Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Blood ; 141(14): 1755-1767, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36574344

ABSTRACT

CD4+FOXP3+ regulatory T cells (Tregs) have demonstrated efficacy in the prevention and treatment of graft-versus-host disease (GVHD). Preclinical and clinical studies indicate that Tregs are able to protect from GVHD without interfering with the graft-versus-tumor (GVT) effect of hematopoietic cell transplantation (HCT), although the underlying molecular mechanisms are largely unknown. To elucidate Treg suppressive function during in vivo suppression of acute GVHD, we performed paired T-cell receptor (TCRα and ΤCRß genes) repertoire sequencing and RNA sequencing analysis on conventional T cells (Tcons) and Tregs before and after transplantation in a major histocompatibility complex -mismatched mouse model of HCT. We show that both Tregs and Tcons underwent clonal restriction, and Tregs did not interfere with the activation of alloreactive Tcon clones and the breadth of their TCR repertoire but markedly suppressed their expansion. Transcriptomic analysis revealed that Tregs predominantly affected the transcriptome of CD4 Tcons and, to a lesser extent, that of CD8 Tcons, thus modulating the transcription of genes encoding pro- and anti-inflammatory molecules as well as enzymes involved in metabolic processes, inducing a switch from glycolysis to oxidative phosphorylation. Finally, Tregs did not interfere with the induction of gene sets involved in the GVT effect. Our results shed light onto the mechanisms of acute GVHD suppression by Tregs and will support the clinical translation of this immunoregulatory approach.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Mice , T-Lymphocytes, Regulatory/pathology , Transcriptome , Graft vs Host Disease/genetics , Graft vs Host Disease/prevention & control , Graft vs Host Disease/pathology , Proteins/genetics
2.
Blood ; 141(11): 1337-1352, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36564052

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative option for patients with hematological disorders and bone marrow (BM) failure syndromes. Graft-versus-host disease (GVHD) remains a leading cause of morbidity posttransplant. Regulatory T cell (Treg) therapies are efficacious in ameliorating GVHD but limited by variable suppressive capacities and the need for a high therapeutic dose. Here, we sought to expand Treg in vivo by expressing an orthogonal interleukin 2 receptor ß (oIL-2Rß) that would selectively interact with oIL-2 cytokine and not wild-type (WT) IL-2. To test whether the orthogonal system would preferentially drive donor Treg expansion, we used a murine major histocompatibility complex-disparate GVHD model of lethally irradiated BALB/c mice given T cell-depleted BM from C57BL/6 (B6) mice alone or together with B6Foxp3+GFP+ Treg or oIL-2Rß-transduced Treg at low cell numbers that typically do not control GVHD with WT Treg. On day 2, B6 activated T cells (Tcons) were injected to induce GVHD. Recipients were treated with phosphate-buffered saline (PBS) or oIL-2 daily for 14 days, then 3 times weekly for an additional 14 days. Mice treated with oIL-2Rß Treg and oIL-2 compared with those treated with PBS had enhanced GVHD survival, in vivo selective expansion of Tregs, and greater suppression of Tcon expansion in secondary lymphoid organs and intestines. Importantly, oIL-2Rß Treg maintained graft-versus-tumor (GVT) responses in 2 distinct tumor models (A20 and MLL-AF9). These data demonstrate a novel approach to enhance the efficacy of Treg therapy in allo-HSCT using an oIL-2/oIL-2Rß system that allows for selective in vivo expansion of Treg leading to GVHD protection and GVT maintenance.


Subject(s)
Graft vs Host Disease , Neoplasms , Animals , Mice , T-Lymphocytes, Regulatory , Interleukin-2/pharmacology , Mice, Inbred C57BL , Bone Marrow Transplantation , Cytokines , Graft vs Host Disease/prevention & control , Mice, Inbred BALB C
3.
Blood ; 138(10): 858-870, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34036317

ABSTRACT

Invariant natural killer T (iNKT) cells are a T-cell subset with potent immunomodulatory properties. Experimental evidence in mice and observational studies in humans indicate that iNKT cells have antitumor potential as well as the ability to suppress acute and chronic graft-versus-host-disease (GVHD). Murine iNKT cells differentiate during thymic development into iNKT1, iNKT2, and iNKT17 sublineages, which differ transcriptomically and epigenomically and have subset-specific developmental requirements. Whether distinct iNKT sublineages also differ in their antitumor effect and their ability to suppress GVHD is currently unknown. In this work, we generated highly purified murine iNKT sublineages, characterized their transcriptomic and epigenomic landscape, and assessed specific functions. We show that iNKT2 and iNKT17, but not iNKT1, cells efficiently suppress T-cell activation in vitro and mitigate murine acute GVHD in vivo. Conversely, we show that iNKT1 cells display the highest antitumor activity against murine B-cell lymphoma cells both in vitro and in vivo. Thus, we report for the first time that iNKT sublineages have distinct and different functions, with iNKT1 cells having the highest antitumor activity and iNKT2 and iNKT17 cells having immune-regulatory properties. These results have important implications for the translation of iNKT cell therapies to the clinic for cancer immunotherapy as well as for the prevention and treatment of GVHD.


Subject(s)
Graft vs Host Disease , Graft vs Tumor Effect/immunology , Lymphocyte Activation , Lymphoma, B-Cell , Natural Killer T-Cells/immunology , Neoplasms, Experimental , Animals , Epigenomics , Female , Gene Expression Profiling , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Male , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy
4.
Am J Transplant ; 22(12): 3061-3068, 2022 12.
Article in English | MEDLINE | ID: mdl-36031344

ABSTRACT

Clinical trials utilizing regulatory T cell (Treg) therapy in organ transplantation have shown promising results, however, the choice of a standard immunosuppressive regimen is still controversial. Calcineurin inhibitors (CNIs) are one of the most common immunosuppressants for organ transplantation, although they may negatively affect Tregs by inhibiting IL-2 production by conventional T cells. As a strategy to replace IL-2 signaling selectively in Tregs, we have introduced an engineered orthogonal IL-2 (ortho IL-2) cytokine/cytokine receptor (R) pair that specifically binds with each other but does not bind with their wild-type counterparts. Murine Tregs were isolated from recipients and retrovirally transduced with ortho IL-2Rß during ex vivo expansion. Transduced Tregs (ortho Tregs) were transferred into recipient mice in a mixed hematopoietic chimerism model with tacrolimus administration. Ortho IL-2 treatment significantly increased the ortho IL-2Rß(+) Treg population in the presence of tacrolimus without stimulating other T cell subsets. All the mice treated with tacrolimus plus ortho IL-2 achieved heart allograft tolerance, even after tacrolimus cessation, whereas those receiving tacrolimus treatment alone did not. These data demonstrate that Treg therapy can be adopted into a CNI-based regimen by utilizing cytokine receptor engineering.


Subject(s)
Calcineurin Inhibitors , Tacrolimus , Mice , Animals , Calcineurin Inhibitors/pharmacology , Tacrolimus/therapeutic use , T-Lymphocytes, Regulatory , Interleukin-2/metabolism , Receptors, Interleukin-2 , Graft Survival , Immunosuppressive Agents/therapeutic use
5.
Blood ; 129(23): 3121-3125, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28416503

ABSTRACT

Chronic graft-versus-host-disease (cGVHD) can cause multiorgan system disease, typically with autoimmune-like features, resulting in high mortality and morbidity caused by treatment limitations. Invariant natural killer T cells (iNKTs), a small population characterized by expression of a semi-invariant T-cell receptor, rapidly produce copious amounts of diverse cytokines on activation that exert potent immune regulatory function. Here, we show that iNKTs are significantly reduced in a cGVHD murine model that recapitulates several aspects of autoimmunity and organ fibrosis observed in patients with cGVHD. Low iNKT infused doses effectively prevented and, importantly, reversed established cGVHD, as did third-party iNKTs. iNKTs suppressed the autoimmune response by reducing the germinal center (GC) reaction, which was associated with an increase in total Tregs and follicular Tregs (Tfr) that control the GC reaction, along with pathogenic antibody production. Treg depletion during iNKT infusions completely abolished iNKT efficacy in treating cGVHD. iNKT cell interleukin 4 production and GC migration were critical to cGVHD reversal. In vivo stimulation of iNKT cells by α-galactosyl-ceramide was effective in both preventing and treating cGVHD. Together, this study demonstrates iNKT deficiency in cGVHD mice and highlights the key role of iNKTs in regulating cGVHD pathogenesis and as a potentially novel prophylactic and therapeutic option for patients with cGVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Graft vs Host Disease/therapy , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation/adverse effects , Diphtheria Toxin/toxicity , Disease Models, Animal , Germinal Center/immunology , Graft vs Host Disease/immunology , Humans , Immunosuppression Therapy/methods , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/transplantation , T-Lymphocytes, Regulatory/drug effects , Tissue Donors
6.
Blood ; 128(6): 866-71, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27365424

ABSTRACT

CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) have been shown to effectively prevent graft-versus-host disease (GVHD) when adoptively transferred in murine models of hematopoietic cell transplantation and in phase 1/2 clinical trials. Critical limitations to Treg clinical application are the paucity of cells and limited knowledge of the mechanisms of in vivo function. We hypothesized that inflammatory conditions in GVHD modify Treg characteristics and activity. We found that peripheral blood of recipient animals during acute GVHD (aGVHD) induces Treg activation and enhances their function. The serum contains high levels of tumor necrosis factor-α (TNF-α) that selectively activates Tregs without impacting CD4(+)FoxP3(-) T cells. TNF-α priming induces Treg in vivo proliferation, whereas it limits the ability of CD4 and CD8 conventional T cells (Tcons) to proliferate and induce GVHD. TNF-α-primed Tregs prolong animal survival as compared with unprimed Tregs when used at an unfavorable Treg:Tcon ratio, demonstrating enhanced in vivo efficacy of TNF-α-primed Tregs. Because TNF-α is produced by several immune cells during inflammation, our work elucidates aspects of the physiologic mechanisms of Treg function. Furthermore, TNF-α priming of Tregs provides a new tool to optimize Treg cellular therapies for GVHD prevention and treatment.


Subject(s)
Adoptive Transfer/methods , CD4 Antigens/immunology , Forkhead Transcription Factors/immunology , Graft vs Host Disease/therapy , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/therapeutic use , Animals , CD4 Antigens/analysis , Cells, Cultured , Forkhead Transcription Factors/analysis , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Lymphocyte Activation , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/transplantation , Tumor Necrosis Factor-alpha/immunology
7.
Blood ; 128(24): 2846-2858, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27760760

ABSTRACT

CD4+Foxp3+ regulatory T cells (Treg) are a subpopulation of T cells, which regulate the immune system and enhance immune tolerance after transplantation. Donor-derived Treg prevent the development of lethal acute graft-versus-host disease (GVHD) in murine models of allogeneic hematopoietic stem cell transplantation. We recently demonstrated that a single treatment of the agonistic antibody to DR3 (death receptor 3, αDR3) to donor mice resulted in the expansion of donor-derived Treg and prevented acute GVHD, although the precise role of DR3 signaling in GVHD has not been elucidated. In this study, we comprehensively analyzed the immunophenotype of Treg after DR3 signal activation, demonstrating that DR3-activated Treg (DR3-Treg) had an activated/mature phenotype. Furthermore, the CD25+Foxp3+ subpopulation in DR3-Treg showed stronger suppressive effects in vivo. Prophylactic treatment of αDR3 to recipient mice expanded recipient-derived Treg and reduced the severity of GVHD, whereas DR3 activation in mice with ongoing GVHD further promoted donor T-cell activation/proliferation. These data suggest that the function of DR3 signaling was highly dependent on the activation status of the T cells. In conclusion, our data demonstrated that DR3 signaling affects the function of Treg and T-cell activation after alloantigen exposure in a time-dependent manner. These observations provide important information for future clinical testing using human DR3 signal modulation and highlight the critical effect of the state of T-cell activation on clinical outcomes after activation of DR3.


Subject(s)
Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction , Acute Disease , Animals , Gene Expression Profiling , Graft vs Host Disease/genetics , Graft vs Host Disease/prevention & control , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/metabolism , Isoantigens/immunology , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
8.
Proc Natl Acad Sci U S A ; 112(49): 15016-23, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26621719

ABSTRACT

Amyloid fibrils composed of peptides as short as six amino acids are therapeutic in experimental autoimmune encephalomyelitis (EAE), reducing paralysis and inflammation, while inducing several pathways of immune suppression. Intraperitoneal injection of fibrils selectively activates B-1a lymphocytes and two populations of resident macrophages (MΦs), increasing IL-10 production, and triggering their exodus from the peritoneum. The importance of IL-10-producing B-1a cells in this effective therapy was established in loss-of-function experiments where neither B-cell-deficient (µMT) nor IL10(-/-) mice with EAE responded to the fibrils. In gain-of-function experiments, B-1a cells, adoptively transferred to µMT mice with EAE, restored their therapeutic efficacy when Amylin 28-33 was administered. Stimulation of adoptively transferred bioluminescent MΦs and B-1a cells by amyloid fibrils resulted in rapid (within 60 min of injection) trafficking of both cell types to draining lymph nodes. Analysis of gene expression indicated that the fibrils activated the CD40/B-cell receptor pathway in B-1a cells and induced a set of immune-suppressive cell-surface proteins, including BTLA, IRF4, and Siglec G. Collectively, these data indicate that the fibrils activate B-1a cells and F4/80(+) MΦs, resulting in their migration to the lymph nodes, where IL-10 and cell-surface receptors associated with immune-suppression limit antigen presentation and T-cell activation. These mechanisms culminate in reduction of paralytic signs of EAE.


Subject(s)
Amyloid/pharmacology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Lymphocyte Activation/drug effects , Adoptive Transfer , Amyloid/metabolism , Amyloid/therapeutic use , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Endocytosis , Female , Interleukin-10/physiology , Islet Amyloid Polypeptide/pharmacology , Mice , Mice, Inbred C57BL
9.
Blood ; 125(22): 3491-500, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25795920

ABSTRACT

Graft-versus-host disease (GVHD) is driven by extensive activation and proliferation of alloreactive donor T cells causing significant morbidity and mortality following allogeneic hematopoietic cell transplantation (HCT). Invariant natural killer T (iNKT) cells are a potent immunoregulatory T-cell subset in both humans and mice. Here, we explored the role of adoptively transferred third-party CD4(+) iNKT cells for protection from lethal GVHD in a murine model of allogeneic HCT across major histocompatibility barriers. We found that low numbers of CD4(+) iNKT cells from third-party mice resulted in a significant survival benefit with retained graft-versus-tumor effects. In vivo expansion of alloreactive T cells was diminished while displaying a T helper cell 2-biased phenotype. Notably, CD4(+) iNKT cells from third-party mice were as protective as CD4(+) iNKT cells from donor mice although third-party CD4(+) iNKT cells were rejected early after allogeneic HCT. Adoptive transfer of third-party CD4(+) iNKT cells resulted in a robust expansion of donor CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) that were required for protection from lethal GVHD. However, in vivo depletion of myeloid-derived suppressor cells abrogated both Treg expansion and protection from lethal GVHD. Despite the fact that iNKT cells are a rare cell population, the almost unlimited third-party availability and feasibility of in vitro expansion provide the basis for clinical translation.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/transplantation , Graft vs Host Disease/mortality , Graft vs Host Disease/prevention & control , Immunotherapy, Adoptive/methods , Killer Cells, Natural/physiology , Killer Cells, Natural/transplantation , Animals , Cell Proliferation , Cells, Cultured , Cytoprotection/immunology , Female , Graft vs Host Disease/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/physiology
10.
Blood ; 126(4): 546-57, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26063163

ABSTRACT

The paucity of regulatory T cells (Tregs) limits clinical translation to control aberrant immune reactions including graft-versus-host disease (GVHD). Recent studies showed that the agonistic antibody to DR3 (αDR3) expanded CD4(+)FoxP3(+) Tregs in vivo. We investigated whether treating donor mice with a single dose of αDR3 could alleviate acute GVHD in a MHC-mismatched bone marrow transplantation model. αDR3 induced selective proliferation of functional Tregs. CD4(+) T cells isolated from αDR3-treated mice contained higher numbers of Tregs and were less proliferative to allogeneic stimuli. In vivo GVHD studies confirmed that Tregs from αDR3-treated donors expanded robustly and higher frequencies of Tregs within donor CD4(+) T cells were maintained, resulting in improved survival. Conventional T cells derived from αDR3-treated donors showed reduced activation and proliferation. Serum levels of proinflammatory cytokines (IFNγ, IL-1ß, and TNFα) and infiltration of donor T cells into GVHD target tissues (gastrointestinal tract and liver) were decreased. T cells from αDR3-treated donors retained graft-vs-tumor (GVT) effects. In conclusion, a single dose of αDR3 alleviates acute GVHD while preserving GVT effects by selectively expanding and maintaining donor Tregs. This novel strategy will facilitate the clinical application of Treg-based therapies.


Subject(s)
Antibodies/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Lymphoma, B-Cell/prevention & control , Receptors, Tumor Necrosis Factor, Member 25/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow Transplantation , Cytokines/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Immunoglobulins/administration & dosage , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Fluorescence , Tissue Donors , Transplantation, Homologous , Tumor Cells, Cultured
11.
J Immunol ; 195(1): 347-55, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25994967

ABSTRACT

Adoptive transfer of freshly isolated natural occurring CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) prevents graft-versus-host disease (GVHD) in several animal models and following hematopoietic cell transplantation (HCT) in clinical trials. Donor-derived Treg have been mainly used, as they share the same MHC with CD4(+) and CD8(+) conventional T cells (Tcon) that are primarily responsible for GVHD. Third party-derived Treg are a promising alternative for cellular therapy, as they can be prepared in advance, screened for pathogens and activity, and banked. We explored MHC disparities between Treg and Tcon in HCT to evaluate the impact of different Treg populations in GVHD prevention and survival. Third-party Treg and donor Treg are equally suppressive in ex vivo assays, whereas both donor and third-party but not host Treg protect from GVHD in allogeneic HCT, with donor Treg being the most effective. In an MHC minor mismatched transplantation model (C57BL/6 → BALB/b), donor and third-party Treg were equally effective in controlling GVHD. Furthermore, using an in vivo Treg depletion mouse model, we found that Treg exert their main suppressive activity in the first 2 d after transplantation. Third-party Treg survive for a shorter period of time after adoptive transfer, but despite the shorter survival, they control Tcon proliferation in the early phases of HCT. These studies provide relevant insights on the mechanisms of Treg-mediated protection from GVHD and support for the use of third-party Treg in clinical trials.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Biomarkers/blood , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Forkhead Transcription Factors/blood , Gene Expression , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Histocompatibility Testing , Interleukin-2 Receptor alpha Subunit/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation , Tissue Donors , Transplantation, Homologous , Whole-Body Irradiation
12.
Blood ; 124(22): 3320-8, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25293774

ABSTRACT

Dysregulated donor T cells lead to destruction of host tissues resulting in graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (HCT). We investigated the impact of highly purified (>95%) donor CD4(+) invariant natural killer T (iNKT) cells on GVHD in a murine model of allogeneic HCT. We found that low doses of adoptively transferred donor CD4(+) iNKT cells protect from GVHD morbidity and mortality through an expansion of donor CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs). These Tregs express high levels of the Ikaros transcription factor Helios and expand from the Treg pool of the donor graft. Furthermore, CD4(+) iNKT cells preserve T-cell-mediated graft-versus-tumor effects. Our studies reveal new aspects of the cellular interplay between iNKT cells and Tregs in the context of tolerance induction after allogeneic HCT and set the stage for clinical translation.


Subject(s)
Adoptive Transfer , CD4-Positive T-Lymphocytes/physiology , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Natural Killer T-Cells/physiology , T-Lymphocytes, Regulatory/immunology , Animals , CD4 Lymphocyte Count , Cells, Cultured , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
13.
Blood ; 124(11): 1832-42, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25030062

ABSTRACT

Acute graft-versus-host disease (GVHD) is induced by alloreactivity of donor T cells toward host antigens presented on antigen-presenting cells (APCs). Apoptotic cells are capable of inducing tolerance by altering APC maturation. Apoptosis can be induced by extracorporeal photopheresis (ECP). We demonstrate that the use of ECP as a prophylaxis prior to conditioning significantly improves survival (P < .0001) after bone marrow transplantation (BMT) by inhibiting the initiation phase of acute GVHD in a murine BMT model. ECP-treated autologous splenocytes resulted in immune tolerance in the host, including reduced dendritic cell activation with decreased nuclear factor-κB engagement, increased regulatory T-cell (Treg) numbers with enhanced expression of cytolytic T lymphocyte-associated antigen 4, potentiating their suppressive function. The protective effect required host production of interleukin-10 and host Tregs. Conventional T cells that entered this tolerant environment experienced reduced proliferation, as well as a reduction of tissue homing and expression of activation markers. The induction of this tolerant state by ECP was obviated by cotreatment with lipopolysaccharide, suggesting that the inflammatory state of the recipient prior to treatment would play a role in potential clinical translation. The use of prophylactic ECP may provide an alternative and safe method for immunosuppression in the bone marrow transplant setting.


Subject(s)
Antigen-Presenting Cells/transplantation , Apoptosis , Graft vs Host Disease/prevention & control , Immune Tolerance , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Autografts , Bone Marrow Transplantation , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
14.
Blood ; 119(24): 5758-68, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22544698

ABSTRACT

Natural killer (NK) cells are potent anti-viral and antitumor "first responders" endowed with natural cytotoxicity and cytokine production capabilities. To date, attempts to translate these promising biologic functions through the adoptive transfer of NK cells for the treatment of cancer have been of limited benefit. Here we trace the fate of adoptively transferred murine NK cells and make the surprising observation that NK cells traffic to tumor sites yet fail to control tumor growth or improve survival. This dysfunction is related to a rapid down-regulation of activating receptor expression and loss of important effector functions. Loss of interferon (IFN)γ production occurs early after transfer, whereas loss of cytotoxicity progresses with homeostatic proliferation and tumor exposure. The dysfunctional phenotype is accompanied by down-regulation of the transcription factors Eomesodermin and T-bet, and can be partially reversed by the forced overexpression of Eomesodermin. These results provide the first demonstration of NK-cell exhaustion and suggest that the NK-cell first-response capability is intrinsically limited. Further, novel approaches may be required to circumvent the described dysfunctional phenotype.


Subject(s)
Adoptive Transfer , Antineoplastic Agents/immunology , Down-Regulation , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Box Domain Proteins/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Movement , Cell Proliferation , Homeostasis , Humans , Killer Cells, Natural/cytology , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Receptors, Immunologic/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
15.
Front Transplant ; 3: 1353803, 2024.
Article in English | MEDLINE | ID: mdl-38993780

ABSTRACT

Invariant Natural Killer T cell therapy is an emerging platform of immunotherapy for cancer treatment. This unique cell population is a promising candidate for cell therapy for cancer treatment because of its inherent cytotoxicity against CD1d positive cancers as well as its ability to induce host CD8 T cell cross priming. Substantial evidence supports that iNKT cells can modulate myelomonocytic populations in the tumor microenvironment to ameliorate immune dysregulation to antagonize tumor progression. iNKT cells can also protect from graft-versus-host disease (GVHD) through several mechanisms, including the expansion of regulatory T cells (Treg). Ultimately, iNKT cell-based therapy can retain antitumor activity while providing protection against GVHD simultaneously. Therefore, these biological properties render iNKT cells as a promising "off-the-shelf" therapy for diverse hematological malignancies and possible solid tumors. Further the introduction of a chimeric antigen recetor (CAR) can further target iNKT cells and enhance function. We foresee that improved vector design and other strategies such as combinatorial treatments with small molecules or immune checkpoint inhibitors could improve CAR iNKT in vivo persistence, functionality and leverage anti-tumor activity along with the abatement of iNKT cell dysfunction or exhaustion.

16.
J Leukoc Biol ; 115(2): 401-409, 2024 01 19.
Article in English | MEDLINE | ID: mdl-37742056

ABSTRACT

Invariant natural killer T cells are a rare, heterogeneous T-cell subset with cytotoxic and immunomodulatory properties. During thymic development, murine invariant natural killer T cells go through different maturation stages differentiating into distinct sublineages, namely, invariant natural killer T1, 2, and 17 cells. Recent reports indicate that invariant natural killer T2 cells display immature properties and give rise to other subsets, whereas invariant natural killer T1 cells seem to be terminally differentiated. Whether human invariant natural killer T cells follow a similar differentiation model is still unknown. To define the maturation stages and assess the sublineage commitment of human invariant natural killer T cells during thymic development, in this study, we performed single-cell RNA sequencing analysis on human Vα24+Vß11+ invariant natural killer T cells isolated from thymocytes. We show that these invariant natural killer T cells displayed heterogeneity, and our unsupervised analysis identified 5 clusters representing different maturation stages, from an immature profile with high expression of genes important for invariant natural killer T cell development and proliferation to a mature, fully differentiated profile with high levels of cytotoxic effector molecules. Evaluation of expression of sublineage-defining gene sets revealed mainly cells with an invariant natural killer T2 signature in the most immature cluster, whereas the more differentiated ones displayed an invariant natural killer T1 signature. Combined analysis with a publicly available single-cell RNA sequencing data set of human invariant natural killer T cells from peripheral blood suggested that the 2 main subsets exist both in thymus and in the periphery, while a third more immature one was restricted to the thymus. Our data point to the existence of different maturation stages of human thymic invariant natural killer T cells and provide evidence for sublineage commitment of invariant natural killer T cells in the human thymus.


Subject(s)
Natural Killer T-Cells , Humans , Mice , Animals , Natural Killer T-Cells/metabolism , Thymus Gland , Thymocytes , T-Lymphocyte Subsets , Cell Differentiation/genetics , Gene Expression Profiling
17.
Blood Adv ; 8(14): 3691-3704, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38701354

ABSTRACT

ABSTRACT: Despite therapeutic advancements, graft-versus-host disease (GVHD) is a major complication of hematopoietic stem cell transplantation (HSCT). In current models of GVHD, tissue injury induced by cytotoxic conditioning regimens, along with translocation of microbes expressing pathogen-associated molecular patterns, result in activation of host antigen-presenting cells (APCs) to stimulate alloreactive donor T lymphocytes. Recent studies have demonstrated that in many pathologic states, tissue injury results in the release of mitochondria from the cytoplasm to the extracellular space. We hypothesized that extracellular mitochondria, which are related to archaebacteria, could also trigger GVHD by stimulation of host APCs. We found that clinically relevant doses of radiation or busulfan induced extracellular release of mitochondria by various cell types, including cultured intestinal epithelial cells. Conditioning-mediated mitochondrial release was associated with mitochondrial damage and impaired quality control but did not affect the viability of the cells. Extracellular mitochondria directly stimulated host APCs to express higher levels of major histocompatibility complex II (MHC-II), costimulatory CD86, and proinflammatory cytokines, resulting in increased donor T-cell activation, and proliferation in mixed lymphocyte reactions. Analyses of plasma from both experimental mice and a cohort of children undergoing HSCT demonstrated that conditioning induced extracellular mitochondrial release in vivo. In mice undergoing MHC-mismatched HSCT, administration of purified syngeneic extracellular mitochondria increased host APC activation and exacerbated GVHD. Our data suggest that pre-HSCT conditioning results in extracellular release of damaged mitochondria, which increase alloreactivity and exacerbate GVHD. Therefore, decreasing the extracellular release of damaged mitochondria after conditioning could serve as a novel strategy for GVHD prevention.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Mitochondria , Transplantation Conditioning , Graft vs Host Disease/etiology , Graft vs Host Disease/metabolism , Animals , Hematopoietic Stem Cell Transplantation/adverse effects , Mitochondria/metabolism , Mice , Humans , Transplantation Conditioning/methods , Disease Models, Animal , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/immunology
18.
Biol Blood Marrow Transplant ; 19(11): 1557-65, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23921175

ABSTRACT

Regulatory T cell (Treg) immunotherapy is a promising strategy for the treatment of graft rejection responses and autoimmune disorders. Our and other laboratories have shown that the transfer of highly purified CD4(+)CD25(+)Foxp3(+) natural Treg can prevent lethal graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation across both major and minor histocompatibility barriers. However, recent evidence suggests that the Treg suppressive phenotype can become unstable, a phenomenon that can culminate in Treg conversion into IL-17-producing cells. We hypothesized that the intense proinflammatory signals released during an ongoing alloreaction might redirect a fraction of the transferred Treg to the Th17 cell fate, thereby losing immunosuppressive potential. We therefore sought to evaluate the impact of Il17 gene ablation on Treg stability and immunosuppressive capacity in a major MHC mismatch model. We show that although Il17 gene ablation results in a mildly enhanced Treg immunosuppressive ability in vitro, such improvement is not observed when IL-17-deficient Treg are used for GVHD suppression in vivo. Similarly, when we selectively blocked IL-1 signaling in Treg, that was shown to be necessary for Th17 conversion, we did not detect any improvement on Treg-mediated GVHD suppressive ability in vivo. Furthermore, upon ex vivo reisolation of transferred wild-type Treg, we detected little or no Treg-mediated IL-17 production upon GVHD induction. Our results indicate that blocking Th17 conversion does not affect the GVHD suppressive ability of highly purified natural Treg in vivo, suggesting that IL-17 targeting is not a valuable strategy to improve Treg immunotherapy after hematopoietic cell transplantation.


Subject(s)
Bone Marrow Transplantation/methods , Graft vs Host Disease/immunology , Immunotherapy, Adoptive/methods , Interleukin-17/genetics , T-Lymphocytes, Regulatory/transplantation , Animals , Graft vs Host Disease/genetics , Humans , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
19.
BMC Med ; 11: 134, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23692886

ABSTRACT

BACKGROUND: Acute graft-versus-host disease (aGVHD) poses a major limitation for broader therapeutic application of allogeneic hematopoietic cell transplantation (allo-HCT). Early diagnosis of aGVHD remains difficult and is based on clinical symptoms and histopathological evaluation of tissue biopsies. Thus, current aGVHD diagnosis is limited to patients with established disease manifestation. Therefore, for improved disease prevention it is important to develop predictive assays to identify patients at risk of developing aGVHD. Here we address whether insights into the timing of the aGVHD initiation and effector phases could allow for the detection of migrating alloreactive T cells before clinical aGVHD onset to permit for efficient therapeutic intervention. METHODS: Murine major histocompatibility complex (MHC) mismatched and minor histocompatibility antigen (miHAg) mismatched allo-HCT models were employed to assess the spatiotemporal distribution of donor T cells with flow cytometry and in vivo bioluminescence imaging (BLI). Daily flow cytometry analysis of peripheral blood mononuclear cells allowed us to identify migrating alloreactive T cells based on homing receptor expression profiles. RESULTS: We identified a time period of 2 weeks of massive alloreactive donor T cell migration in the blood after miHAg mismatch allo-HCT before clinical aGVHD symptoms appeared. Alloreactive T cells upregulated α4ß7 integrin and P-selectin ligand during this migration phase. Consequently, targeted preemptive treatment with rapamycin, starting at the earliest detection time of alloreactive donor T cells in the peripheral blood, prevented lethal aGVHD. CONCLUSIONS: Based on this data we propose a critical time frame prior to the onset of aGVHD symptoms to identify alloreactive T cells in the peripheral blood for timely and effective therapeutic intervention.


Subject(s)
Disease Models, Animal , Graft vs Host Disease/diagnosis , Graft vs Host Disease/surgery , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocyte Subsets/transplantation , Acute Disease , Animals , Blood Group Incompatibility/immunology , Female , Forecasting , Graft vs Host Disease/immunology , HLA Antigens/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocyte Subsets/immunology , Time Factors , Transplantation, Homologous , Treatment Outcome
20.
Blood ; 117(11): 3230-9, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21239702

ABSTRACT

Allogeneic hematopoietic cell transplantation can be curative in patients with leukemia and lymphoma. However, progressive growth of malignant cells, relapse after transplantation, and graft-versus-host disease (GVHD) remain important problems. The goal of the current murine study was to select a freshly isolated donor T-cell subset for infusion that separates antilymphoma activity from GVHD, and to determine whether the selected subset could effectively prevent or treat progressive growth of a naturally occurring B-cell lymphoma (BCL(1)) without GVHD after recipients were given T cell-depleted bone marrow transplantations from major histocompatibility complex-mismatched donors. Lethal GVHD was observed when total T cells, naive CD4(+) T cells, or naive CD8(+) T cells were used. Memory CD4(+)CD44(hi) and CD8(+)CD44(hi) T cells containing both central and effector memory cells did not induce lethal GVHD, but only memory CD8(+) T cells had potent antilymphoma activity and promoted complete chimerism. Infusion of CD8(+) memory T cells after transplantation was able to eradicate the BCL(1) lymphoma even after progressive growth without inducing severe GVHD. In conclusion, the memory CD8(+) T-cell subset separated graft antilymphoma activity from GVHD more effectively than naive T cells, memory CD4(+) T cells, or memory total T cells.


Subject(s)
Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Hyaluronan Receptors/metabolism , Immunologic Memory/immunology , Lymphoma/immunology , Animals , Body Weight , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Division , Cell Line, Tumor , Disease Progression , Graft vs Host Disease/complications , Immunization , Lymphoma/complications , Lymphoma/therapy , Mice , Organ Specificity/immunology , Phenotype , Spleen/cytology , Spleen/immunology , Survival Analysis , T-Lymphocyte Subsets/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL