198 results on '"Angus W. Thomson"'
Search Results
2. Regulatory T lymphocytes as a therapy for ischemic stroke
- Author
-
Miao Wang, Angus W. Thomson, Fang Yu, Rimi Hazra, Aditi Junagade, and Xiaoming Hu
- Subjects
Immunology ,Immunology and Allergy - Abstract
Unrestrained excessive inflammatory responses exacerbate ischemic brain injury and impede post-stroke brain recovery. CD4
- Published
- 2022
- Full Text
- View/download PDF
3. Tacrolimus before CTLA4Ig and rapamycin promotes vascularized composite allograft survival in MGH miniature swine
- Author
-
Tarek Y. Elgendy, Matthias Waldner, Wensheng Zhang, Deokyeol Y. Kim, Marta I. Minervini, Chiaki Komatsu, Yalcin Kulahci, Kia M. Washington, Vijay S. Gorantla, Mohamed B. Ezzelarab, Mario G. Solari, Angus W. Thomson, University of Zurich, and Thomson, Angus W
- Subjects
Sirolimus ,Graft Rejection ,Transplantation ,2403 Immunology ,Swine ,2747 Transplantation ,Graft Survival ,Immunology ,610 Medicine & health ,Tacrolimus ,Abatacept ,2723 Immunology and Allergy ,Animals ,Swine, Miniature ,Immunology and Allergy ,Composite Tissue Allografts ,10266 Clinic for Reconstructive Surgery ,Immunosuppressive Agents - Abstract
We evaluated the outcome of vertical rectus abdominus myocutaneous flap (VRAM) allotransplantation in a mini-pig model, using a combined co-stimulation blockade (Co-SB) and mechanistic target of rapamycin inhibition (mTORi)-based regimen, with or without preceding calcineurin inhibition (CNI).VRAM allotransplants were performed between SLA-mismatched MGH miniature swine. Group A (n = 2) was treated continuously with the mTOR inhibitor rapamycin from day -1 in combination with the Co-SB agent cytotoxic T lymphocyte antigen 4-Ig (CTLA4-Ig) from post-operative day (POD) 0. In group B (n = 3), animals received tacrolimus daily from POD 0 to POD 13, followed by rapamycin daily from POD 7 and CTLA4-Ig weekly from POD 7-28. Graft rejection was determined by Banff criteria and host cellular and humoral immunity monitored.In group A, allografts developed grade-I acute rejection by POD 2 and POD 7, and reached grade-IV by POD 17 and POD 20, respectively. By contrast, in group B, two allografts demonstrated grade-I rejection on POD 30 and grade-IV on POD 74, while the third exhibited grade-I rejection starting on POD 50, though this animal had to be euthanized on POD 58 due to Pneumocystis jirovecii infection. Time-to-event incidence of grade-I rejection was significantly lower in group A compared to group B. During the first 3 weeks post-transplant, no significant differences in anti-donor immunity were observed between the groups.A short course of CNI, followed by combined Co-SB and mTORi significantly delays acute rejection of VRAM allografts in SLA-mismatched miniature swine.
- Published
- 2022
- Full Text
- View/download PDF
4. A Comparison of Ex Vivo Expanded Human Regulatory T Cells Using Allogeneic Stimulated B Cells or Monocyte-Derived Dendritic Cells
- Author
-
Linda M. Lee, Hong Zhang, Karim Lee, Horace Liang, Alexander Merleev, Flavio Vincenti, Emanual Maverakis, Angus W. Thomson, and Qizhi Tang
- Subjects
0301 basic medicine ,Isoantigens ,regulatory T cell ,T-Lymphocytes ,Cell Culture Techniques ,T-Cell Antigen Receptor Specificity ,Lymphocyte Activation ,0302 clinical medicine ,Receptors ,Immunology and Allergy ,IL-2 receptor ,B-Lymphocytes ,Cultured ,Chemistry ,FOXP3 ,hemic and immune systems ,Regulatory ,Mixed ,Cell biology ,transplant tolerance ,Phenotype ,medicine.anatomical_structure ,Medical Microbiology ,Antigen ,030220 oncology & carcinogenesis ,Cytokines ,Treg therapy ,Regulatory T cell ,Cells ,1.1 Normal biological development and functioning ,Immunology ,chemical and pharmacologic phenomena ,Immunophenotyping ,03 medical and health sciences ,Immune system ,Underpinning research ,medicine ,Humans ,dendritic cells ,human ,Lymphocyte Culture Test ,CD86 ,Transplantation ,B cells ,Inflammatory and immune system ,immune regulation ,RC581-607 ,T-Cell ,030104 developmental biology ,Immunologic diseases. Allergy ,Biomarkers ,Ex vivo ,CD80 - Abstract
Alloreactive regulatory T cells (arTregs) are more potent than polyclonal Tregs at suppressing immune responses to transplant antigens. Human arTregs can be expanded with allogeneic CD40L-stimulated B cells (sBcs) or stimulated-matured monocyte-derived dendritic cells (sDCs). Here, we compared the expansion efficiency and properties of arTregs stimulated ex vivo using these two types of antigen-presenting cells. Compared to sBcs, sDCs stimulated Tregs to expand two times more in number. The superior expansion-inducing capacity of sDCs correlated with their higher expression of CD80, CD86, and T cell-attracting chemokines. sBc- and sDC-arTregs expressed comparable levels of FOXP3, HELIOS, CD25, CD27, and CD62L, demethylated FOXP3 enhancer and in vitro suppressive function. sBc- and sDCs-arTregs had similar gene expression profiles that were distinct from primary Tregs. sBc- and sDC-arTregs exhibited similar low frequencies of IFN-γ, IL-4, and IL-17A-producing cells, and the cytokine-producing arTregs expressed high levels of FOXP3. Almost all sBc- and sDC-arTregs expressed CXCR3, which may enable them traffic to inflammatory sites. Thus, sDCs-arTregs that expand more readily, are phenotypically similar to sBc-arTregs, supporting sDCs as a viable alternative for arTreg production for clinical evaluation.
- Published
- 2021
- Full Text
- View/download PDF
5. Dendritic Cell-Mediated Regulation of Liver Ischemia-Reperfusion Injury and Liver Transplant Rejection
- Author
-
Ryosuke Nakano, Lillian M. Tran, David A. Geller, Camila Macedo, Diana M. Metes, and Angus W. Thomson
- Subjects
0301 basic medicine ,Graft Rejection ,medicine.medical_treatment ,T cell ,Mini Review ,Immunology ,ischemia-reperfusion injury ,chemical and pharmacologic phenomena ,Liver transplantation ,Major histocompatibility complex ,liver ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Immunology and Allergy ,Animals ,Humans ,Transplantation, Homologous ,Innate immune system ,biology ,business.industry ,immune regulation ,Graft Survival ,Dendritic cell ,Dendritic Cells ,RC581-607 ,Liver Transplantation ,Transplantation ,transplant tolerance ,030104 developmental biology ,Immunosuppressive drug ,medicine.anatomical_structure ,surgical procedures, operative ,Reperfusion Injury ,biology.protein ,030211 gastroenterology & hepatology ,Transplantation Tolerance ,Immunologic diseases. Allergy ,business - Abstract
Liver allograft recipients are more likely to develop transplantation tolerance than those that receive other types of organ graft. Experimental studies suggest that immune cells and other non-parenchymal cells in the unique liver microenvironment play critical roles in promoting liver tolerogenicity. Of these, liver interstitial dendritic cells (DCs) are heterogeneous, innate immune cells that appear to play pivotal roles in the instigation, integration and regulation of inflammatory responses after liver transplantation. Interstitial liver DCs (recruited in situ or derived from circulating precursors) have been implicated in regulation of both ischemia/reperfusion injury (IRI) and anti-donor immunity. Thus, livers transplanted from mice constitutively lacking DCs into syngeneic, wild-type recipients, display increased tissue injury, indicating a protective role of liver-resident donor DCs against transplant IRI. Also, donor DC depletion before transplant prevents mouse spontaneous liver allograft tolerance across major histocompatibility complex (MHC) barriers. On the other hand, mouse liver graft-infiltrating host DCs that acquire donor MHC antigen via “cross-dressing”, regulate anti-donor T cell reactivity in association with exhaustion of graft-infiltrating T cells and promote allograft tolerance. In an early phase clinical trial, infusion of donor-derived regulatory DCs (DCreg) before living donor liver transplantation can induce alterations in host T cell populations that may be conducive to attenuation of anti-donor immune reactivity. We discuss the role of DCs in regulation of warm and liver transplant IRI and the induction of liver allograft tolerance. We also address design of cell therapies using DCreg to reduce the immunosuppressive drug burden and promote clinical liver allograft tolerance.
- Published
- 2021
6. Detection and Monitoring of Regulatory Immune Cells Following Their Adoptive Transfer in Organ Transplantation
- Author
-
Lillian M. Tran and Angus W. Thomson
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,regulatory T cell ,medicine.medical_specialty ,adoptive transfer ,Regulatory T cell ,Mini Review ,medicine.medical_treatment ,Immunology ,Cell- and Tissue-Based Therapy ,Context (language use) ,030230 surgery ,Bioinformatics ,T-Lymphocytes, Regulatory ,Organ transplantation ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Transplantation, Homologous ,Immunology and Allergy ,Myeloid Cells ,business.industry ,Mesenchymal Stem Cells ,Organ Transplantation ,Immunotherapy ,regulatory myeloid cell ,Transplantation ,Tolerance induction ,030104 developmental biology ,medicine.anatomical_structure ,cell tracking ,mesenchymal stromal cell ,Transplantation Tolerance ,cell therapy ,lcsh:RC581-607 ,business ,transplantation - Abstract
Application of cell-based immunotherapy in organ transplantation to minimize the burden of immunosuppressive medication and promote allograft tolerance has expanded significantly over the past decade. Adoptively transferred regulatory immune cells prolong allograft survival and transplant tolerance in pre-clinical models. Many cell products are currently under investigation in early phase human clinical trials designed to assess feasibility and safety. Despite rapid advances in manufacturing practices, defining the appropriate protocol that will optimize in vivo conditions for tolerance induction remains a major challenge and depends heavily on understanding the fate, biodistribution, functional stability and longevity of the cell product after administration. This review focuses on in vivo detection and monitoring of various regulatory immune cell types administered for allograft tolerance induction in both pre-clinical animal models and early human clinical trials. We discuss the current status of various non-invasive methods for tracking regulatory cell products in the context of organ transplantation and implications for enhanced understanding of the therapeutic potential of cell-based therapy in the broad context of control of immune-mediated inflammatory disorders.
- Published
- 2020
- Full Text
- View/download PDF
7. Donor plasmacytoid dendritic cells modulate effector and regulatory T cell responses in mouse spontaneous liver transplant tolerance
- Author
-
Toshimasa Nakao, Shinichiro Yokota, Yoshihiro Ono, David A. Geller, Angus W. Thomson, Shoko Kimura, Osamu Yoshida, Marta I. Minervini, and Ryosuke Nakano
- Subjects
Regulatory T cell ,T cell ,Spleen ,030230 surgery ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Living Donors ,Immunology and Allergy ,Animals ,Humans ,Pharmacology (medical) ,CD86 ,Transplantation ,Mice, Inbred C3H ,business.industry ,FOXP3 ,hemic and immune systems ,Dendritic cell ,Dendritic Cells ,Liver Transplantation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cancer research ,business ,CD8 ,CD80 - Abstract
We assessed the role of donor liver non-conventional plasmacytoid dendritic cells (pDCs) in spontaneous liver transplant tolerance in a fully MHC-mismatched (C57BL/6 (H2(b)) to C3H (H2(k))) mouse model. Compared with spleen pDCs, liver pDCs expressed higher levels of DNAX-activating protein of 12 kDa and its co-receptor, triggering receptor expressed by myeloid cells 2, and higher ratios of programed death ligand-1 (PD-L1):costimulatory CD80/CD86 in the steady state and after Toll-like receptor 9 ligation. Moreover, liver pDCs potently suppressed allogeneic CD4(+) and CD8(+) T cell proliferative responses. Survival of pDC-depleted livers was much poorer (median survival time: 25 days) than that of either untreated donor livers or pDC-depleted syngeneic donor livers that survived indefinitely. Numbers of forkhead box p3 (FoxP3)(+) regulatory T cells in grafts and mesenteric lymph nodes of mice given pDC-depleted allogeneic livers were reduced significantly compared with those in recipients of untreated livers. Graft-infiltrating CD8(+) T cells with an exhausted phenotype (programed cell death protein 1(+), T cell immunoglobulin and mucin domain-containing protein 3(+)) were also reduced in recipients of pDC-depleted livers. PD1-PD-L1 pathway blockade reversed the reduction in exhausted T cells. These novel observations link immunoregulatory functions of liver interstitial pDCs, alloreactive T cell exhaustion, and spontaneous liver transplant tolerance.
- Published
- 2020
8. Donor-derived regulatory dendritic cell infusion results in host cell cross-dressing and T cell subset changes in prospective living donor liver transplant recipients
- Author
-
Fadi G. Lakkis, Camila Macedo, Angus W. Thomson, Alan F. Zahorchak, Thalachallour Mohanakumar, Lillian M. Tran, Xinyan Gu, Ranjithkumar Ravichandran, Adriana Zeevi, Diana Metes, Beth Elinoff, Abhinav Humar, Mindi A. Styn, and Helong Dai
- Subjects
Adoptive cell transfer ,T cell ,030230 surgery ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,0302 clinical medicine ,T-Lymphocyte Subsets ,Living Donors ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Pharmacology (medical) ,Prospective Studies ,Interleukin-7 receptor ,CD86 ,Transplantation ,business.industry ,Graft Survival ,FOXP3 ,Dendritic cell ,Dendritic Cells ,Bandages ,Liver Transplantation ,medicine.anatomical_structure ,Immunology ,business ,CD8 - Abstract
Regulatory dendritic cells (DCreg) promote transplant tolerance following their adoptive transfer in experimental animals. We investigated the feasibility, safety, fate, and impact on host T cells of donor monocyte-derived DCreg infused into prospective, living donor liver transplant patients, 7 days before transplantation. The DCreg expressed a tolerogenic gene transcriptional profile, high cell surface programed death ligand-1 (PD-L1):CD86 ratios, high IL-10/no IL-12 productivity and poor ability to stimulate allogeneic T cell proliferation. Target DCreg doses (range 2.5-10 × 106 cells/kg) were achieved in all but 1 of 15 recipients, with no infusion reactions. Following DCreg infusion, transiently elevated levels of donor HLA and immunoregulatory PD-L1, CD39, and CD73 were detected in circulating small extracellular vesicles. At the same time, flow and advanced image stream analysis revealed intact DCreg and "cross-dressing" of host DCs in blood and lymph nodes. PD-L1 co-localization with donor HLA was observed at higher levels than with recipient HLA. Between DCreg infusion and transplantation, T-bethi Eomeshi memory CD8+ T cells decreased, whereas regulatory (CD25hi CD127- Foxp3+ ): T-bethi Eomeshi CD8+ T cell ratios increased. Thus, donor-derived DCreg infusion may induce systemic changes in host antigen-presenting cells and T cells potentially conducive to modulated anti-donor immune reactivity at the time of transplant.
- Published
- 2020
9. Treg cell-derived osteopontin promotes microglia-mediated white matter repair after ischemic stroke
- Author
-
Qingxiu Zhang, Di Xie, Ligen Shi, Donna B. Stolz, Xuejiao Dai, Kong Chen, Lesley M. Foley, Kartik Iyer, Jun Chen, T. Kevin Hitchens, Xiaoming Hu, Rehana K. Leak, Zeyu Sun, Sicheng Li, Wei Su, Ying Ding, Fei Xu, and Angus W. Thomson
- Subjects
0301 basic medicine ,Male ,Immunology ,chemical and pharmacologic phenomena ,T-Lymphocytes, Regulatory ,Flow cytometry ,Brain Ischemia ,Transcriptome ,White matter ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Immunology and Allergy ,Animals ,Osteopontin ,Ischemic Stroke ,biology ,Microglia ,medicine.diagnostic_test ,Regeneration (biology) ,Macrophages ,hemic and immune systems ,Recovery of Function ,White Matter ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Interleukin-2 ,Antibody - Abstract
Summary The precise mechanisms underlying the beneficial effects of regulatory T (Treg) cells on long-term tissue repair remain elusive. Here, using single-cell RNA sequencing and flow cytometry, we found that Treg cells infiltrated the brain 1 to 5 weeks after experimental stroke in mice. Selective depletion of Treg cells diminished oligodendrogenesis, white matter repair, and functional recovery after stroke. Transcriptomic analyses revealed potent immunomodulatory effects of brain-infiltrating Treg cells on other immune cells, including monocyte-lineage cells. Microglia depletion, but not T cell lymphopenia, mitigated the beneficial effects of transferred Treg cells on white matter regeneration. Mechanistically, Treg cell-derived osteopontin acted through integrin receptors on microglia to enhance microglial reparative activity, consequently promoting oligodendrogenesis and white matter repair. Increasing Treg cell numbers by delivering IL-2:IL-2 antibody complexes after stroke improved white matter integrity and rescued neurological functions over the long term. These findings reveal Treg cells as a neurorestorative target for stroke recovery.
- Published
- 2020
10. The fourth international workshop on clinical transplant tolerance
- Author
-
Dixon B. Kaufman, Sindhu Chandran, Megan Sykes, Josh Levitsky, Nancy D. Bridges, Lisbeth A. Welniak, Fadi Issa, Samuel Strober, Peter Nickerson, Joseph R. Leventhal, Tatsuo Kawai, Fadi G. Lakkis, Joren C. Madsen, V. Mas, Angus W. Thomson, Kathryn J. Wood, and Anthony J. Demetris
- Subjects
Graft Rejection ,medicine.medical_specialty ,medicine.medical_treatment ,education ,030230 surgery ,Organ transplantation ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immune Tolerance ,Immunology and Allergy ,Lung transplantation ,Humans ,Pharmacology (medical) ,Intensive care medicine ,Immunosuppression Therapy ,Transplantation ,Hematopoietic cell ,business.industry ,Immunosuppression ,Organ Transplantation ,Pennsylvania ,Clinical trial ,Risk stratification ,Transplantation Tolerance ,business ,Solid organ transplantation ,Immunosuppressive Agents - Abstract
The International Workshop on Clinical Transplant Tolerance is a biennial meeting that aims to provide an update on the progress of studies of immunosuppression minimization or withdrawal in solid organ transplantation. The Fourth International Workshop on Clinical Tolerance was held in Pittsburgh, Pennsylvania, September 5-6, 2019. This report is a summary of presentations on the status of clinical trials designed to minimize or withdraw immunosuppressive drugs in kidney, liver, and lung transplantation without subsequent evidence of rejection. All protocols had in common the use of donor or recipient cell therapy combined with organ transplantation. The workshop also included presentations of mechanistic studies designed to improve understanding of the cellular and molecular basis of tolerance and to identify potential predictors/biomarkers of tolerance. Strategies to enhance the safety of hematopoietic cell transplantation and to improve patient selection/risk stratification for clinical trials were also discussed.
- Published
- 2020
- Full Text
- View/download PDF
11. Transplant Tolerance Induction: Insights From the Liver
- Author
-
Helong Dai, Yawen Zheng, Angus W. Thomson, and Natasha M. Rogers
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,immune tolerance ,medicine.medical_treatment ,Immunology ,Cell- and Tissue-Based Therapy ,Review ,Liver transplantation ,Bioinformatics ,Immune tolerance ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Species Specificity ,Immunity ,medicine ,Animals ,Humans ,Immunology and Allergy ,Immunosuppression Therapy ,Clinical Trials as Topic ,mechanisms ,liver transplantation ,business.industry ,Models, Immunological ,Immunosuppression ,Tolerance induction ,030104 developmental biology ,Immunosuppressive drug ,Cellular Microenvironment ,Liver ,Organ Specificity ,Models, Animal ,immunosuppression withdrawal ,Transplantation Tolerance ,cell therapy ,lcsh:RC581-607 ,business ,030215 immunology - Abstract
A comparison of pre-clinical transplant models and of solid organs transplanted in routine clinical practice demonstrates that the liver is most amenable to the development of tolerance. This phenomenon arises in the absence of stringent conditioning regimens that accompany published tolerizing protocols for other organs, particularly the kidney. The unique immunologic facet of the liver has assisted our understanding of the alloimmune response and how it can be manipulated to improve graft function and survival. This review will briefly address important experimental findings following liver transplantation in both animals and humans, and how these have driven the understanding and development of therapeutic immunosuppressive options. We will discuss the liver’s unique system of immune and non-immune cells that regulate immunity, yet maintain effective responses to pathogens, as well as mechanisms of liver transplant tolerance in pre-clinical models and humans, including current immunosuppressive drug withdrawal trials and biomarkers of tolerance. In addition, we will address innovative therapeutic strategies, including mesenchymal stem cell, regulatory T cells and dendritic cell therapy to promote liver allograft tolerance or minimization and withdrawal of immunosuppression in the clinic.
- Published
- 2020
- Full Text
- View/download PDF
12. Regulatory dendritic cells: profiling, targeting, and therapeutic application
- Author
-
Angus W. Thomson and Mohamed Ezzelarab
- Subjects
0301 basic medicine ,Transplantation ,medicine.medical_specialty ,biology ,Dendritic Cells ,Dendritic cell ,Major histocompatibility complex ,Living donor ,Article ,Organ transplantation ,Secondary lymphoid organs ,Cell biology ,Regulatory molecules ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,In vivo ,Immune Tolerance ,biology.protein ,medicine ,Humans ,Immunology and Allergy ,030215 immunology - Abstract
Purpose of review There is currently increased focus on improved understanding of how dendritic cell tolerogenicity is determined and maintained, and on their therapeutic potential. We review recent progress in profiling of regulatory dendritic cells (DCreg), innovative approaches to enhancing dendritic cell tolerogenicity in situ, ex-vivo generation of DCreg and initial clinical testing of these cells in organ transplantation. Recent findings "Omics' studies indicate that the distinctive properties of DCreg are the result of a specific transcriptional program characterized by activation of tolerance-enhancing genes, rather than the retention of an immature state. In situ dendritic cell-directed targeting of nanovesicles bearing immune regulatory molecules can trigger in-vivo expansion of Ag-specific regulatory cells. Innovative approaches to ex-vivo modification of dendritic cells to enhance their regulatory function and capacity to migrate to secondary lymphoid organs has been described. Cross-dressing (with donor major histocompatibility complex molecules) of graft-infiltrating host dendritic cells that regulate antidonor T-cell responses has been implicated in "spontaneous' liver transplant tolerance. Clinical trials of DCreg therapy have begun in living donor renal and liver transplantation. Summary Further definition of molecules that can be targeted to promote the function and stability of DCreg in vivo may lead to standardization of DCreg manufacturing for therapeutic application.
- Published
- 2018
- Full Text
- View/download PDF
13. Characterization of eomesodermin and T-bet expression by allostimulated CD8+ T cells of healthy volunteers and kidney transplant patients in relation to graft outcome
- Author
-
Angus W. Thomson, Lien Lu, Diana Metes, S Hariharan, Angelica Perez-Gutierrez, and Mohamed Ezzelarab
- Subjects
0301 basic medicine ,genetic structures ,business.industry ,T cell ,Immunology ,Eomesodermin ,030230 surgery ,eye diseases ,Transplantation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Downregulation and upregulation ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Tumor necrosis factor alpha ,sense organs ,business ,Memory T cell ,CD8 - Abstract
Memory T cell (Tmem) responses play a critical role in the outcome of allo-transplantation. While the role of the T-box transcription factor Eomesodermin (Eomes) in the maintenance of antigen-specific Tmem is well studied, little is known about Eomes+CD8+T cell responses after transplantation. We evaluated the phenotype and function of allo-reactive Eomes+CD8+T cells in healthy volunteers and kidney transplant patients and their relation to transplant outcome. High Eomes expression by steady-state CD8+T cells correlated with effector and memory phenotype. Following allo-stimulation, the expression of both the T-box proteins Eomes and T-bet by proliferating cells increased significantly, where high expression of Eomes and T-bet correlated with higher incidence of allo-stimulated IFNγ+TNFα+ CD8+T cells. In patients with no subsequent rejection, Eomes but not T-bet expression by donor-stimulated CD8+T cells, increased significantly after transplantation. This was characterized by increased EomeshiT-bet-/lo and decreased Eomes-/loT-bethi CD8+T cell subsets, with no significant changes in the EomeshiT-bethi CD8+T cell subset. No upregulation of exhaustion markers programmed-death-1 (PD-1) and cytotoxic-T-lymphocyte-associated-antigen-4 (CTLA4) by donor-stimulated Eomes+CD8+T cells was observed. Before transplantation, in patients without rejection, there were higher incidences of EomeshiT-bet-/lo, and lower incidences of EomeshiT-bethi and Eomes-/loT-bethi donor-stimulated CD8+T cell subsets, compared to those with subsequent rejection. Overall, our findings indicate that high Eomes expression by allo-stimulated T-bet+CD8+T cells is associated with enhanced effector function, and that an elevated incidence of donor-stimulated CD8+T cells co-expressing high levels of Eomes and T-bet before transplantation, may correlate with an increased incidence of acute cellular rejection.
- Published
- 2018
- Full Text
- View/download PDF
14. Regulatory dendritic cells for promotion of liver transplant operational tolerance: Rationale for a clinical trial and accompanying mechanistic studies
- Author
-
Angus W. Thomson, Diana Metes, Fadi G. Lakkis, and Abhinav Humar
- Subjects
Graft Rejection ,0301 basic medicine ,medicine.medical_treatment ,T cell ,Immunology ,030230 surgery ,Liver transplantation ,T-Lymphocytes, Regulatory ,Article ,Cell therapy ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,business.industry ,Graft Survival ,Dendritic Cells ,General Medicine ,Kidney Transplantation ,Tacrolimus ,Liver Transplantation ,030104 developmental biology ,Immunosuppressive drug ,medicine.anatomical_structure ,Models, Animal ,Transplantation Tolerance ,Bone marrow ,business ,Immunologic Memory ,Memory T cell ,CD8 - Abstract
Dendritic cells (DC) are rare, bone marrow (BM)-derived innate immune cells that critically maintain self-tolerance in the healthy steady-state. Regulatory DC (DCreg) with capacity to suppress allograft rejection and promote transplant tolerance in pre-clinical models can readily be generated from BM precursors or circulating blood monocytes. These DCreg enhance allograft survival via various mechanisms, including promotion of regulatory T cells. In non-human primates receiving minimal immunosuppressive drug therapy (IS), infusion of DCreg of donor origin, one week before transplant, safely prolongs renal allograft survival and selectively attenuates anti-donor CD8+ memory T cell responses in the early post-transplant period. Based on these observations, and in view of the critical need to reduce patient dependence on non-specific IS agents that predispose to cardiometabolic side effects and renal insufficiency, we will conduct a first-in-human safety and preliminary efficacy study of donor-derived DCreg infusion to achieve early (18 months post-transplant) complete IS withdrawal in low-risk, living donor liver transplant recipients receiving standard-of-care IS (mycophenolate mofetil, tacrolimus and steroids). We will test the hypothesis that, although donor-derived DCreg are short-lived, they will induce robust donor-specific T cell hyporesponsiveness. We will examine immunological mechanisms by sequential analysis of blood and tissue samples, incorporating cutting-edge technologies.
- Published
- 2018
- Full Text
- View/download PDF
15. Combined GM-CSF and G-CSF administration mobilizes CD4(+)CD25(hi)Foxp3(hi) Treg in leukapheresis products of rhesus monkeys
- Author
-
Veronica Vujevich, Lien Lu, Julia Hughes, Mohamed Ezzelarab, Kazuki Sasaki, Angus W. Thomson, and Yu-Chao Wang
- Subjects
medicine.medical_treatment ,T cell ,chemical and pharmacologic phenomena ,030230 surgery ,T-Lymphocytes, Regulatory ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Granulocyte Colony-Stimulating Factor ,medicine ,Immunology and Allergy ,Animals ,Pharmacology (medical) ,Leukapheresis ,Transplantation ,business.industry ,FOXP3 ,Granulocyte-Macrophage Colony-Stimulating Factor ,hemic and immune systems ,Macaca mulatta ,Cytokine ,medicine.anatomical_structure ,Immunology ,business ,Ex vivo ,CD8 ,Transcription Factors - Abstract
Early phase clinical trials are evaluating the feasibility, safety and therapeutic potential of ex vivo expanded regulatory T cells (Treg) in transplantation. A limitation is the paucity of naturally-occurring Treg numbers in peripheral blood. Hence, protracted ex vivo expansion is required to obtain sufficient Treg in order to meet target cell doses. Since cytokine administration has been used successfully to mobilize immune cells to the peripheral blood in experimental and clinical studies, we hypothesized that granulocyte-macrophage-colony stimulating factor (GM-CSF) and granulocyte-CSF (G-CSF) administration would enhance Treg percentages in leukapheresis products of rhesus monkeys. Following combined GM-CSF and G-CSF administration, the incidence of Treg in peripheral blood and leukapheresis products was elevated significantly, where approximately 3.7x10(6)/kg CD4(+)CD25(hi)Foxp3(hi) or 6.8x10(6)/kg CD4(+)CD25(hi)CD127(lo) Treg can be collected from individual products. Mobilized Treg expressed a comparable repertoire of surface markers, chemokine receptors and transcription factors to naïve monkey peripheral blood Treg. Furthermore, when expanded ex vivo, mobilized leukapheresis product and peripheral blood Treg exhibited similar ability to suppress autologous CD4(+) and CD8(+) T cell proliferation. These observations indicate that leukapheresis products from combined GM-CSF and G-CSF-mobilized individuals are a comparatively rich source of Treg and may circumvent long-term ex vivo expansion required for therapeutic application.
- Published
- 2020
16. Dendritic Cells as Sensors, Mediators, and Regulators of Ischemic Injury
- Author
-
Helong Dai, Angus W. Thomson, and Natasha M. Rogers
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,kidney ,Regulatory T cell ,medicine.medical_treatment ,Mini Review ,Cell Plasticity ,Immunology ,Inflammation ,Cell Communication ,heart ,Biology ,liver ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Ischemia ,medicine ,Immunology and Allergy ,Animals ,Humans ,dendritic cells ,Hypoxia ,Ischemic Preconditioning ,fungi ,Pattern recognition receptor ,Immunity ,Acquired immune system ,Natural killer T cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Phenotype ,Organ Specificity ,Reperfusion Injury ,Disease Susceptibility ,medicine.symptom ,Signal transduction ,lcsh:RC581-607 ,ischemic injury ,Biomarkers ,030215 immunology - Abstract
Dendritic cells (DCs) are highly specialized, bone marrow (BM)-derived antigen-processing and -presenting cells crucial to the induction, integration and regulation of innate, and adaptive immunity. They are stimulated by damage-associated molecular patterns (DAMPS) via pattern recognition receptors to promote inflammation and initiate immune responses. In addition to residing within the parenchyma of all organs as part of the heterogeneous mononuclear phagocyte system, DCs are an abundant component of the inflammatory cell infiltrate that appears in response to ischemia reperfusion injury (IRI). They can play disparate roles in the pathogenesis of IRI since their selective depletion has been found to be protective, deleterious, or of no benefit in mouse models of IRI. In addition, administration of DC generated and manipulated ex vivo can protect organs from IRI by suppressing inflammatory cytokine production, limiting the capacity of DCs to activate NKT cells, or enhancing regulatory T cell function. Few studies however have investigated specific signal transduction mechanisms underlying DC function and how these affect IRI. Here, we address current knowledge of the role of DCs in regulation of IRI, current gaps in understanding and prospects for innovative therapeutic intervention at the biological and pharmacological levels.
- Published
- 2019
- Full Text
- View/download PDF
17. Preliminary assessment of the feasibility of autologous myeloid-derived suppressor cell infusion in non-human primate kidney transplantation
- Author
-
Martin Wijkstrom, Marta I. Minervini, Roger W. Wiseman, Abhinav Humar, Yu-Chao Wang, Alan F. Zahorchak, Angelica Perez-Gutierrez, Lien Lu-Casto, Mohamed Ezzelarab, and Angus W. Thomson
- Subjects
Graft Rejection ,medicine.medical_specialty ,Cell Transplantation ,T cell ,T-Lymphocytes ,Immunology ,Population ,030230 surgery ,Belatacept ,Transplantation, Autologous ,Organ transplantation ,Article ,Abatacept ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Histocompatibility Antigens ,Immune Tolerance ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Transplantation, Homologous ,education ,Kidney transplantation ,Cells, Cultured ,Sirolimus ,Transplantation ,education.field_of_study ,business.industry ,Myeloid-Derived Suppressor Cells ,medicine.disease ,Kidney Transplantation ,Macaca mulatta ,Disease Models, Animal ,medicine.anatomical_structure ,Myeloid-derived Suppressor Cell ,Feasibility Studies ,business ,Immunosuppressive Agents ,030215 immunology ,medicine.drug - Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogenous population of immunosuppressive myeloid cells now considered important immune regulatory cells in diverse clinical conditions, including cancer, chronic inflammatory disorders and transplantation. In rodents, MDSC administration can inhibit graft-versus-host disease lethality and enhance organ or pancreatic islet allograft survival. There is also evidence, however, that under systemic inflammatory conditions, adoptively-transferred MDSC can rapidly lose their suppressive function. To our knowledge, there are no reports of autologous MDSC administration to either human or clinically-relevant non-human primate (NHP) transplant recipients. Monocytic (m) MDSC have been shown to be more potent suppressors of T cell responses than other subsets of MDSC. Following their characterization in rhesus macaques, we have conducted a preliminary analysis of the feasibility and preliminary efficacy of purified mMDSC infusion into MHC-mismatched rhesus kidney allograft recipients. The graft recipients were treated with rapamycin and the high affinity variant of the T cell co-stimulation blocking agent cytotoxic T lymphocyte antigen 4 Ig (Belatacept) that targets the B7-CD28 pathway. Graft survival and histology were not affected by infusions of autologous, leukapheresis product-derived mMDSC on days 7 and 14 post-transplant (cumulative totals of 3.19 and 1.98 × 106 cells/kg in n = 2 recipients) compared with control monkeys that did not receive MDSC (n = 2). Sequential analyses of effector T cell populations revealed no differences between the groups. While these initial findings do not provide evidence of efficacy under the conditions adopted, further studies in NHP, designed to ascertain the appropriate mMDSC source and dose, timing and anti-inflammatory/immunosuppressive agent support are likely to prove instructive regarding the therapeutic potential of MDSC in organ transplantation.
- Published
- 2019
18. The 'other' mTOR complex: new insights into mTORC2 immunobiology and their implications
- Author
-
Helong Dai and Angus W. Thomson
- Subjects
Cell ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,mTORC2 ,Article ,Mice ,T-Lymphocyte Subsets ,Transplantation Immunology ,medicine ,Immunology and Allergy ,Animals ,Humans ,Pharmacology (medical) ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Transplantation ,B-Lymphocytes ,biology ,business.industry ,Models, Immunological ,Cell migration ,medicine.disease ,Immunity, Innate ,Transplant rejection ,Cell biology ,Mitochondria ,Cell metabolism ,medicine.anatomical_structure ,Reperfusion Injury ,biology.protein ,business - Abstract
A central role of the mechanistic target of rapamycin (mTOR) in regulation of fundamental cell processes is well recognized. mTOR functions in two distinct complexes: rapamycin-sensitive mTOR complex (C) 1 and rapamycin-insensitive mTORC2. While the role of mTORC1 in shaping immune responses, including transplant rejection, and the influence of its antagonism in promoting allograft tolerance have been studied extensively using rapamycin, lack of selective small molecule inhibitors has limited understanding of mTORC2 biology. Within the past few years, however, intracellular localization of mTORC2, its contribution to mitochondrial fitness, cell metabolism, cytoskeletal modeling and cell migration, and its role in differentiation and function of immune cells have been described. Studies in mTORC2 knockdown/knockout mouse models and a new class of dual mTORC1/2 inhibitors, have shed light on the immune regulatory functions of mTORC2. These include regulation of antigen-presenting cell, NK cell, T cell subset, and B cell differentiation and function. mTORC2 has been implicated in regulation of ischemia/reperfusion injury and graft rejection. Potential therapeutic benefits of antagonizing mTORC2 to inhibit chronic rejection have also been described, while selective in vivo targeting strategies using nanotechnology have been developed. We briefly review and discuss these developments and their implications.
- Published
- 2019
19. Hepatic stellate cells increase the immunosuppressive function of natural Foxp3+ regulatory T cells via IDO-induced AhR activation
- Author
-
Angus W. Thomson, Chandrashekhar R. Gandhi, Sudhir Kumar, and Jiang Wang
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Adoptive cell transfer ,Immunoprecipitation ,Immunology ,Biology ,Retinoblastoma Protein ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,0302 clinical medicine ,Hepatic Stellate Cells ,Animals ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Immunology and Allergy ,IL-2 receptor ,Phosphorylation ,Receptor ,Cell Proliferation ,Cryopreservation ,Immunosuppression Therapy ,Mice, Inbred BALB C ,Protein Stability ,Tryptophan ,FOXP3 ,Acetylation ,Forkhead Transcription Factors ,hemic and immune systems ,Cell Biology ,Aryl hydrocarbon receptor ,Coculture Techniques ,Receptors, Signal Transduction, & Genes ,Cell biology ,Blot ,030104 developmental biology ,Receptors, Aryl Hydrocarbon ,Hepatic stellate cell ,biology.protein ,Cytokines ,Signal Transduction ,030215 immunology - Abstract
Immunosuppressive, naturally occurring CD4+CD25+forkhead box p3+ (Foxp3+) regulatory T cells (nTregs) offer potential for the treatment of immune-mediated inflammatory disorders. However, potential instability of ex vivo-expanded nTregs following their adoptive transfer may be a significant limitation. LPS-stimulated hepatic stellate cells (HSCs) induce expansion and enhance the suppressive function and stability of allogeneic nTregs. We aimed to delineate mechanisms underlying HSC-induced expansion and increased potency of nTregs. HSCs and nTregs were isolated from mouse livers and spleens, respectively. Following coculture with LPS-pretreated allogeneic HSCs (LPS/HSCs), proliferation of nTregs was measured by CFSE dilution, and Foxp3 expression and acetylation were determined by immunoprecipitation (IP) and Western blotting analysis. Expression of various genes associated with immunologic tolerance was determined by quantitative RT-PCR (qRT-PCR). LPS stimulation increased the expression and activity of the immunoregulatory enzyme IDO1 in HSCs, and LPS/HSCs stimulated aryl hydrocarbon receptor (AhR) signaling in cocultured nTregs. Reciprocally, Tregs increased IDO1 expression in HSCs. IDO1−/− LPS/HSCs were inferior to WT LPS/HSCs in stimulating nTreg expansion. Pharmacologic inhibition of IDO1 in HSCs by 1-methyltryptophan (1MT) inhibited LPS/HSC-induced AhR signaling in nTregs, which was responsible for their expansion, Foxp3 expression, and stabilization of Foxp3 by increasing acetylation of lysine residues. Finally, HSCs cryopreserved, following 2–3 passages, were as potent as primary-cultured HSCs in expanding nTregs. In conclusion, LPS/HSCs expand allogeneic nTregs through an IDO-dependent, AhR-mediated mechanism and increase their stability through lysine-acetylation of Foxp3. nTregs expanded by cryopreserved HSCs may have potential for clinical use.
- Published
- 2016
- Full Text
- View/download PDF
20. mTORC2 deficiency in cutaneous dendritic cells potentiates CD8(+) effector T cell responses and accelerates skin graft rejection
- Author
-
Alicia R. Watson, Helong Dai, Angus W. Thomson, Julio A. Diaz-Perez, Alicia R. Mathers, and Meaghan E. Killeen
- Subjects
Graft Rejection ,Male ,T cell ,CD11c ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,030230 surgery ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Immunology and Allergy ,Animals ,Pharmacology (medical) ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Skin ,Mice, Knockout ,Transplantation ,biology ,business.industry ,Graft Survival ,Dendritic cell ,Dendritic Cells ,Skin Transplantation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Female ,business ,CD8 - Abstract
Mechanistic target of rapamycin (mTOR) complex (mTORC)1 and mTORC2 regulate the differentiation and function of immune cells. While inhibition of mTORC1 antagonizes dendritic cell (DC) differentiation and suppresses graft rejection, the role of mTORC2 in DCs in determining host responses to transplanted tissue remains undefined. Using a mouse model in which mTORC2 was deleted specifically in CD11c+ DCs (TORC2DC-/- ), we show that the transplant of minor histocompatibility Ag (HY)-mismatched skin grafts from TORC2DC-/- donors into wild-type recipients results in accelerated rejection characterized by enhanced CD8+ T cell responses in the graft and regional lymphoid tissue [Correction added on January 9, 2019, after first online publication: in the previous sentence, major was changed to minor]. Similar enhancement of CD8+ effector T cell responses was observed in MHC-mismatched recipients of TORC2DC-/- grafts. Augmented CD8+ T cell responses were also observed in a delayed-type hypersensitivity model in which mTORC2 was absent in cutaneous DCs. These elevated responses could be ascribed to an increased T cell stimulatory phenotype of TORC2DC-/- and not to enhanced lymph node homing of the cells. In contrast, rejection of ovalbumin transgenic skin grafts in TORC2DC-/- recipients was unaffected. These findings suggest that mTORC2 in skin DCs restrains effector CD8+ T cell responses and have implications for understanding of the influence of mTOR inhibitors that target mTORC2 in transplant.
- Published
- 2018
21. Donor-Derived Regulatory Dendritic Cell Infusion Maintains Donor-Reactive CD4+CTLA4hi T Cells in Non-Human Primate Renal Allograft Recipients Treated with CD28 Co-Stimulation Blockade
- Author
-
Mohamed B. Ezzelarab, Lien Lu, William F. Shufesky, Adrian E. Morelli, and Angus W. Thomson
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,medicine.medical_specialty ,T cell ,Immunology ,030230 surgery ,regulatory T cells ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Downregulation and upregulation ,Internal medicine ,medicine ,Cytotoxic T cell ,Immunology and Allergy ,co-stimulation blockade ,dendritic cells ,Chemistry ,CD28 ,Dendritic cell ,Blockade ,Transplantation ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,renal allografts ,non-human primates ,lcsh:RC581-607 - Abstract
Donor-derived regulatory dendritic cell (DCreg) infusion before transplantation, significantly prolongs renal allograft survival in non-human primates. This is associated with enhanced expression of the immunoregulatory molecules cytotoxic T-lymphocyte-associated antigen (Ag) 4 (CTLA4) and programmed cell death protein 1 (PD1) by host donor-reactive T cells. In rodents and humans, CD28 co-stimulatory pathway blockade with the fusion protein CTLA4:Ig (CTLA4Ig) is associated with reduced differentiation and development of regulatory T cells (Treg). We hypothesized that upregulation of CTLA4 by donor-reactive CD4+ T cells in DCreg-infused recipients treated with CTLA4Ig, might be associated with higher incidences of donor-reactive CD4+ T cells with a Treg phenotype. In normal rhesus monkeys, allo-stimulated CD4+CTLA4hi, but not CD4+CTLA4med/lo T cells exhibited a regulatory phenotype, irrespective of PD1 expression. CTLA4Ig significantly reduced the incidence of CD4+CTLA4hi, but not CD4+CTLA4med/lo T cells following allo-stimulation, associated with a significant reduction in the CD4+CTLA4hi/CD4+CTLA4med/lo T cell ratio. In CTLA4Ig-treated renal allograft recipient monkeys, there was a marked reduction in circulating donor-reactive CD4+CTLA4hi T cells. In contrast, in CTLA4Ig-treated monkeys with DCreg infusion, no such reduction was observed. In parallel, the donor-reactive CD4+CTLA4hi/CD4+CTLA4med/lo T cell ratio was reduced significantly in graft recipients without DCreg infusion, but increased in those given DCreg. These observations suggest that pre-transplant DCreg infusion promotes and maintains donor-reactive CD4+CTLA4hi T cells with a regulatory phenotype after transplantation, even in the presence of CD28 co-stimulation blockade.
- Published
- 2018
- Full Text
- View/download PDF
22. Clinical Implications of Basic Science Discoveries: Immune Homeostasis and the Microbiome—Dietary and Therapeutic Modulation and Implications for Transplantation
- Author
-
Jay A. Fishman and Angus W. Thomson
- Subjects
Transplantation ,Innate immune system ,Effector ,Microbiota ,Probiotics ,Human microbiome ,Organ Transplantation ,Dendritic cell ,Biology ,Immunity, Innate ,Immune system ,Immune System Diseases ,Antigen ,Immune System ,Immunology ,Homeostasis ,Humans ,Immunology and Allergy ,Pharmacology (medical) ,Microbiome - Abstract
Links between the human microbiome and the innate and adaptive immune systems and their impact on autoimmune and inflammatory diseases are only beginning to be recognized. Characterization of the complex human microbial community is facilitated by culture-independent nucleic acid sequencing tools and bioinformatics systems. Specific organisms and microbial antigens are linked with initiation of innate immune responses that, depending on the context, may be associated with tolerogenic or effector immune responses. Further complexity is introduced by preclinical data that demonstrate the impacts of dietary manipulation on the prevention of genetically determined, systemic autoimmune disorders and on gastrointestinal microbiota. Investigation of interactions of complex microbial populations with the human immune system may provide new targets for clinical management in allotransplantation.
- Published
- 2015
- Full Text
- View/download PDF
23. IRF-1 Promotes Liver Transplant Ischemia/Reperfusion Injury via Hepatocyte IL-15/IL-15Rα Production
- Author
-
Shinichiro Yokota, David A. Geller, Donna B. Stolz, Qiang Du, Osamu Yoshida, Mark A. Ross, Anthony V. Spadaro, Kumiko Isse, Anthony J. Demetris, Lei Dou, Angus W. Thomson, and Shoko Kimura
- Subjects
Male ,medicine.medical_treatment ,T cell ,Immunology ,Cell Culture Techniques ,Liver transplantation ,Biology ,Models, Biological ,Article ,Gene Knockout Techniques ,Mice ,Interleukin-15 Receptor alpha Subunit ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Gene Silencing ,Interleukin-15 ,Mice, Knockout ,Liver injury ,Cell Death ,Dendritic Cells ,Allografts ,medicine.disease ,Lymphocyte Subsets ,Liver Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,Gene Expression Regulation ,Liver ,Interleukin 15 ,Reperfusion Injury ,Hepatocyte ,Hepatocytes ,Cancer research ,Cytokines ,Inflammation Mediators ,Reperfusion injury ,CD8 ,Interferon Regulatory Factor-1 ,Protein Binding - Abstract
Ischemia and reperfusion (I/R) injury following liver transplantation (LTx) is an important problem that significantly impacts clinical outcomes. IFN regulatory factor-1 (IRF-1) is a nuclear transcription factor that plays a critical role in liver injury. Our objective was to determine the immunomodulatory role of IRF-1 during I/R injury following allogeneic LTx. IRF-1 was induced in liver grafts immediately after reperfusion in both human and mouse LTx. IRF-1 contributed significantly to I/R injury because IRF-1–knockout (KO) grafts displayed much less damage as assessed by serum alanine aminotransferase and histology. In vitro, IRF-1 regulated both constitutive and induced expression of IL-15, as well as IL-15Rα mRNA expression in murine hepatocytes and liver dendritic cells. Specific knockdown of IRF-1 in human primary hepatocytes gave similar results. In addition, we identified hepatocytes as the major producer of soluble IL-15/IL-15Rα complexes in the liver. IRF-1–KO livers had significantly reduced NK, NKT, and CD8+ T cell numbers, whereas rIL-15/IL-15Rα restored these immune cells, augmented cytotoxic effector molecules, promoted systemic inflammatory responses, and exacerbated liver injury in IRF-1–KO graft recipients. These results indicate that IRF-1 promotes LTx I/R injury via hepatocyte IL-15/IL-15Rα production and suggest that targeting IRF-1 and IL-15/IL-15Rα may be effective in reducing I/R injury associated with LTx.
- Published
- 2015
- Full Text
- View/download PDF
24. Sequential Monitoring and Stability of Ex Vivo–Expanded Autologous and Nonautologous Regulatory T Cells Following Infusion in Nonhuman Primates
- Author
-
Alan F. Zahorchak, Lien Lu, Hong Zhang, Giorgio Raimondi, Mohamed Ezzelarab, David K. C. Cooper, H. Guo, Roger W. Wiseman, and Angus W. Thomson
- Subjects
Male ,medicine.medical_specialty ,Globulin ,T cell ,chemical and pharmacologic phenomena ,Endogeny ,Methylation ,T-Lymphocytes, Regulatory ,Article ,Interleukin-7 Receptor alpha Subunit ,Major Histocompatibility Complex ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,IL-2 receptor ,Interleukin-7 receptor ,Antilymphocyte Serum ,Sirolimus ,Transplantation ,biology ,business.industry ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Forkhead Transcription Factors ,hemic and immune systems ,Carboxyfluorescein succinimidyl ester ,Macaca fascicularis ,Ki-67 Antigen ,Phenotype ,medicine.anatomical_structure ,Endocrinology ,Haplotypes ,chemistry ,biology.protein ,business ,Immunosuppressive Agents ,Ex vivo - Abstract
Ex vivo-expanded cynomolgus monkey CD4(+)CD25(+)CD127(-) regulatory T cells (Treg) maintained Foxp3 demethylation status at the Treg-specific demethylation region, and potently suppressed T cell proliferation through three rounds of expansion. When carboxyfluorescein succinimidyl ester- or violet proliferation dye 450-labeled autologous (auto) and nonautologous (non-auto)-expanded Treg were infused into monkeys, the number of labeled auto-Treg in peripheral blood declined rapidly during the first week, but persisted at low levels in both normal and anti-thymocyte globulin plus rapamycin-treated (immunosuppressed; IS) animals for at least 3 weeks. By contrast, MHC-mismatched non-auto-Treg could not be detected in normal monkey blood or in blood of two out of the three IS monkeys by day 6 postinfusion. They were also more difficult to detect than auto-Treg in peripheral lymphoid tissue. Both auto- and non-auto-Treg maintained Ki67 expression early after infusion. Sequential monitoring revealed that adoptively transferred auto-Treg maintained similarly high levels of Foxp3 and CD25 and low CD127 compared with endogenous Treg, although Foxp3 staining diminished over time in these nontransplanted recipients. Thus, infused ex vivo-expanded auto-Treg persist longer than MHC-mismatched non-auto-Treg in blood of nonhuman primates and can be detected in secondary lymphoid tissue. Host lymphodepletion and rapamycin administration did not consistently prolong the persistence of non-auto-Treg in these sites.
- Published
- 2015
- Full Text
- View/download PDF
25. Evolving Perspectives of mTOR Complexes in Immunity and Transplantation
- Author
-
D. Fantus and Angus W. Thomson
- Subjects
Transplantation ,Kinase ,TOR Serine-Threonine Kinases ,Mechanistic Target of Rapamycin Complex 2 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,mTORC2 ,Cell biology ,Immune system ,Immunity ,Immune System ,Multiprotein Complexes ,biology.protein ,Animals ,Humans ,Immunology and Allergy ,Pharmacology (medical) ,Mechanistic target of rapamycin ,Immunosuppressive Agents ,PI3K/AKT/mTOR pathway - Abstract
Since the discovery of Rapamycin (RAPA) and its immunosuppressive properties, enormous progress has been made in characterizing the mechanistic target of rapamycin (mTOR). Use of RAPA and its analogues (rapalogs) as anti-rejection agents has been accompanied by extensive investigation of how targeting of mTOR complex 1 (mTORC1), the principal target of RAPA, and more recently mTORC2, affects the function of immune cells, as well as vascular endothelial cells, that play crucial roles in regulation of allograft rejection. While considerable knowledge has accumulated on the function of mTORC1 and 2 in T cells, understanding of the differential roles of these complexes in antigen-presenting cells, NK cells and B cells/plasma cells is only beginning to emerge. Immune cell-specific targeting of mTORC1 or mTORC2, together with use of novel, second generation, dual mTORC kinase inhibitors (TORKinibs) have started to play an important role in elucidating the roles of these complexes and their potential for targeting in transplantation. Much remains unknown about the role of mTOR complexes and the consequences of mTOR targeting on immune reactivity in clinical transplantation. Here we address recent advances in understanding and evolving perspectives of the role of mTOR complexes and mTOR targeting in immunity, with extrapolation to transplantation.
- Published
- 2015
- Full Text
- View/download PDF
26. Renal Allograft Survival in Nonhuman Primates Infused with Donor Antigen-Pulsed Autologous Regulatory Dendritic Cells
- Author
-
Angus W. Thomson, Roger W. Wiseman, Alan F. Zahorchak, Martin Wijkstrom, Lien Lu, Angelica Perez-Gutierrez, David K. C. Cooper, Mohamed Ezzelarab, Dàlia Raïch-Regué, A. Humar, Adrian E. Morelli, and Marta I. Minervini
- Subjects
0301 basic medicine ,Male ,Isoantigens ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,030230 surgery ,Peripheral blood mononuclear cell ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Transplantation, Homologous ,Pharmacology (medical) ,Transplantation ,business.industry ,Graft Survival ,Immunosuppression ,Dendritic cell ,Dendritic Cells ,Kidney Transplantation ,Macaca mulatta ,Tissue Donors ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Leukocytes, Mononuclear ,Kidney Failure, Chronic ,Transplantation Tolerance ,business ,CD8 - Abstract
Systemic administration of autologous regulatory dendritic cells (DCreg; unpulsed or pulsed with donor antigen [Ag]), prolongs allograft survival and promotes transplant tolerance in rodents. Here, we demonstrate that nonhuman primate (NHP) monocyte-derived DCreg preloaded with cell membrane vesicles from allogeneic peripheral blood mononuclear cells induce T cell hyporesponsiveness to donor alloantigen (alloAg) in vitro. These donor alloAg-pulsed autologous DCreg (1.4-3.6 × 106 /kg) were administered intravenously, 1 day before MHC-mismatched renal transplantation to rhesus monkeys treated with costimulation blockade (cytotoxic T lymphocyte Ag 4 immunoglobulin [CTLA4] Ig) and tapered rapamycin. Prolongation of graft median survival time from 39.5 days (no DCreg infusion; n = 6 historical controls) and 29 days with control unpulsed DCreg (n = 2), to 56 days with donor Ag-pulsed DCreg (n = 5) was associated with evidence of modulated host CD4+ and CD8+ T cell responses to donor Ag and attenuation of systemic IL-17 production. Circulating anti-donor antibody (Ab) was not detected until CTLA4 Ig withdrawal. One monkey treated with donor Ag-pulsed DCreg rejected its graft in association with progressively elevated anti-donor Ab, 525 days posttransplant (160 days after withdrawal of immunosuppression). These findings indicate a modest but not statistically significant beneficial effect of donor Ag-pulsed autologous DCreg infusion on NHP graft survival when administered with a minimal immunosuppressive drug regimen.
- Published
- 2017
27. Adenosine Triphosphate-Competitive mTOR Inhibitors: A New Class of Immunosuppressive Agents That Inhibit Allograft Rejection
- Author
-
Quan Liu, Dàlia Raïch-Regué, Brian R. Rosborough, Raman Venkataramanan, Heth R. Turnquist, and Angus W. Thomson
- Subjects
CD4-Positive T-Lymphocytes ,Graft Rejection ,Male ,Morpholines ,T cell ,mTORC1 ,CD8-Positive T-Lymphocytes ,Binding, Competitive ,Polymerase Chain Reaction ,mTORC2 ,Article ,Mice ,Adenosine Triphosphate ,medicine ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,DNA Primers ,Sirolimus ,Transplantation ,Base Sequence ,business.industry ,Cell growth ,TOR Serine-Threonine Kinases ,Graft Survival ,RPTOR ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Immunology ,Cancer research ,business ,Immunosuppressive Agents ,medicine.drug - Abstract
The mechanistic/mammalian target of rapamycin (mTOR) is inhibited clinically to suppress T cell function and prevent allograft rejection. mTOR is the kinase subunit of two mTOR-containing complexes, mTOR complex (mTORC) 1 and 2. Although mTORC1 is inhibited by the macrolide immunosuppressant rapamycin (RAPA), its efficacy may be limited by its inability to block mTORC1 completely and its limited effect on mTORC2. Adenosine triphosphate (ATP)-competitive mTOR inhibitors are an emerging class of mTOR inhibitors that compete with ATP at the mTOR active site and inhibit any mTOR-containing complex. Since this class of compounds has not been investigated for their immunosuppressive potential, our goal was to determine the influence of a prototypic ATP-competitive mTOR inhibitor on allograft survival. AZD8055 proved to be a potent suppressor of T cell proliferation. Moreover, a short, 10-day course of the agent successfully prolonged murine MHC-mismatched, vascularized heart transplant survival. This therapeutic effect was associated with increased graft-infiltrating regulatory T cells and reduced CD4(+) and CD8(+) T cell interferon-γ production. These studies establish for the first time, that ATP-competitive mTOR inhibition can prolong organ allograft survival and warrant further investigation of this next generation mTOR inhibitors.
- Published
- 2014
- Full Text
- View/download PDF
28. DAP12 Deficiency in Liver Allografts Results in Enhanced Donor DC Migration, Augmented Effector T Cell Responses and Abrogation of Transplant Tolerance
- Author
-
Mark A. Ross, Angus W. Thomson, Benjamin M. Matta, Osamu Yoshida, Shoko Kimura, Shinichiro Yokota, David A. Geller, and Lei Dou
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Cell Transplantation ,T-Lymphocytes ,T cell ,medicine.medical_treatment ,Mice, Transgenic ,Biology ,Liver transplantation ,Article ,Flow cytometry ,Mice ,Cell Movement ,Leukocytes ,medicine ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,Interferon gamma ,Adaptor Proteins, Signal Transducing ,Inflammation ,Transplantation ,medicine.diagnostic_test ,FOXP3 ,Dendritic Cells ,Molecular biology ,Liver Transplantation ,Mice, Inbred C57BL ,Tolerance induction ,Phenotype ,medicine.anatomical_structure ,Liver ,Immunology ,Spleen ,CD8 ,Ex vivo ,medicine.drug - Abstract
Liver interstitial dendritic cells (DC) have been implicated in immune regulation and tolerance induction. We found that the transmembrane immuno-adaptor DNAX-activating protein of 12 kDa (DAP12) negatively regulated conventional liver myeloid (m) DC maturation and their in vivo migratory and T cell allostimulatory ability. Livers were transplanted from C57BL/6(H2(b) ) (B6) WT or DAP12(-/-) mice into WT C3H (H2(k) ) recipients. Donor mDC (H2-K(b+) CD11c(+) ) were quantified in spleens by flow cytometry. Anti-donor T cell reactivity was evaluated by ex vivo carboxyfluorescein diacetate succinimidyl ester-mixed leukocyte reaction and delayed-type hypersensitivity responses, while T effector and regulatory T cells were determined by flow analysis. A threefold to fourfold increase in donor-derived DC was detected in spleens of DAP12(-/-) liver recipients compared with those given WT grafts. Moreover, pro-inflammatory cytokine gene expression in the graft, interferon gamma (IFNγ) production by graft-infiltrating CD8(+) T cells and systemic levels of IFNγ were all elevated significantly in DAP12(-/-) liver recipients. DAP12(-/-) grafts also exhibited reduced incidences of CD4(+) Foxp3(+) cells and enhanced CD8(+) T cell IFNγ secretion in response to donor antigen challenge. Unlike WT grafts, DAP12(-/-) livers failed to induce tolerance and were rejected acutely. Thus, DAP12 expression in liver grafts regulates donor mDC migration to host lymphoid tissue, alloreactive T cell responses and transplant tolerance.
- Published
- 2014
- Full Text
- View/download PDF
29. A view of the future of regulatory immune cell therapy in organ transplantation
- Author
-
Angus W. Thomson
- Subjects
0301 basic medicine ,Transplantation ,medicine.medical_specialty ,business.industry ,Bioinformatics ,Organ transplantation ,Cell therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,Immune system ,030220 oncology & carcinogenesis ,Immunology and Allergy ,Medicine ,business - Published
- 2018
- Full Text
- View/download PDF
30. All-trans retinoic acid and rapamycin synergize with transforming growth factor-β1 to induce regulatory T cells but confer different migratory capacities
- Author
-
Siddharth Jhunjhunwala, Angus W. Thomson, Giorgio Raimondi, Erin E. Nichols, Leo C. Chen, and Steven R. Little
- Subjects
Chemokine ,Immunology ,Retinoic acid ,Tretinoin ,chemical and pharmacologic phenomena ,Cell Development, Differentiation, & Trafficking ,T-Lymphocytes, Regulatory ,Transforming Growth Factor beta1 ,Mice ,Receptors, CCR ,chemistry.chemical_compound ,Antigens, CD ,Cell Movement ,In vivo ,medicine ,Animals ,Immunology and Allergy ,Sirolimus ,biology ,Drug Synergism ,Cell Biology ,Phenotype ,Mice, Inbred C57BL ,chemistry ,Cancer research ,biology.protein ,Integrin alpha Chains ,medicine.drug ,Homing (hematopoietic) ,Transforming growth factor - Abstract
Tregs play important roles in maintaining immune homeostasis, and thus, therapies based on Treg are promising candidates for the treatment for a variety of immune-mediated disorders. These therapies, however, face the significant challenge of obtaining adequate numbers of Tregs from peripheral blood that maintains suppressive function following extensive expansion. Inducing Tregs from non-Tregs offers a viable alternative. Different methods to induce Tregs have been proposed and involve mainly treating cells with TGF-β-iTreg. However, use of TGF-β alone is not sufficient to induce stable Tregs. ATRA or rapa has been shown to synergize with TGF-β to induce stable Tregs. Whereas TGF-β plus RA-iTregs have been well-described in the literature, the phenotype, function, and migratory characteristics of TGF-β plus rapa-iTreg have yet to be elucidated. Herein, we describe the phenotype and function of mouse rapa-iTreg and reveal that these cells differ in their in vivo homing capacity when compared with mouse RA-iTreg and mouse TGF-β-iTreg. This difference in migratory activity significantly affects the therapeutic capacity of each subset in a mouse model of colitis. We also describe the characteristics of iTreg generated in the presence of TGF-β, RA, and rapa.
- Published
- 2013
- Full Text
- View/download PDF
31. Regulatory Dendritic Cell Infusion Prolongs Kidney Allograft Survival in Nonhuman Primates
- Author
-
Alan F. Zahorchak, Angus W. Thomson, Mohamed Ezzelarab, David K. C. Cooper, Ron Shapiro, Geetha Chalasani, Martin Wijkstrom, Anthony J. Demetris, Lien Lu, Noriko Murase, Adrian E. Morelli, Fadi G. Lakkis, and A. Humar
- Subjects
Male ,Immunoconjugates ,Regulatory T cell ,T cell ,T-Lymphocytes, Regulatory ,Article ,Immune tolerance ,Abatacept ,Immune Tolerance ,medicine ,Animals ,Transplantation, Homologous ,Immunology and Allergy ,Pharmacology (medical) ,Kidney transplantation ,Sirolimus ,Transplantation ,Kidney ,business.industry ,Graft Survival ,Dendritic Cells ,Dendritic cell ,medicine.disease ,Combined Modality Therapy ,Kidney Transplantation ,Macaca mulatta ,medicine.anatomical_structure ,Immunology ,Kidney Diseases ,business ,Immunologic Memory ,Immunosuppressive Agents ,medicine.drug - Abstract
We examined the influence of regulatory dendritic cells (DCreg), generated from cytokine-mobilized donor blood monocytes in vitamin D3 and IL-10, on renal allograft survival in a clinically relevant rhesus macaque model. DCreg expressed low MHC class II and costimulatory molecules, but comparatively high levels of programmed death ligand-1 (B7-H1), and were resistant to pro-inflammatory cytokine-induced maturation. They were infused intravenously (3.5-10 × 10(6) /kg), together with the B7-CD28 costimulation blocking agent CTLA4Ig, 7 days before renal transplantation. CTLA4Ig was given for up to 8 weeks and rapamycin, started on Day -2, was maintained with tapering of blood levels until full withdrawal at 6 months. Median graft survival time was 39.5 days in control monkeys (no DC infusion; n = 6) and 113.5 days (p0.05) in DCreg-treated animals (n = 6). No adverse events were associated with DCreg infusion, and there was no evidence of induction of host sensitization based on circulating donor-specific alloantibody levels. Immunologic monitoring also revealed regulation of donor-reactive memory CD95(+) T cells and reduced memory/regulatory T cell ratios in DCreg-treated monkeys compared with controls. Termination allograft histology showed moderate combined T cell- and Ab-mediated rejection in both groups. These findings justify further preclinical evaluation of DCreg therapy and their therapeutic potential in organ transplantation.
- Published
- 2013
- Full Text
- View/download PDF
32. Ex Vivo-Expanded Cynomolgus Macaque Regulatory T Cells Are Resistant to Alemtuzumab-Mediated Cytotoxicity
- Author
-
Lien Lu, Hong Zhang, David K. C. Cooper, Mohamed Ezzelarab, Jay K. Bhama, Jan N. M. IJzermans, Eefje M. Dons, Angus W. Thomson, Giorgio Raimondi, Alan F. Zahorchak, and Surgery
- Subjects
Erythrocytes ,CD52 ,medicine.drug_class ,Antibodies, Neoplasm ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Peripheral blood mononuclear cell ,T-Lymphocytes, Regulatory ,Article ,Cell therapy ,Antigens, CD ,Antigens, Neoplasm ,Immunology and Allergy ,Medicine ,Animals ,Pharmacology (medical) ,Alemtuzumab ,Glycoproteins ,Antibody-dependent cell-mediated cytotoxicity ,Transplantation ,business.industry ,Antibody-Dependent Cell Cytotoxicity ,hemic and immune systems ,Macaca fascicularis ,CD52 Antigen ,Immunology ,Leukocytes, Mononuclear ,business ,Ex vivo ,medicine.drug - Abstract
Alemtuzumab (Campath-1H) is a humanized monoclonal antibody (Ab) directed against CD52 that depletes lymphocytes and other leukocytes, mainly by complement-dependent mechanisms. We investigated the influence of alemtuzumab (i) on ex vivo-expanded cynomolgus monkeys regulatory T cells (Treg) generated for prospective use in adoptive cell therapy and (ii) on naturally-occurring Treg following alemtuzumab infusion. Treg were isolated from PBMC and lymph nodes and expanded for two rounds. CD52 expression, binding of alemtuzumab, and both complement-mediated killing and Ab-dependent cell-mediated cytotoxicity (ADCC) were compared between freshly-isolated and expanded Treg and effector T cells. Monkeys undergoing allogeneic heart transplantation given alemtuzumab were monitored for Treg and serum alemtuzumab activity. Ex vivo-expanded Treg showed progressive downregulation of CD52 expression, absence of alemtuzumab binding, minimal change in complement inhibitory protein (CD46) expression and no complement-dependent killing or ADCC. Infusion of alemtuzumab caused potent depletion of all lymphocytes, but a transient increase in the incidence of circulating Treg. After infusion of alemtuzumab, monkey serum killed fresh PBMC, but not expanded Treg. Thus, expanded cynomolgus monkey Treg are resistant to alemtuzumab-mediated, complement-dependent cytotoxicity. Furthermore, our data suggest that these expanded monkey Treg can be infused into graft recipients given alemtuzumab without risk of complement-mediated killing.
- Published
- 2013
- Full Text
- View/download PDF
33. IL-33 expands suppressive CD11b+ Gr-1(int) and regulatory T cells, including ST2L+ Foxp3+ cells, and mediates regulatory T cell-dependent promotion of cardiac allograft survival
- Author
-
Heth R. Turnquist, Angus W. Thomson, Brian R. Rosborough, A. Jake Demetris, Kathryn J. Wood, Foo Y. Liew, Zhiliang Wang, Kumiko Isse, Donna B. Stolz, Quan Liu, Zhenlin Zhao, Xin Xiao Zheng, Megan Lang, and A. Castellaneta
- Subjects
Male ,Regulatory T cell ,T cell ,Cellular differentiation ,Immunology ,Population ,Down-Regulation ,Mice, Transgenic ,Biology ,T-Lymphocytes, Regulatory ,Article ,Mice ,T-Lymphocyte Subsets ,medicine ,Immunology and Allergy ,Animals ,Myeloid Cells ,education ,Cells, Cultured ,education.field_of_study ,Mice, Inbred BALB C ,Mice, Inbred C3H ,CD11b Antigen ,Interleukins ,Graft Survival ,FOXP3 ,Receptors, Interleukin-1 ,Cell Differentiation ,Forkhead Transcription Factors ,Atherosclerosis ,Interleukin-33 ,Transplantation ,Interleukin 33 ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cancer research ,Heart Transplantation ,Receptors, Chemokine ,CD8 - Abstract
IL-33 administration is associated with facilitation of Th2 responses and cardioprotective properties in rodent models. However, in heart transplantation, the mechanism by which IL-33, signaling through ST2L (the membrane-bound form of ST2), promotes transplant survival is unclear. We report that IL-33 administration, while facilitating Th2 responses, also increases immunoregulatory myeloid cells and CD4+ Foxp3+ regulatory T cells (Tregs) in mice. IL-33 expands functional myeloid-derived suppressor cells, CD11b+ cells that exhibit intermediate (int) levels of Gr-1 and potent T cell suppressive function. Furthermore, IL-33 administration causes an St2-dependent expansion of suppressive CD4+ Foxp3+ Tregs, including an ST2L+ population. IL-33 monotherapy after fully allogeneic mouse heart transplantation resulted in significant graft prolongation associated with increased Th2-type responses and decreased systemic CD8+ IFN-γ+ cells. Also, despite reducing overall CD3+ cell infiltration of the graft, IL-33 administration markedly increased intragraft Foxp3+ cells. Whereas control graft recipients displayed increases in systemic CD11b+ Gr-1hi cells, IL-33–treated recipients exhibited increased CD11b+ Gr-1int cells. Enhanced ST2 expression was observed in the myocardium and endothelium of rejecting allografts, however the therapeutic effect of IL-33 required recipient St2 expression and was dependent on Tregs. These findings reveal a new immunoregulatory property of IL-33. Specifically, in addition to supporting Th2 responses, IL-33 facilitates regulatory cells, particularly functional CD4+ Foxp3+ Tregs that underlie IL-33–mediated cardiac allograft survival.
- Published
- 2016
34. A novel mTORC1-dependent, Akt-independent pathway differentiates the gut tropism of regulatory and conventional CD4 T cells
- Author
-
Giorgio Raimondi, Yawah Nicholson, Angus W. Thomson, Brian R. Rosborough, and Leo C. Chen
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Immunology ,Retinoic acid ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Tretinoin ,mTORC1 ,Cell Separation ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,T-Lymphocytes, Regulatory ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Chemokine receptor ,Mice ,Receptors, CCR ,0302 clinical medicine ,Immunology and Allergy ,Animals ,Protein kinase B ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,TOR Serine-Threonine Kinases ,hemic and immune systems ,Flow Cytometry ,Mice, Inbred C57BL ,Retinoic acid receptor ,Chemotaxis, Leukocyte ,030104 developmental biology ,chemistry ,Multiprotein Complexes ,biology.protein ,Cancer research ,Signal transduction ,Proto-Oncogene Proteins c-akt ,030215 immunology ,Signal Transduction - Abstract
The vitamin A metabolite all-trans retinoic acid (ATRA) induces a gut-homing phenotype in activated CD4+ conventional T cells (Tconv) by upregulating the integrin α4β7 and the chemokine receptor CCR9. We report that, in contrast to mouse Tconv, only ∼50% of regulatory T cells (Treg) upregulate CCR9 when stimulated by physiological levels of ATRA, even though Tconv and Treg express similar levels of the retinoic acid receptor (RAR). The resulting bimodal CCR9 expression is not associated with differences in the extent of their proliferation, level of Foxp3 expression, or affiliation with naturally occurring Treg or induced Treg in the circulating Treg pool. Furthermore, we find that exposure of Treg to the mechanistic target of rapamycin (mTOR) inhibitor rapamycin suppresses upregulation of both CCR9 and α4β7, an effect that is not evident with Tconv. This suggests that in Treg, ATRA-induced upregulation of CCR9 and α4β7 is dependent on activation of a mTOR signaling pathway. The involvement of mTOR is independent of Akt activity, because specific inhibition of Akt, pyruvate dehydrogenase kinase-1, or its downstream target glycogen synthase kinase-3 did not prevent CCR9 expression. Additionally, Rictor (mTOR complex [mTORC]2)-deficient Treg showed unaltered ability to express CCR9, whereas Raptor (mTORC1)-deficient Treg were unable to upregulate CCR9, suggesting the selective participation of mTORC1. These findings reveal a novel difference between ATRA signaling and chemokine receptor induction in Treg versus Tconv and provide a framework via which the migratory behavior of Treg versus Tconv might be regulated differentially for therapeutic purposes.
- Published
- 2016
35. Hepatic Stellate Cells Undermine the Allostimulatory Function of Liver Myeloid Dendritic Cells via STAT3-Dependent Induction of IDO
- Author
-
Chandrashekhar R. Gandhi, Angus W. Thomson, Donna B. Stolz, Tina L. Sumpter, Yoram Vodovotz, Benjamin M. Matta, Chao Huang, and Anil Dangi
- Subjects
Male ,STAT3 Transcription Factor ,Isoantigens ,Chemokine ,Liver cytology ,T cell ,Immunology ,Population ,Down-Regulation ,Mice, Transgenic ,Biology ,Article ,Immunophenotyping ,Mice ,Downregulation and upregulation ,Hepatic Stellate Cells ,medicine ,Animals ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Immunology and Allergy ,Myeloid Cells ,education ,Cells, Cultured ,Mice, Knockout ,CD86 ,Mice, Inbred BALB C ,education.field_of_study ,hemic and immune systems ,Dendritic Cells ,Coculture Techniques ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Liver ,Enzyme Induction ,biology.protein ,Hepatic stellate cell ,CD80 - Abstract
Hepatic stellate cells (HSCs) are critical for hepatic wound repair and tissue remodeling. They also produce cytokines and chemokines that may contribute to the maintenance of hepatic immune homeostasis and the inherent tolerogenicity of the liver. The functional relationship between HSCs and the professional migratory APCs in the liver, that is, dendritic cells (DCs), has not been evaluated. In this article, we report that murine liver DCs colocalize with HSCs in vivo under normal, steady-state conditions, and cluster with HSCs in vitro. In vitro, HSCs secrete high levels of DC chemoattractants, such as MΙP-1α and MCP-1, as well as cytokines that modulate DC activation, including TNF-α, IL-6, and IL-1β. Culture of HSCs with conventional liver myeloid (m) DCs resulted in increased IL-6 and IL-10 secretion compared with that of either cell population alone. Coculture also resulted in enhanced expression of costimulatory (CD80, CD86) and coinhibitory (B7-H1) molecules on mDCs. HSC-induced mDC maturation required cell–cell contact and could be blocked, in part, by neutralizing MΙP-1α or MCP-1. HSC-induced mDC maturation was dependent on activation of STAT3 in mDCs and, in part, on HSC-secreted IL-6. Despite upregulation of costimulatory molecules, mDCs conditioned by HSCs demonstrated impaired ability to induce allogeneic T cell proliferation, which was independent of B7-H1, but dependent upon HSC-induced STAT3 activation and subsequent upregulation of IDO. In conclusion, by promoting IDO expression, HSCs may act as potent regulators of liver mDCs and function to maintain hepatic homeostasis and tolerogenicity.
- Published
- 2012
- Full Text
- View/download PDF
36. Dendritic Cell Activation and Memory Cell Development Are Impaired among Mice Administered Medroxyprogesterone Acetate Prior to Mucosal Herpes Simplex Virus Type 1 Infection
- Author
-
Tracy Terry-Allison, Rodolfo D. Vicetti Miguel, Robert L. Hendricks, Stephen A. K. Harvey, Alfredo J. Aguirre, Melissa A. Melan, Thomas L. Cherpes, Angus W. Thomson, and Anthony J. St. Leger
- Subjects
T cell ,Immunology ,Herpesvirus 1, Human ,Medroxyprogesterone Acetate ,Article ,Mice ,Immunity ,medicine ,Animals ,Immunology and Allergy ,CD154 ,Cells, Cultured ,Mice, Knockout ,Immunity, Cellular ,Mice, Inbred BALB C ,Mucous Membrane ,CD40 ,biology ,Cell Differentiation ,Dendritic Cells ,Dendritic cell ,Coculture Techniques ,Mucosal Infection ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Keratitis, Herpetic ,biology.protein ,Female ,Immunologic Memory ,CD80 ,Ex vivo - Abstract
Epidemiological studies indicate that the exogenous sex steroid medroxyprogesterone acetate (MPA) can impair cell-mediated immunity, but mechanisms responsible for this observation are not well defined. In this study, MPA administered to mice 1 wk prior to HSV type 1 (HSV-1) infection of their corneal mucosa impaired initial expansion of viral-specific effector and memory precursor T cells and reduced the number of viral-specific memory T cells found in latently infected mice. MPA treatment also dampened expression of the costimulatory molecules CD40, CD70, and CD80 by dendritic cells (DC) in lymph nodes draining acute infection, whereas coculture of such DC with T cells from uninfected mice dramatically impaired ex vivo T cell proliferation compared with the use of DC from mice that did not receive MPA prior to HSV-1 infection. In addition, T cell expansion was comparable to that seen in untreated controls if MPA-treated mice were administered recombinant soluble CD154 (CD40L) concomitant with their mucosal infection. In contrast, the immunomodulatory effects of MPA were infection site dependent, because MPA-treated mice exhibited normal expansion of virus-specific T cells when infection was systemic rather than mucosal. Taken together, our results reveal that the administration of MPA prior to viral infection of mucosal tissue impairs DC activation, virus-specific T cell expansion, and development of virus-specific immunological memory.
- Published
- 2012
- Full Text
- View/download PDF
37. IL-27 Production and STAT3-Dependent Upregulation of B7-H1 Mediate Immune Regulatory Functions of Liver Plasmacytoid Dendritic Cells
- Author
-
Brian R. Rosborough, Benjamin M. Matta, Tina L. Sumpter, Angus W. Thomson, and Giorgio Raimondi
- Subjects
medicine.medical_treatment ,T cell ,Immunology ,FOXP3 ,hemic and immune systems ,Spleen ,EBI3 ,Biology ,Immune tolerance ,medicine.anatomical_structure ,Immune system ,Cytokine ,Downregulation and upregulation ,medicine ,Immunology and Allergy - Abstract
Plasmacytoid dendritic cells (pDCs) are highly specialized APCs that, in addition to their well-recognized role in anti-viral immunity, also regulate immune responses. Liver-resident pDCs are considerably less immunostimulatory than those from secondary lymphoid tissues and are equipped to promote immune tolerance/regulation through various mechanisms. IL-27 is an IL-12 family cytokine that regulates the function of both APCs and T cells, although little is known about its role in pDC immunobiology. In this study, we show that mouse liver pDCs express higher levels of IL-27p28 and EBV-induced protein 3 (Ebi3) compared with those of splenic pDCs. Both populations of pDCs express the IL-27Rα/WSX-1; however, only liver pDCs significantly upregulate expression of the coregulatory molecule B7 homolog-1 (B7-H1) in response to IL-27. Inhibition of STAT3 activation completely abrogates IL-27–induced upregulation of B7-H1 expression on liver pDCs. Liver pDCs treated with IL-27 increase the percentage of CD4+Foxp3+ T cells in MLR, which is dependent upon expression of B7-H1. pDCs from Ebi3-deficient mice lacking functional IL-27 show increased capacity to stimulate allogeneic T cell proliferation and IFN-γ production in MLR. Liver but not spleen pDCs suppress delayed-type hypersensitivity responses to OVA, an effect that is lost with Ebi3−/− and B7-H1−/− liver pDCs compared with wild-type liver pDCs. These data suggest that IL-27 signaling in pDCs promotes their immunoregulatory function and that IL-27 produced by pDCs contributes to their capacity to regulate immune responses in vitro and in vivo.
- Published
- 2012
- Full Text
- View/download PDF
38. Histone deacetylase inhibition facilitates GM-CSF-mediated expansion of myeloid-derived suppressor cells in vitro and in vivo
- Author
-
Sudha Natarajan, Brian R. Rosborough, Heth R. Turnquist, A. Castellaneta, and Angus W. Thomson
- Subjects
education.field_of_study ,Cell growth ,Cellular differentiation ,T cell ,Immunology ,Population ,Cell Biology ,Biology ,Molecular biology ,Haematopoiesis ,medicine.anatomical_structure ,Granulocyte macrophage colony-stimulating factor ,Myeloid Cell Differentiation ,Myeloid-derived Suppressor Cell ,Cancer research ,medicine ,Immunology and Allergy ,education ,medicine.drug - Abstract
Chromatin-modifying HDACi exhibit anti-inflammatory properties that reflect their ability to suppress DC function and enhance regulatory T cells. The influence of HDACi on MDSCs, an emerging regulatory leukocyte population that potently inhibits T cell proliferation, has not been examined. Exposure of GM-CSF-stimulated murine BM cells to HDACi led to a robust expansion of monocytic MDSC (CD11b+Ly6C+F4/80intCD115+), which suppressed allogeneic T cell proliferation in a NOS- and HO-1-dependent manner with similar potency to control MDSCs. The increased yield of MDSCs correlated with blocked differentiation of BM cells and an overall increase in HSPCs (Lin–Sca-1+c-Kit+). In vivo, TSA enhanced the mobilization of splenic HSPCs following GM-CSF administration and increased the number of CD11b+Gr1+ cells in BM and spleen. Increased numbers of Gr1+ cells, which suppressed T cell proliferation, were recovered from spleens of TSA-treated mice. Overall, HDACi enhance MDSC expansion in vitro and in vivo, suggesting that acetylation regulates myeloid cell differentiation. These findings establish a clinically applicable approach to augment this rare and potent suppressive immune cell population and support a novel mechanism underlying the anti-inflammatory action of HDACi.
- Published
- 2012
- Full Text
- View/download PDF
39. A Tale of Two Pathways: Renewing the Promise of Anti-CD40L Blockade
- Author
-
Mohamed Ezzelarab and Angus W. Thomson
- Subjects
Graft Rejection ,Primates ,CD40 Ligand ,chemical and pharmacologic phenomena ,030230 surgery ,Pharmacology ,Article ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,parasitic diseases ,Animals ,Immunology and Allergy ,Medicine ,Pharmacology (medical) ,Kidney transplantation ,Transplantation ,CD40 ,Graft rejection ,biology ,business.industry ,Graft Survival ,hemic and immune systems ,medicine.disease ,Kidney Transplantation ,Blockade ,surgical procedures, operative ,Anti cd40l ,biology.protein ,Graft survival ,business ,030215 immunology - Abstract
The advent of costimulation blockade provides the prospect for targeted therapy with improved graft survival in transplant patients. Perhaps the most effective costimulation blockade in experimental models is the use of reagents to block the CD40/CD154 pathway. Unfortunately, successful clinical translation of anti-CD154 therapy has not been achieved. In an attempt to develop an agent that is as effective as previous CD154 blocking antibodies but lacks the risk of thromboembolism, we evaluated the efficacy and safety of a novel anti-human CD154 domain antibody (dAb, BMS-986004). The anti-CD154 dAb effectively blocked CD40-CD154 interactions but lacked Fc binding activity and resultant platelet activation. In a non-human primate kidney transplant model, anti-CD154 dAb was safe and efficacious, significantly prolonging allograft survival without evidence of thromboembolism (MST 103 days). The combination of anti-CD154 dAb and conventional immunosuppression synergized to effectively control allograft rejection (MST 397 days). Furthermore, anti-CD154 dAb treatment increased the frequency of CD4+CD25+Foxp3+ regulatory T cells. This study demonstrates that the use of a novel anti-CD154 dAb that lacks Fc binding activity is safe without evidence of thromboembolism and is equally as potent as previous anti-CD154 agents at prolonging renal allograft survival in a non-human primate preclinical model.
- Published
- 2017
- Full Text
- View/download PDF
40. Rapamycin-conditioned, alloantigen-pulsed myeloid dendritic cells present donor MHC class I/peptide via the semi-direct pathway and inhibit survival of antigen-specific CD8+ T cells in vitro and in vivo
- Author
-
Heth R. Turnquist, Angus W. Thomson, Donna Beer-Stolz, Zhiliang Wang, and Ryan T. Fischer
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Isoantigens ,Adoptive cell transfer ,Cell Survival ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Article ,Mice ,Interleukin 21 ,MHC class I ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Myeloid Cells ,Antigen-presenting cell ,Bone Marrow Transplantation ,Mice, Knockout ,Sirolimus ,Mice, Inbred BALB C ,Transplantation ,biology ,Histocompatibility Antigens Class I ,Dendritic Cells ,Acquired immune system ,Transplantation, Isogeneic ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Immunosuppressive Agents ,CD8 - Abstract
Dendritic cells (DC) are “professional” bone marrow-derived antigen (Ag)-presenting cells of interest both as therapeutic targets and potential cellular vaccines due to their ability to regulate innate and adaptive immunity. Harnessing the inherent tolerogenicity of DC is a promising and incompletely explored approach to the prevention of allograft rejection. Previously, we and others have reported the ability of pharmacologically-modified DC, that resist maturation, to inhibit CD4 + T cell responses and prolong allograft survival. Here we evaluated the ability of murine myeloid DC conditioned with the immunosuppressive pro-drug rapamycin (RAPA) to acquire and directly present alloAg to syngeneic CD8 + T cells. RAPA-conditioned DC (RAPA-DC) pulsed with allogeneic splenocyte lysate acquired and expressed donor MHC class I and enhanced the apoptotic death of directly-reactive donor Ag-specific CD8 + T cells in vitro. Moreover, following their adoptive transfer, they reduced the survival of these T cells in vivo. The ability of RAPA-DC to inhibit the survival of alloAg-specific CD8 + T cells provides a potential mechanism by which host-derived DC may act as negative regulators of T cell alloreactivity and support donor-specific unresponsiveness. Adoptive cell therapy with alloAg-pulsed RAPA-DC may offer an effective approach to suppression of alloimmunity, with reduced dependence on systemic immunosuppression.
- Published
- 2011
- Full Text
- View/download PDF
41. DAP12 Promotes IRAK-M Expression and IL-10 Production by Liver Myeloid Dendritic Cells and Restrains Their T Cell Allostimulatory Ability
- Author
-
Heth R. Turnquist, Tina L. Sumpter, Angus W. Thomson, Osamu Yoshida, A. Castellaneta, and Vignesh T. Packiam
- Subjects
Small interfering RNA ,Liver cytology ,T cell ,Cellular differentiation ,Immunology ,Biology ,Molecular biology ,Cell biology ,Immune tolerance ,Interleukin 10 ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Receptor ,CD8 - Abstract
Freshly isolated hepatic dendritic cells (DC) are comparatively immature, relatively resistant to maturation, and can downmodulate effector T cell responses. Molecular mechanisms that underlie these properties are ill defined. DNAX-activating protein of 12 kDa (DAP12) is an ITAM-bearing transmembrane adaptor protein that integrates signals through several receptors, including triggering receptor expressed on myeloid cells-1, -2, and CD200R. Notably, DC propagated from DAP12-deficient mice exhibit enhanced maturation in response to TLR ligation. Given the constitutive exposure of liver DC to endotoxin draining from the gut, we hypothesized that DAP12 might regulate liver DC maturation. We show that DAP12 is expressed by freshly isolated liver, spleen, kidney, and lung myeloid DC. Moreover, inhibition of DAP12 expression by liver DC using small interfering RNA promotes their phenotypic and functional maturation, resulting in enhanced TNF-α, IL-6, and IL-12p70 production, reduced secretion of IL-10, and enhanced CD4+ and CD8+ T cell proliferation. Furthermore, DAP12 silencing correlates with decreased STAT3 phosphorylation in mature liver DC and with diminished expression of the IL-1R–associated kinase-M, a negative regulator of TLR signaling. These findings highlight a regulatory role for DAP12 in hepatic DC maturation, and suggest a mechanism whereby this function may be induced/maintained.
- Published
- 2011
- Full Text
- View/download PDF
42. The STATus of PD-L1 (B7-H1) on tolerogenic APCs
- Author
-
Angus W. Thomson and Tina L. Sumpter
- Subjects
STAT3 Transcription Factor ,Programmed Cell Death 1 Receptor ,Immunology ,Lipopolysaccharide Receptors ,Antigen-Presenting Cells ,T-Lymphocytes, Regulatory ,B7-H1 Antigen ,Monocytes ,Downregulation and upregulation ,Antigens, CD ,T-Lymphocyte Subsets ,PD-L1 ,Immune Tolerance ,Humans ,Immunology and Allergy ,Immune homeostasis ,biology ,Interleukin-6 ,Toll-Like Receptors ,Models, Immunological ,Cell Differentiation ,SUPERFAMILY ,Dendritic Cells ,Tlr agonists ,Phenotype ,Interleukin-10 ,Cell biology ,Gene Expression Regulation ,biology.protein ,Mitogen-Activated Protein Kinases ,Apoptosis Regulatory Proteins ,Function (biology) ,Signal Transduction ,Programmed death - Abstract
Expression by DCs of co-inhibitory molecules such as programmed death ligand-1 (PD-L1/B7-H1/CD274), a member of the B7 superfamily, is crucial for the downregulation of T-cell responses and the maintenance of immune homeostasis. Exposure of immature DCs to danger-associated molecular patterns (DAMPS) or pathogen-associated molecular patterns (PAMPs) generally results in their maturation and acquisition of immunostimulatory function. However, exposure of DCs to TLR ligands early during their differentiation can inhibit further differentiation and confer tolerogenic properties on these APCs. A report in this issue of The European Journal of Immunology reveals that early inhibition of human DC differentiation from blood monocytes by TLR agonists is associated with a tolerogenic phenotype and Treg generation. The tolerogenic function of these APCs is dependent on MAPK-induced IL-6 and IL-10 production, which drives STAT-3-mediated PD-L1 expression. These observations link IL-10 and IL-6 to PD-L1 expression, providing a new dimension to the anti-inflammatory properties of these cytokines. These findings also have implications for understanding the inherent function of DCs in non-lymphoid tissues such as the liver and lung, where they are exposed to PAMPs that are found constitutively in the local microenvironment.
- Published
- 2011
- Full Text
- View/download PDF
43. Combined Administration of a Mutant TGF-β1/Fc and Rapamycin Promotes Induction of Regulatory T Cells and Islet Allograft Tolerance
- Author
-
Angus W. Thomson, Xin Xiao Zheng, Yun Liu, Dong Zhang, Wensheng Zhang, Yan Tian, Miaoda Shen, and W. P. Andrew Lee
- Subjects
geography ,geography.geographical_feature_category ,Cellular differentiation ,T cell ,Immunology ,FOXP3 ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,Islet ,Proinflammatory cytokine ,Transplantation ,Tolerance induction ,medicine.anatomical_structure ,medicine ,Cancer research ,Immunology and Allergy ,Interleukin 17 - Abstract
The critical roles of TGF-β in the reciprocal differentiation of tolerance-promoting CD4+Foxp3+ regulatory T cells (Tregs) and proinflammatory Th17 effector cells affect alloimmune reactivity and transplant outcome. We reasoned that a strategy to harness TGF-β and block proinflammatory cytokines would inhibit the differentiation of Th17 cells and strengthen the cadre of Tregs to promote tolerance induction and long-term allograft survival. In this study, we report the development of a long-lasting autoactive human mutant TGF-β1/Fc fusion protein that acts in conjunction with rapamycin to inhibit T cell proliferation and induce the de novo generation of Foxp3+ Treg in the periphery, while at the same time inhibiting IL-6–mediated Th17 cell differentiation. Short-term combined treatment with TGF-β1/Fc and rapamycin achieved long-term pancreatic islet allograft survival and donor-specific tolerance in a mouse model. This effect was accompanied by expansion of Foxp3+ Tregs, enhanced alloantigen-specific Treg function, and modulation of transcript levels of Foxp3, IL-6, and IL-17. Our strategy of combined TGF-β1/Fc and rapamycin to target the IL-6–related Tregs and Th17 signaling pathways provides a promising approach for inducing transplant tolerance and its clinical application.
- Published
- 2010
- Full Text
- View/download PDF
44. Human alloreactive-regulatory T cell profile after ex vivo expansion using CD40L-stimulated B cells or monocyte-derived dendritic cells
- Author
-
Linda M. Lee, Hong Zhang, Horace Liang, Karim Lee, Angus W. Thomson, and Qizhi Tang
- Subjects
Immunology ,Immunology and Allergy - Abstract
Alloreactive regulatory T cells (arTregs) expanded with CD40L-stimulated B cells (sBcs) are being tested clinically to induce tolerance and control rejection in liver and kidney transplantation. In research settings, allogeneic monocyte-derived DCs (moDCs) have been shown to expand arTregs. We directly compared the efficacy of sBcs and cytokine-matured moDCs in expanding arTregs, and the phenotype of sBc-stimulated arTregs (sBc-arTregs) and moDC-stimulated arTregs (moDC-arTregs). arTregs expanded twice as much after stimulation with moDCs than with sBcs. Neither sBcs nor moDCs produced much TNFα, IL-1α, IL-1β, IL-6, IL-12P70, or IL-23 that can destabilize Tregs. sBc- and moDC-arTregs expressed similar percentages of Treg-defining markers - FOXP3, HELIOS, CD25, CD27, and CD62L. moDC-arTregs had slightly elevated FOXP3 and CD62L MFI. Compared to primary Tregs, sBc- and moDC-arTregs expressed similar or decreased levels of TBX21, GATA3, and RORC mRNA. However, greater percentages of moDC-arTregs produced IFNγ, IL-4, and IL-17A, and a greater percentage of sBc-arTregs produced IL-17A, but far less than those percentages produced by similarly stimulated conventional CD4+ T cells (Tconvs). The majority of cytokine-expressing arTregs were FOXP3+. Percentages of CXCR3 and CCR4, but not CCR6, increased after sBc and moDC stimulation. Using a 770-gene Nanostring panel and unsupervised clustering, sBc- and moDCs-arTregs were found to be mostly similar, but distinct from primary Tregs and further separated from Tconvs. Thus, arTregs expanded by sBcs or moDCs are phenotypically similar, likely stable, and had features that may enable them to traffic to inflammatory sites, e.g. transplanted organs undergoing alloimmune attack.
- Published
- 2018
- Full Text
- View/download PDF
45. Monitoring the operationally tolerant liver allograft recipient
- Author
-
Angus W. Thomson, A. Castellaneta, George V. Mazariegos, Michael E. de Vera, and Navdeep Nayyar
- Subjects
Graft Rejection ,medicine.medical_specialty ,Time Factors ,Biopsy ,medicine.medical_treatment ,Drug Administration Schedule ,Drug withdrawal ,Liver Function Tests ,Predictive Value of Tests ,T-Lymphocyte Subsets ,medicine ,Humans ,Transplantation, Homologous ,Immunology and Allergy ,Registries ,Biomarker Analysis ,Intensive care medicine ,Transplantation ,medicine.diagnostic_test ,Human liver ,business.industry ,Graft Survival ,Immunosuppression ,Microarray Analysis ,medicine.disease ,Liver Transplantation ,Treatment Outcome ,Biomarker (medicine) ,Transplantation Tolerance ,business ,Biomarkers ,Immunosuppressive Agents ,Blood sampling - Abstract
Purpose of review A fundamental goal in transplantation is the establishment of allograft function without ongoing immunosuppression. Robust allograft tolerance has been established in experimental transplantation models, whereas clinical operational tolerance has been described most frequently following human liver transplantation. Recent findings Clinical assessment of tolerance has been limited to laboratory evaluation of organ function. Additional tools include graft monitoring through biopsy and blood sampling for biomarker analysis. Current biomarkers under assessment in recent years include dendritic cell subsets, regulatory T cells, antidonor antibodies, and gene polymorphisms. Emerging microarray analysis that is being prospectively validated will also be reviewed. Recent findings A further tool in the characterization of the tolerant patient will be the accurate enrollment of such patients into a multicenter registry that will prospectively follow the natural history of the patient withdrawn from immunosuppression and help facilitate the entry of interested patients to mechanistic and immune monitoring trials. The International Solid Organ Transplant Tolerance Registry (www.transplant-tolerance.org) will be briefly described. Summary Effective biomarker characterization of the operationally tolerant liver allograft recipient would allow earlier, well tolerated, prospective drug withdrawal with the goal of extending the potential benefits of drug minimization to an increasing number of patients in a more predictable fashion.
- Published
- 2010
- Full Text
- View/download PDF
46. Composite Tissue Vasculopathy and Degeneration Following Multiple Episodes of Acute Rejection in Reconstructive Transplantation
- Author
-
Angus W. Thomson, Jignesh V. Unadkat, Stefan Schneeberger, Mario G. Solari, E. H. Horibe, Kia M. Washington, Vijay S. Gorantla, C. Goldbach, G. M. Cooper, and W. P. Andrew Lee
- Subjects
Male ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Fibrosis ,Rats, Inbred BN ,Animals ,Immunology and Allergy ,Medicine ,Pharmacology (medical) ,Dexamethasone ,Transplantation ,business.industry ,Vascular disease ,medicine.disease ,Muscle atrophy ,Hindlimb ,Rats ,Specific Pathogen-Free Organisms ,Transplantation, Isogeneic ,medicine.anatomical_structure ,Rats, Inbred Lew ,Circulatory system ,Cyclosporine ,medicine.symptom ,business ,Allotransplantation ,Blood vessel ,medicine.drug - Abstract
Transplant vasculopathy has not been systematically investigated in composite tissue allotransplantation (CTA). The impact of multiple acute rejections (ARs) on long-term graft outcomes in reconstructive transplantation remains unknown. This study in a rat hind-limb allotransplantation model systematically analyzes vasculopathy and tissue-specific pathological changes secondary to multiple AR episodes. LEW rats were transplanted with BN rat hind limbs and treated as follows: Group 1 (Iso): isografts. Group 2 (CsA): Cyclosporine (CsA) qd; Group 3 (mult AR): CsA and dexamethasone only when AR was observed. No AR was observed in Groups 1 and 2. Multiple AR were observed in Group 3, and each episode was completely reversed (clinically) with pulsed CsA + dexamethasone treatment. Group 3 animals demonstrated significant vascular lesions along with skin and muscle atrophy, upregulation of profibrotic gene expression and fibrosis when compared to Groups 1 and 2. In addition, allograft bone was sclerotic, weak and prone to malunion and nonunion. Interestingly, vasculopathy was a late finding, whereas muscle atrophy with macrophage infiltration was seen early, after only a few AR episodes. Taken together, multiple AR episodes lead to vasculopathy and tissue-specific pathology in CTA. This is the first evidence of 'composite tissue vasculopathy and degeneration (CTVD)' in CTA.
- Published
- 2010
- Full Text
- View/download PDF
47. Mammalian Target of Rapamycin Inhibition and Alloantigen-Specific Regulatory T Cells Synergize To Promote Long-Term Graft Survival in Immunocompetent Recipients
- Author
-
Benjamin M. Matta, Tina L. Sumpter, Yoram Vodovotz, Zhiliang Wang, Giorgio Raimondi, Natasha Corbitt, Angus W. Thomson, and Mahesh Pillai
- Subjects
Isoantigens ,medicine.medical_treatment ,T cell ,Immunology ,T-Cell Antigen Receptor Specificity ,Protein Serine-Threonine Kinases ,Biology ,T-Lymphocytes, Regulatory ,Article ,Proinflammatory cytokine ,Mice ,Adjuvants, Immunologic ,In vivo ,medicine ,Animals ,Humans ,Immunology and Allergy ,Heart transplantation ,TOR Serine-Threonine Kinases ,Graft Survival ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Immunosuppression ,In vitro ,Mice, Inbred C57BL ,Treatment Outcome ,medicine.anatomical_structure ,Lymphocyte Transfusion ,Heart Transplantation ,Immunocompetence - Abstract
Minimization of immunosuppression and donor-specific tolerance to MHC-mismatched organ grafts are important clinical goals. The therapeutic potential of regulatory T cells (Tregs) has been demonstrated, but conditions for optimizing their in vivo function posttransplant in nonlymphocyte-depleted hosts remain undefined. In this study, we address mechanisms through which inhibition of the mammalian target of rapamycin (Rapa) synergizes with alloantigen-specific Treg (AAsTreg) to permit long-term, donor-specific heart graft survival in immunocompetent hosts. Crucially, immature allogeneic dendritic cells allowed AAsTreg selection in vitro, with minimal expansion of unwanted (Th17) cells. The rendered Treg potently inhibited T cell proliferation in an Ag-specific manner. However, these AAsTreg remained unable to control T cells stimulated by allogeneic mature dendritic cells, a phenomenon dependent on the release of proinflammatory cytokines. In vivo, Rapa administration reduced danger-associated IL-6 production, T cell proliferation, and graft infiltration. Based on these observations, AAsTreg were administered posttransplant (day 7) in combination with a short course of Rapa and rendered >80% long-term (>150 d) graft survival, a result superior to that achieved with polyclonal Treg. Moreover, graft protection was alloantigen-specific. Significantly, long-term graft survival was associated with alloreactive T cell anergy. These findings delineate combination of transient mammalian target of Rapa inhibition with appropriate AAsTreg selection as an effective approach to promote long-term organ graft survival.
- Published
- 2009
- Full Text
- View/download PDF
48. NOD2 Ligation Subverts IFN-α Production by Liver Plasmacytoid Dendritic Cells and Inhibits Their T Cell Allostimulatory Activity via B7-H1 Up-Regulation
- Author
-
Tina L. Sumpter, Angus W. Thomson, Daisuke Tokita, Lieping Chen, and A. Castellaneta
- Subjects
Male ,Adoptive cell transfer ,T-Lymphocytes ,T cell ,Blotting, Western ,Immunology ,Nod2 Signaling Adaptor Protein ,Enzyme-Linked Immunosorbent Assay ,Spleen ,Biology ,Lymphocyte Activation ,B7-H1 Antigen ,Article ,Interferon-gamma ,Mice ,chemistry.chemical_compound ,Adjuvants, Immunologic ,NF-KappaB Inhibitor alpha ,medicine ,Animals ,Immunology and Allergy ,CD86 ,Mice, Inbred BALB C ,Membrane Glycoproteins ,Reverse Transcriptase Polymerase Chain Reaction ,hemic and immune systems ,Dendritic Cells ,Flow Cytometry ,Acquired immune system ,Molecular biology ,Up-Regulation ,Mice, Inbred C57BL ,IκBα ,medicine.anatomical_structure ,Liver ,chemistry ,Interferon Regulatory Factors ,B7-1 Antigen ,I-kappa B Proteins ,Lymphocyte Culture Test, Mixed ,Peptides ,Acetylmuramyl-Alanyl-Isoglutamine ,Muramyl dipeptide ,Ex vivo - Abstract
The nucleotide-binding oligomerization domain (NOD)2/CARD15 protein, which senses muramyl dipeptide (MDP), a product of bacterial peptidoglycan, appears to play an important role in regulating intestinal immunity. Although the liver is exposed to gut-derived MDP, the influence of NOD2 ligation on hepatic APC, in particular dendritic cells (DC), is unknown. Freshly isolated mouse liver and spleen plasmacytoid (p)DC expressed higher levels of NOD2 message than conventional myeloid (m)DC. Following MDP stimulation in vivo, liver pDC, but not mDC, up-regulated expression of IFN regulatory factor 4 (IRF-4), a negative regulator of TLR signaling, and induced less allogeneic T cell proliferation and IFN-γ production. The adoptive transfer of liver pDC from MDP-treated mice failed to prime allogeneic T cells in vivo. By contrast, splenic DC IRF-4 levels and T cell stimulatory activity remained unchanged. Liver pDC from MDP-stimulated mice also displayed greater IκBα, cell surface B7-H1, and B7-H1 relative to CD86 than control liver pDC. No similar effects were observed for liver mDC or spleen DC. Absence of B7-H1 on liver pDC reversed the inhibitory effect of MDP. After ex vivo stimulation with LPS or CpG, liver pDC but not mDC from MDP-treated animals secreted less IL-12p70, IL-6, and TNF-α and induced weaker allogeneic T cell proliferation than those from controls. Moreover, CpG-stimulated liver pDC from MDP-treated mice secreted less IFN-α than their splenic counterparts, and systemic levels of IFN-α were reduced in MDP-treated animals after CpG administration. These findings suggest that differential effects of NOD2 ligation on liver pDC may play a role in regulating hepatic innate and adaptive immunity.
- Published
- 2009
- Full Text
- View/download PDF
49. IL-33 broadens its repertoire to affect DC
- Author
-
Heth R. Turnquist and Angus W. Thomson
- Subjects
medicine.diagnostic_test ,Immunology ,Arthritis ,Biology ,Ligand (biochemistry) ,Acquired immune system ,medicine.disease ,Flow cytometry ,Interleukin 33 ,Cell culture ,medicine ,Immunology and Allergy ,Receptor ,Function (biology) - Abstract
IL-33 is a novel multi-functional IL-1 family member that, in contrast to other family members, is associated with Th2 responses. IL-33 signals via a heterodimer composed of its receptor, IL-1 receptor-like-1 (IL-1RL1), more commonly known as ST2L, and the IL-1R accessory protein. ST2L is expressed by endothelial cells, mast cells, basophils, Th2 cells, and DC. IL-33 has been associated with several immune-mediated disorders, including asthma, arthritis, and inflammatory bowel disease. In contrast, there is evidence that IL-33 can inhibit atherosclerosis development. A report in this issue of the European Journal of Immunology reveals a novel function of IL-33: the ability to promote myeloid DC generation in murine BM cell cultures, by triggering GM-CSF production by other BM cells, likely basophils. DC generated in the presence of IL-33 are maturation resistant, with only minimal T-cell stimulatory ability, associated with comparatively high levels of programmed death receptor ligand expression. This commentary discusses several questions raised by these findings, and provides a basis for further evaluation of IL-33 and ST2L in regulation of APC generation and function in both innate and adaptive immunity.
- Published
- 2009
- Full Text
- View/download PDF
50. Allospecific CD154+ T Cells Associate with Rejection Risk After Pediatric Liver Transplantation
- Author
-
Chethan Ashokkumar, Jennifer Dobberstein, Ronald Jaffe, Brandon W. Higgs, Geoffrey Bond, Rakesh Sindhi, Kyle Soltys, Anjan Talukdar, George V. Mazariegos, Adriana Zeevi, Patrick Wilson, Qing Sun, Anthony J. Demetris, Angus W. Thomson, and Janine E. Janosky
- Subjects
Graft Rejection ,Male ,T-Lymphocytes ,medicine.medical_treatment ,CD40 Ligand ,chemical and pharmacologic phenomena ,Liver transplantation ,Article ,Flow cytometry ,Cohort Studies ,Antigens, CD ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,CTLA-4 Antigen ,Pharmacology (medical) ,CD154 ,Child ,Transplantation ,CD40 ,biology ,medicine.diagnostic_test ,business.industry ,Incidence (epidemiology) ,Alloimmunity ,hemic and immune systems ,Tacrolimus ,Liver Transplantation ,surgical procedures, operative ,Child, Preschool ,Immunology ,biology.protein ,Female ,business ,Immunologic Memory - Abstract
Antigen-specific T cells, which express CD154 rapidly, but remain untested in alloimmunity, were measured with flow cytometry in 16-h MLR of 58 identically-immunosuppressed children with liver transplantation (LTx), to identify Rejectors (who had experienced biopsy-proven rejection within 60 days posttransplantation). Thirty-one children were sampled once, cross-sectionally. Twenty-seven children were sampled longitudinally, pre-LTx, and at 1-60 and 61-200 days after LTx. Results were correlated with proliferative alloresponses measured by CFSE-dye dilution (n = 23), and CTLA4, a negative T-cell costimulator, which antagonizes CD154-mediated effects (n = 31). In cross-sectional observations, logistic regression and leave-one-out cross-validation identified donor-specific, CD154 + T-cytotoxic (Tc)-memory cells as best associated with rejection outcomes. In the longitudinal cohort, (1) the association between CD154 + Tc-memory cells and rejection outcomes was replicated with sensitivity/specificity 92.3%/84.6% for observations at 1-60 days, and (2) elevated pre-LTx CD154 + Tc-memory cell responses were associated with significantly increased incidence (p = 0.02) and hazard (HR = 7.355) of rejection in survival/proportional hazard analysis. CD154 expression correlated with proliferative alloresponses (r = 0.835, p = 7.1e-07), and inversely with CTLA4 expression of allospecific CD154 + Tc-memory cells (r =-0.706, p = 3.0e-05). Allospecific CD154 + T-helper-memory cells, not CD154 + Tc-memory, were inhibited by increasing Tacrolimus concentrations (p = 0.026). Collectively, allospecific CD154 + T cells provide an estimate of rejection risk in children with LTx.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.