Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Proc Natl Acad Sci U S A ; 116(14): 6995-7004, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30894483

ABSTRACT

Aging is associated with impaired tissue regeneration. Stem cell number and function have been identified as potential culprits. We first demonstrate a direct correlation between stem cell number and time to bone fracture union in a human patient cohort. We then devised an animal model recapitulating this age-associated decline in bone healing and identified increased cellular senescence caused by a systemic and local proinflammatory environment as the major contributor to the decline in skeletal stem/progenitor cell (SSPC) number and function. Decoupling age-associated systemic inflammation from chronological aging by using transgenic Nfkb1KO mice, we determined that the elevated inflammatory environment, and not chronological age, was responsible for the decrease in SSPC number and function. By using a pharmacological approach inhibiting NF-κB activation, we demonstrate a functional rejuvenation of aged SSPCs with decreased senescence, increased SSPC number, and increased osteogenic function. Unbiased, whole-genome RNA sequencing confirmed the reversal of the aging phenotype. Finally, in an ectopic model of bone healing, we demonstrate a functional restoration of regenerative potential in aged SSPCs. These data identify aging-associated inflammation as the cause of SSPC dysfunction and provide mechanistic insights into its reversal.


Subject(s)
Aging/metabolism , Fracture Healing , Fractures, Bone/metabolism , Osteogenesis , Stem Cells/metabolism , Aging/genetics , Aging/pathology , Animals , Female , Fractures, Bone/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Stem Cells/pathology
2.
Immunity ; 37(2): 339-50, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22921121

ABSTRACT

Little is known about the maintenance of intestinal stem cells (ISCs) and progenitors during immune-mediated tissue damage or about the susceptibility of transplant recipients to tissue damage mediated by the donor immune system during graft versus host disease (GVHD). We demonstrate here that deficiency of recipient-derived IL-22 increased acute GVHD tissue damage and mortality, that ISCs were eliminated during GVHD, and that ISCs as well as their downstream progenitors expressed the IL-22 receptor. Intestinal IL-22 was produced after bone marrow transplant by IL-23-responsive innate lymphoid cells (ILCs) from the transplant recipients, and intestinal IL-22 increased in response to pretransplant conditioning. However, ILC frequency and IL-22 amounts were decreased by GVHD. Recipient IL-22 deficiency led to increased crypt apoptosis, depletion of ISCs, and loss of epithelial integrity. Our findings reveal IL-22 as a critical regulator of tissue sensitivity to GVHD and a protective factor for ISCs during inflammatory intestinal damage.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Interleukins/metabolism , Intestine, Small/immunology , Stem Cells/metabolism , Animals , Bone Marrow Transplantation/adverse effects , Disease Models, Animal , Flow Cytometry , Graft vs Host Disease/mortality , Immunohistochemistry , Interleukin-23/metabolism , Interleukins/genetics , Interleukins/immunology , Intestine, Small/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/metabolism , Interleukin-22
3.
Blood ; 132(26): 2763-2774, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30381375

ABSTRACT

Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a ubiquitously expressed transcription factor that is well known for its role in regulating the cellular redox pathway. Although there is mounting evidence suggesting a critical role for Nrf2 in hematopoietic stem cells and innate leukocytes, little is known about its involvement in T-cell biology. In this study, we identified a novel role for Nrf2 in regulating alloreactive T-cell function during allogeneic hematopoietic cell transplantation (allo-HCT). We observed increased expression and nuclear translocation of Nrf2 upon T-cell activation in vitro, especially in CD4+ donor T cells after allo-HCT. Allo-HCT recipients of Nrf2 -/- donor T cells had significantly less acute graft-versus-host disease (GVHD)-induced mortality, morbidity, and pathology. This reduction in GVHD was associated with the persistence of Helios+ donor regulatory T cells in the allograft, as well as defective upregulation of the gut-homing receptor LPAM-1 on alloreactive CD8+ T cells. Additionally, Nrf2 -/- donor CD8+ T cells demonstrated intact cytotoxicity against allogeneic target cells. Tumor-bearing allo-HCT recipients of Nrf2 -/- donor T cells had overall improved survival as a result of preserved graft-versus-tumor activity and reduced GVHD activity. Our findings characterized a previously unrecognized role for Nrf2 in T-cell function, as well as revealed a novel therapeutic target to improve the outcomes of allo-HCT.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Lymphocyte Activation , NF-E2-Related Factor 2/immunology , Neoplasms, Experimental/immunology , Acute Disease , Allografts , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy
4.
Blood ; 118(2): 446-55, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21596854

ABSTRACT

IL-21 is a proinflammatory cytokine produced by Th17 cells. Abrogation of IL-21 signaling has recently been shown to reduce GVHD while retaining graft-versus-leukemia/lymphoma (GVL) responses. However, the mechanisms by which IL-21 may lead to a separation of GVHD and GVL remain incompletely understood. In a murine MHC-mismatched BM transplantation model, we observed that IL-21 receptor knockout (IL-21R KO) donor T cells mediate decreased systemic and gastrointestinal GVHD in recipients of a transplant. This reduction in GVHD was associated with expansion of transplanted donor regulatory T cells and with tissue-specific modulation of Th-cell function. IL-21R KO and wild-type donor T cells showed equivalent alloactivation, but IL-21R KO T cells showed decreased infiltration and inflammatory cytokine production within the mesenteric lymph nodes. However, Th-cell cytokine production was maintained peripherally, and IL-21R KO T cells mediated equivalent immunity against A20 and P815 hematopoietic tumors. In summary, abrogation of IL-21 signaling in donor T cells leads to tissue-specific modulation of immunity, such that gastrointestinal GVHD is reduced, but peripheral T-cell function and GVL capacity are retained. IL-21 is thus an exciting target for therapeutic intervention and improvement of clinical transplantation outcomes.


Subject(s)
Graft vs Host Disease/genetics , Graft vs Leukemia Effect/genetics , Immunity, Innate/genetics , Interleukins/physiology , T-Lymphocytes/metabolism , Tissue Donors , Animals , Gene Knockdown Techniques , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Humans , Immunity, Innate/physiology , Interleukin-21 Receptor alpha Subunit/genetics , Interleukin-21 Receptor alpha Subunit/metabolism , Interleukin-21 Receptor alpha Subunit/physiology , Interleukins/genetics , Interleukins/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/immunology , Signal Transduction/genetics , T-Lymphocytes/physiology , Transplantation Immunology
5.
Blood ; 116(11): e18-25, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20511541

ABSTRACT

We have developed a dual bioluminescent reporter system allowing noninvasive, concomitant imaging of T-cell trafficking, expansion, and activation of nuclear factor of activated T cells (NFAT) in vivo. NFAT activation plays an important role in T-cell activation and T-cell development. Therefore we used this system to determine spatial-temporal activation patterns of (1) proliferating T lymphocytes during graft-versus-host disease (GVHD) and (2) T-cell precursors during T-cell development after allogeneic hematopoietic stem cell transplantation (HSCT). In the first days after HSCT, donor T cells migrated to the peripheral lymph nodes and the intestines, whereas the NFAT activation was dominant in the intestines, suggesting an important role for the intestines in the early stages of alloactivation during development of GVHD. After adoptive transfer of in vitro-derived T-cell receptor (TCR) H-Y transgenic T-cell precursors into B6 (H-2(b)) hosts of both sexes, NFAT signaling and development into CD4(+) or CD8(+) single-positive cells could only be detected in the thymus of female recipients indicating either absence of positive selection or prompt depletion of double-positive thymocytes in the male recipients. Because NFAT plays an important role in a wide range of cell types, our system could provide new insights into a variety of biologic processes.


Subject(s)
Cell Movement , Cell Proliferation , Precursor Cells, T-Lymphoid/cytology , T-Lymphocytes/cytology , 3T3 Cells , Adoptive Transfer , Animals , Cell Line , Cell Line, Tumor , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Jurkat Cells , Lentivirus/genetics , Luciferases/genetics , Luciferases/metabolism , Luminescence , Luminescent Measurements/methods , Lymphocyte Activation , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , Precursor Cells, T-Lymphoid/metabolism , Precursor Cells, T-Lymphoid/transplantation , Promoter Regions, Genetic/genetics , T-Lymphocytes/metabolism
6.
Blood ; 113(7): 1574-80, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19011222

ABSTRACT

Keratinocyte growth factor (KGF), which is given exogenously to allogeneic bone marrow transplantation (allo-BMT) recipients, supports thymic epithelial cells and increases thymic output of naive T cells. Here, we demonstrate that this improved T-cell reconstitution leads to enhanced responses to DNA plasmid tumor vaccination. Tumor-bearing mice treated with KGF and DNA vaccination have improved long-term survival and decreased tumor burden after allo-BMT. When assayed before vaccination, KGF-treated allo-BMT recipients have increased numbers of peripheral T cells, including CD8(+) T cells with vaccine-recognition potential. In response to vaccination, KGF-treated allo-BMT recipients, compared with control subjects, generate increased numbers of tumor-specific CD8(+) cells, as well as increased numbers of CD8(+) cells producing interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). We also found unanticipated benefits to antitumor immunity with the administration of KGF. KGF-treated allo-BMT recipients have an improved ratio of T effector cells to regulatory T cells, a larger fraction of effector cells that display a central memory phenotype, and effector cells that are derived from a broader T-cell-receptor repertoire. In conclusion, our data suggest that KGF can function as a potent vaccine adjuvant after allo-BMT through its effects on posttransplantation T-cell reconstitution.


Subject(s)
Bone Marrow Transplantation/immunology , Cancer Vaccines/immunology , Fibroblast Growth Factor 7/pharmacology , Thymus Gland/drug effects , Vaccines, DNA/immunology , Animals , Bone Marrow Transplantation/methods , Bone Marrow Transplantation/mortality , CD4 Antigens/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Division/drug effects , Cell Division/immunology , Female , Forkhead Transcription Factors/metabolism , Immunologic Memory/drug effects , Immunologic Memory/immunology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Plasmids , Survival Rate , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Transplantation Chimera , Transplantation, Homologous
7.
J Bone Miner Res ; 35(5): 932-941, 2020 05.
Article in English | MEDLINE | ID: mdl-31881108

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are one of the most commonly prescribed antidepressants worldwide and recent data show significant impairment of fracture healing after treatment with the SSRI fluoxetine in mice. Here, we provide evidence that the negative effects of SSRIs can be overcome by administration of the beta-blocker propranolol at the time of fracture. First, in vitro experiments established that propranolol does not affect osteogenic differentiation. We then used a murine model of intramembranous ossification to study the potential rescue effect of propranolol on SSRI-induced impaired fracture healing. Micro-CT analysis revealed that fluoxetine treatment resulted in a smaller bony regenerate and that this decrease in bone formation can be overcome by co-treatment with propranolol. We then tested this in a clinically relevant model of endochondral ossification. Fluoxetine-treated mice with a femur fracture were treated with propranolol initiated at the time of fracture, and a battery of analyses demonstrated a reversal of the detrimental effect of fluoxetine on fracture healing in response to propranolol treatment. These experiments show for the first time to our knowledge that the negative effects of SSRIs on fracture healing can be overcome by co-treatment with a beta-blocker. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Femoral Fractures , Selective Serotonin Reuptake Inhibitors , Animals , Fluoxetine/pharmacology , Fracture Healing , Mice , Osteogenesis , Propranolol/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology
8.
Cancer Res ; 73(15): 4687-96, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23733752

ABSTRACT

Efforts to limit GVHD mediated by alloreactive donor T cells after allogeneic bone marrow transplantation are limited by a concomitant decrease in graft-versus-tumor (GVT) activity and increased possibilities of tumor relapse. Using a novel approach, we adoptively transferred conventional T cells expressing the transcription factor promyelocytic leukemia zinc finger (PLZF), which confers effector properties resembling invariant natural killer T cells, such as copious production of cytokines under suboptimal stimulation. PLZF expression in T-cell allografts attenuates expansion of alloreactive T cells, leading to lower GVHD. Intact alloreactivity-driven antitumor cytokine responses result in preserved GVT effects, leading to improved survival. Our findings suggest that therapy with PLZF-overexpressing T cells would result in overall improved outcomes due to less GVHD and intact GVT effects.


Subject(s)
Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Kruppel-Like Transcription Factors/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Bone Marrow Transplantation , Flow Cytometry , Graft vs Host Disease/immunology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/therapy , Promyelocytic Leukemia Zinc Finger Protein , T-Lymphocytes/transplantation , Transplantation, Homologous
9.
J Clin Invest ; 123(6): 2654-62, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23676461

ABSTRACT

Current strategies to suppress graft-versus-host disease (GVHD) also compromise graft-versus-tumor (GVT) responses. Furthermore, most experimental strategies to separate GVHD and GVT responses merely spare GVT function without actually enhancing it. We have previously shown that endogenously expressed TNF-related apoptosis-inducing ligand (TRAIL) is required for optimal GVT activity against certain malignancies in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). In order to model a donor-derived cellular therapy, we genetically engineered T cells to overexpress TRAIL and adoptively transferred donor-type unsorted TRAIL+ T cells into mouse models of allo-HSCT. We found that murine TRAIL+ T cells induced apoptosis of alloreactive T cells, thereby reducing GVHD in a DR5-dependent manner. Furthermore, murine TRAIL+ T cells mediated enhanced in vitro and in vivo antilymphoma GVT response. Moreover, human TRAIL+ T cells mediated enhanced in vitro cytotoxicity against both human leukemia cell lines and against freshly isolated chronic lymphocytic leukemia (CLL) cells. Finally, as a model of off-the-shelf, donor-unrestricted antitumor cellular therapy, in vitro-generated TRAIL+ precursor T cells from third-party donors also mediated enhanced GVT response in the absence of GVHD. These data indicate that TRAIL-overexpressing donor T cells could potentially enhance the curative potential of allo-HSCT by increasing GVT response and suppressing GVHD.


Subject(s)
Graft Rejection/prevention & control , Graft vs Host Disease/immunology , T-Lymphocytes/transplantation , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Adoptive Transfer , Animals , Antigen-Presenting Cells , Cell Line, Tumor , Cytotoxicity, Immunologic , Graft Rejection/immunology , Graft vs Host Disease/prevention & control , HEK293 Cells , Humans , Immunotherapy, Adoptive , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , T-Lymphocytes/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/physiology
10.
J Clin Invest ; 120(1): 343-56, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19955659

ABSTRACT

Thymic graft-versus-host disease (tGVHD) can contribute to profound T cell deficiency and repertoire restriction after allogeneic BM transplantation (allo-BMT). However, the cellular mechanisms of tGVHD and interactions between donor alloreactive T cells and thymic tissues remain poorly defined. Using clinically relevant murine allo-BMT models, we show here that even minimal numbers of donor alloreactive T cells, which caused mild nonlethal systemic graft-versus-host disease, were sufficient to damage the thymus, delay T lineage reconstitution, and compromise donor peripheral T cell function. Furthermore, to mediate tGVHD, donor alloreactive T cells required trafficking molecules, including CCR9, L selectin, P selectin glycoprotein ligand-1, the integrin subunits alphaE and beta7, CCR2, and CXCR3, and costimulatory/inhibitory molecules, including Ox40 and carcinoembryonic antigen-associated cell adhesion molecule 1. We found that radiation in BMT conditioning regimens upregulated expression of the death receptors Fas and death receptor 5 (DR5) on thymic stromal cells (especially epithelium), while decreasing expression of the antiapoptotic regulator cellular caspase-8-like inhibitory protein. Donor alloreactive T cells used the cognate proteins FasL and TNF-related apoptosis-inducing ligand (TRAIL) (but not TNF or perforin) to mediate tGVHD, thereby damaging thymic stromal cells, cytoarchitecture, and function. Strategies that interfere with Fas/FasL and TRAIL/DR5 interactions may therefore represent a means to attenuate tGVHD and improve T cell reconstitution in allo-BMT recipients.


Subject(s)
Bone Marrow Transplantation/immunology , Fas Ligand Protein/physiology , Graft vs Host Disease/etiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Thymus Gland/immunology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/analysis , Cell Movement , Graft vs Host Disease/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, OX40/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , Stromal Cells/physiology , T-Lymphocytes/immunology , Thymus Gland/cytology , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL