25(-) proliferation. Allo-stimulated CD4(+)CD25(-) cells show increased apoptosis upon GITR stimulation that is dependent on the Fas-FasL pathway. Recipients of an allograft containing CD8(+)CD25(-) donor T cells had increased GVHD morbidity and mortality in the presence of GITR-activating antibody (Ab). Conversely, recipients of an allograft with CD4(+)CD25(-) T cells showed a significant decrease in GVHD when treated with a GITR-activating Ab. Our findings indicate that GITR has opposite effects on the regulation of alloreactive CD4(+) and CD8(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Animals , Apoptosis , Bone Marrow Transplantation , Cell Division , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Glucocorticoid-Induced TNFR-Related Protein , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin-2/biosynthesis , Up-Regulation
9.
J Clin Invest ; 112(7): 1095-107, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14523046

ABSTRACT

We used clinically relevant murine allogeneic bone marrow transplantation (BMT) models to study the mechanisms by which IL-7 administration can improve posttransplant peripheral T cell reconstitution. After transplant we could distinguish two populations of mature donor T cells: (a) alloreactive T cells with decreased expression of CD127 (IL-7 receptor alpha chain) and (b) nonalloreactive T cells, which express CD127 and undergo homeostatic proliferation. IL-7 administration increased the homeostatic proliferation of nonalloreactive T cells, but had no effect on alloreactive T cells and the development of graft-versus-host disease. Allogeneic transplant of purified hematopoietic stem cells and adoptive transfer of thymocytes into lethally irradiated hosts suggested that recent thymic emigrants can undergo homeostatic proliferation and acquire a memory-like phenotype. We found by BrdU pulse-chase, cell cycle, and annexin V analyses that IL-7 administration has significant proliferative and antiapoptotic effects on posttransplant peripheral T cells. We conclude that homeostatic expansion is important for T cell reconstitution after allogeneic BMT and involves both transferred mature T cells and recent thymic emigrants. Apart from its thymopoietic effects, IL-7 promotes peripheral T cell reconstitution through its selective proliferative and antiapoptotic effects on nonalloreactive and de novo-generated T cells, but has no effect on alloreactive T cells.


Subject(s)
Hematopoietic Stem Cell Transplantation , Interleukin-7/pharmacology , T-Lymphocytes/drug effects , Animals , Female , Graft vs Host Disease/mortality , Homeostasis , Hyaluronan Receptors/analysis , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Receptors, Interleukin-2/analysis , Receptors, Interleukin-7/analysis , T-Lymphocytes/immunology , Transplantation, Homologous
10.
Transplantation ; 75(12): 1977-83, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12829897

ABSTRACT

BACKGROUND: Prolonged immunodeficiency after allogeneic bone marrow transplantation (allo BMT) results in significant morbidity and mortality from infection. Previous studies in murine syngeneic BMT models have demonstrated that posttransplantation insulin-like growth factor (IGF)-I administration could enhance immune reconstitution. METHODS: To analyze the effects of IGF-I on immune reconstitution and graft-versus-host disease (GVHD) after allo BMT, we used murine models for MHC-matched and -mismatched allo BMT. Young (3-month-old) recipient mice received 4 mg/kg per day of human IGF-I from days 14 to 28 by continuous subcutaneous administration. RESULTS: IGF-I administration resulted in increased thymic precursor populations (triple negative-2 and triple negative-3) as determined on day 28 but had no effect on overall thymic cellularity. In the periphery, the numbers of donor-derived splenic CD3+ T cells were increased and these cells had an improved proliferative response to mitogen stimulation. IGF-I treatment also significantly increased the numbers of pro-, pre-, and mature B cells and myeloid cell populations in the spleens of allo BMT recipients on day 28. The administration of IGF-I in combination with interleukin 7 had a remarkable additive effect on B-cell, but not on T-cell, lymphopoiesis. Finally, we tested the effects of IGF-I administration on the development of GVHD in three different MHC-matched and -mismatched models and found no changes in GVHD morbidity and mortality. CONCLUSION: IGF-I administration can enhance lymphoid and myeloid reconstitution after allo BMT without aggravating GVHD.


Subject(s)
Bone Marrow Transplantation/physiology , Graft vs Host Disease/prevention & control , Insulin-Like Growth Factor I/therapeutic use , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Transplantation/immunology , Cell Differentiation/drug effects , Granulocytes/drug effects , Granulocytes/physiology , Humans , Infusions, Parenteral , Insulin-Like Growth Factor I/administration & dosage , Lymphocyte Activation/drug effects , Major Histocompatibility Complex , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , T-Lymphocytes/drug effects , Transplantation, Homologous , Transplantation, Isogeneic
11.
Blood ; 101(6): 2440-5, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12424195

ABSTRACT

Previous studies in murine bone marrow transplantation (BMT) models using neutralizing anti-tumor necrosis factor (TNF) antibodies or TNF receptor (TNFR)-deficient recipients have demonstrated that TNF can be involved in both graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL). TNF in these GVHD and GVL models was thought to be primarily produced by activated monocytes and macrophages, and the role of T cell-derived TNF was not determined. We used TNF(-/-) mice to study the specific role of TNF produced by donor T cells in a well-established parent-into-F1 hybrid model (C57BL/6J-->C3FeB6F1/J). Recipients of TNF(-/-) T cells developed significantly less morbidity and mortality from GVHD than recipients of wild-type (wt) T cells. Histology of GVHD target organs revealed significantly less damage in thymus, small bowel, and large bowel, but not in liver or skin tissues from recipients of TNF(-/-) T cells. Recipients of TNF(-/-) T cells which were also inoculated with leukemia cells at the time of BMT showed increased mortality from leukemia when compared with recipients of wt cells. We found that TNF(-/-) T cells do not have intrinsic defects in vitro or in vivo in proliferation, IFN-gamma production, or alloactivation. We could not detect TNF in the serum of our transplant recipients, suggesting that T cells contribute to GVHD and GVL via membrane-bound or locally released TNF.


Subject(s)
Bone Marrow Transplantation , Graft vs Host Disease , Graft vs Leukemia Effect , T-Lymphocytes/metabolism , Tissue Donors , Tumor Necrosis Factor-alpha/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/immunology , Intestine, Large/pathology , Intestine, Small/pathology , Leukemia, Experimental/pathology , Liver/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , T-Lymphocytes/immunology , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/deficiency
12.
Nat Med ; 8(12): 1433-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12426560

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that exhibits specific tumoricidal activity against a variety of tumors. It is expressed on different cells of the immune system and plays a role in natural killer cell-mediated tumor surveillance. In allogeneic hematopoietic-cell transplantation, the reactivity of the donor T cell against malignant cells is essential for the graft-versus-tumor (GVT) effect. Cytolytic activity of T cells is primarily mediated through the Fas-Fas ligand and perforin-granzyme pathways. However, T cells deficient for both Fas ligand and perforin can still exert GVT activity in vivo in mouse models. To uncover a potential role for TRAIL in donor T cell-mediated GVT activity, we compared donor T cells from TRAIL-deficient and wild-type mice in clinically relevant mouse bone-marrow transplantation models. We found that alloreactive T cells can express TRAIL, but the absence of TRAIL had no effect on their proliferative and cytokine response to alloantigens. TRAIL-deficient T cells showed significantly lower GVT activity than did TRAIL-expressing T cells, but no important differences in graft-versus-host disease, a major complication of allogeneic hematopoietic cell transplantation, were observed. These data suggest that strategies to enhance TRAIL-mediated GVT activity could decrease relapse rates of malignancies after hematopoietic cell transplantation without exacerbation of graft-versus-host disease.


Subject(s)
Graft vs Tumor Effect/immunology , Membrane Glycoproteins/physiology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis Regulatory Proteins , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , TNF-Related Apoptosis-Inducing Ligand , Transplantation, Homologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...