1
|
Singh RB, Cho W, Liu C, Naderi A, Surico PL, Kahale F, Dohlman TH, Chauhan SK, Dana R. Immunopathological mechanisms and clinical manifestations of ocular graft-versus-host disease following hematopoietic stem cell transplantation. Bone Marrow Transplant 2024; 59:1049-1056. [PMID: 38822141 DOI: 10.1038/s41409-024-02321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Graft-versus-host disease is among the most common clinical complications following allogeneic hematopoietic stem cell transplantation. It causes inflammation-mediated destruction and dysfunction of various organ systems including ocular tissues in 60-90% of the patients and is termed ocular GVHD (oGVHD). In oGVHD, donor-derived T-cells recognize host antigens as foreign, resulting in immune dysregulation, inflammation and fibrosis of lacrimal glands, meibomian glands, cornea, and conjunctiva. The clinical presentation in oGVHD patients range from mild dry eye symptoms to catastrophic inflammation mediated pathological changes which can cause corneal perforation and blindness. In this review article, we provide detailed insights into the impact of mucosal barrier disruption, the afferent and efferent phases of immunological response involving activation of antigen presenting cells and T cells, respectively. We evaluate the evidence outlining the effector phase of the disease leading to cellular destruction and eventually fibrosis in patients with oGVHD. Finally, we discuss the well-established criteria for the diagnosis of oGVHD.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Wonkyung Cho
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Catherine Liu
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Amirreza Naderi
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Pier Luigi Surico
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Francesca Kahale
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Thomas H Dohlman
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Troise D, Infante B, Mercuri S, Catalano V, Ranieri E, Stallone G. Dendritic Cells: A Bridge between Tolerance Induction and Cancer Development in Transplantation Setting. Biomedicines 2024; 12:1240. [PMID: 38927447 PMCID: PMC11200833 DOI: 10.3390/biomedicines12061240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Dendritic cells (DCs) are a heterogeneous group of antigen-presenting cells crucial for fostering allograft tolerance while simultaneously supporting host defense against infections and cancer. Within the tumor microenvironment, DCs can either mount an immune response against cancer cells or foster immunotolerance, presenting a dual role. In immunocompromised individuals, posttransplant malignancies pose a significant health concern, with DCs serving as vital players in immune responses against cancer cells. Both recipient- and donor-derived DCs play a critical role in the rejection process, infiltrating the transplanted organ and sustaining T-cell responses. The use of immunosuppressive drugs represents the predominant approach to control this immunological barrier in transplanted organs. Evidence has shed light on the immunopharmacology of these drugs and novel strategies for manipulating DCs to promote allograft survival. Therefore, comprehending the mechanisms underlying this intricate microenvironment and the effects of immunosuppressive therapy on DCs is crucial for developing targeted therapies to reduce graft failure rates. This review will delve into the fundamental immunobiology of DCs and provide a detailed exploration of their clinical significance concerning alloimmune responses and posttransplant malignancies.
Collapse
Affiliation(s)
- Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Valeria Catalano
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Elena Ranieri
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
3
|
Gray G, Scroggins DG, Wilson KT, Scroggins SM. Cellular Immunotherapy in Mice Prevents Maternal Hypertension and Restores Anti-Inflammatory Cytokine Balance in Maternal and Fetal Tissues. Int J Mol Sci 2023; 24:13594. [PMID: 37686399 PMCID: PMC10487605 DOI: 10.3390/ijms241713594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Preeclampsia is the leading cause of maternal-fetal morbidity worldwide. The concept that persistent feto-placental intolerance is important in the pathogenesis of preeclampsia (PreE) has been demonstrated by our lab and others. Arginine vasopressin (AVP) infusion during pregnancy induces cardiovascular, renal, and T helper (TH) cell alterations in mice consistent with human PreE. In addition to their conventional immuno-stimulatory role, dendritic cells (DCs) also play a vital role in immune tolerance. In contrast to conventional DCs, regulatory DCs (DCregs) express low levels of co-stimulatory markers, produce anti-inflammatory cytokines, induce T regulatory (Treg) cells, and promote tolerance. In mice, DCregs prevent pro-inflammatory responses and induce antigen-specific tolerance. Given these known functions of DCregs, we hypothesize that DCregs will prevent the development of AVP-induced PreE in mice. C57BL/6J females were infused with AVP (24 ng/h) or saline throughout gestation via an osmotic minipump. Bone-marrow-derived DCregs were injected into AVP-infused dams at the time of the pump implantation or on gestational day (GD) 7. The blood pressure of the mice was taken throughout their pregnancy. The maternal urine proteins and TH-associated cytokines in maternal and fetal tissues were measured on GD 18. The treatment with DCregs effectively prevented the elevation of maternal blood pressure, proteinuria, and fetal growth restriction that were observed in AVP-infused dams. Furthermore, we noted a reduction in the pro-inflammatory TH-associated cytokines IFNγ and IL-17, while anti-inflammatory cytokines IL-4, IL-10, and TGFβ showed an increase following DCreg treatment. These outcomes provide strong evidence supporting the potential of DCregs as a valuable therapeutic approach in addressing PreE.
Collapse
Affiliation(s)
- Gabrielle Gray
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Douglas G. Scroggins
- School of Medicine, Department of Biomedical Sciences, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Katlin T. Wilson
- School of Medicine, Department of Biomedical Sciences, University of Minnesota Duluth, Duluth, MN 55812, USA
| | - Sabrina M. Scroggins
- School of Medicine, Department of Biomedical Sciences, University of Minnesota Duluth, Duluth, MN 55812, USA
| |
Collapse
|
4
|
Shi H, Gao L, Zhang W, Jiang M. Long non-coding RNAs regulate treatment outcome in leukemia: What have we learnt recently? Cancer Med 2023. [PMID: 37148556 DOI: 10.1002/cam4.6027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/03/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023] Open
Abstract
Leukemia is a group of highly heterogeneous and life-threatening blood cancers that originate from abnormal hematopoietic stem cells. Multiple treatments are approved for leukemia, including chemotherapy, targeted therapy, hematopoietic stem cell transplantation, radiation therapy, and immunotherapy. Unfortunately, therapeutic resistance occurs in a substantial proportion of patients and greatly compromises the treatment efficacy of leukemia, resulting in relapse and mortality. The abnormal activity of receptor tyrosine kinases, cell membrane transporters, intracellular signal transducers, transcription factors, and anti-apoptotic proteins have been shown to contribute to the emergence of therapeutic resistance. Despite these findings, the exact mechanisms of treatment resistance are still not fully understood, which limits the development of effective measures to overcome it. Long non-coding RNAs (lncRNA) are a class of regulatory molecules that are gaining increasing attention, and lncRNA-mediated regulation of therapeutic resistance against multiple drugs for leukemia is being revealed. These dysregulated lncRNAs not only serve as potential targets to reduce resistance but also might improve treatment response prediction and individualized treatment decision. Here, we summarize the recent findings on lncRNA-mediated regulation of therapeutic resistance in leukemia and discuss future perspectives on how to make use of the dysregulated lncRNAs in leukemia to improve treatment outcome.
Collapse
Affiliation(s)
- Huiping Shi
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Liang Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Weili Zhang
- Department of Gastroenterology, Xiangcheng People's Hospital, Suzhou, Jiangsu, People's Republic of China
| | - Min Jiang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| |
Collapse
|
5
|
A Multiparameter Prognostic Risk Score of Chronic Graft-versus-Host Disease Based on CXCL10 and Plasmacytoid Dendritic Cell Levels in the Peripheral Blood at 3 Months after Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2023; 29:302.e1-302.e8. [PMID: 36796518 DOI: 10.1016/j.jtct.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/26/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
Chronic GVHD (cGVHD) is the major cause of long-term morbidity after allogeneic hematopoietic stem cell transplantation (HSCT). There are no biomarkers that can consistently predict its occurrence. We aimed to evaluate whether numbers of antigen-presenting cell subsets in peripheral blood (PB) or serum chemokine concentrations are biomarkers of cGVHD occurrence. The study cohort comprised 101 consecutive patients undergoing allogeneic HSCT between January 2007 and 2011. cGVHD was diagnosed by both modified Seattle criteria and National Institutes of Health (NIH) criteria. Multicolor flow cytometry was used to determine the number of PB myeloid dendritic cells (DCs), plasmacytoid DCs, CD16+ DCs, and CD16+ and CD16- monocytes, as well as CD4+ and CD8+ T cells, CD56+ natural killer cells, and CD19+ B cells. Serum concentrations of CXCL8, CXCL10, CCL2, CCL3, CCL4, and CCL5 were measured by a cytometry bead array assay. At a median of 60 days after enrollment, 37 patients had developed cGVHD. Patients with cGVHD and those without cGVHD had comparable clinical characteristics. However, previous acute GVHD (aGVHD) was strongly correlated with later cGVHD (57% versus 24%, respectively; P = .0024). Each potential biomarker was screened for its association with cGVHD using the Mann-Whitney U test. Biomarkers that differed significantly (P < .05) between patients with cGVHD and those without cGVHD were analyzed by receiver operating characteristic (ROC) curve analysis to select the variables predicting cGVHD with an area under the ROC curve (AUC) >.5 and a P value <.05. A multivariate Fine-Gray model identified the following variables as independently associated with the risk of cGVHD: CXCL10 ≥592.650 pg/mL (hazard ratio [HR], 2.655; 95% confidence interval [CI], 1.298 to 5.433; P = .008), pDC ≥2.448/μL (HR, .286; 95% CI, .142 to .577; P < .001) and previous aGVHD (HR, 2.635; 95% CI, 1.298 to 5.347; P = .007). A risk score was derived based on the weighted coefficients of each variable (2 points each), resulting in the identification of 4 cohorts of patients (scores of 0, 2, 4, and 6). In a competing risk analysis to stratify patients at differing risk levels of cGVHD, the cumulative incidence of cGVHD was 9.7%, 34.3%, 57.7%, and 100% in patients with scores of 0, 2, 4, and 6, respectively (P < .0001). The score could nicely stratify the patients based on the risk of extensive cGVHD as well as NIH-based global and moderate to severe cGVHD. Based on ROC analysis, the score could predict the occurrence of cGVHD with an AUC of .791 (95% CI, .703 to .880; P < .001). Finally, a cutoff score ≥4 was identified as the optimal cutoff by Youden J index with a sensitivity of 57.1% and a specificity of 85.0%. A multiparameter score including a history of previous aGVHD, serum CXCL10 concentration, and number of pDCs in the PB at 3 months post-HSCT stratify patients at varying risk levels of cGVHD. However, the score needs to be validated in a much larger independent and possibly multicenter cohort of patients undergoing transplantation from different donor types and with distinct GVHD prophylaxis regimens.
Collapse
|
6
|
Shaikh H, Pezoldt J, Mokhtari Z, Gamboa Vargas J, Le DD, Peña Mosca J, Arellano Viera E, Kern MA, Graf C, Beyersdorf N, Lutz MB, Riedel A, Büttner-Herold M, Zernecke A, Einsele H, Saliba AE, Ludewig B, Huehn J, Beilhack A. Fibroblastic reticular cells mitigate acute GvHD via MHCII-dependent maintenance of regulatory T cells. JCI Insight 2022; 7:154250. [PMID: 36227687 DOI: 10.1172/jci.insight.154250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/07/2022] [Indexed: 12/15/2022] Open
Abstract
Acute graft versus host disease (aGvHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT) inflicted by alloreactive T cells primed in secondary lymphoid organs (SLOs) and subsequent damage to aGvHD target tissues. In recent years, Treg transfer and/or expansion has emerged as a promising therapy to modulate aGvHD. However, cellular niches essential for fostering Tregs to prevent aGvHD have not been explored. Here, we tested whether and to what extent MHC class II (MHCII) expressed on Ccl19+ fibroblastic reticular cells (FRCs) shape the donor CD4+ T cell response during aGvHD. Animals lacking MHCII expression on Ccl19-Cre-expressing FRCs (MHCIIΔCcl19) showed aberrant CD4+ T cell activation in the effector phase, resulting in exacerbated aGvHD that was associated with significantly reduced expansion of Foxp3+ Tregs and invariant NK T (iNKT) cells. Skewed Treg maintenance in MHCIIΔCcl19 mice resulted in loss of protection from aGvHD provided by adoptively transferred donor Tregs. In contrast, although FRCs upregulated costimulatory surface receptors, and although they degraded and processed exogenous antigens after myeloablative irradiation, FRCs were dispensable to activate alloreactive CD4+ T cells in 2 mouse models of aGvHD. In summary, these data reveal an immunoprotective, MHCII-mediated function of FRC niches in secondary lymphoid organs (SLOs) after allo-HCT and highlight a framework of cellular and molecular interactions that regulate CD4+ T cell alloimmunity.
Collapse
Affiliation(s)
- Haroon Shaikh
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Joern Pezoldt
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Zeinab Mokhtari
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Juan Gamboa Vargas
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Duc-Dung Le
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Josefina Peña Mosca
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Estibaliz Arellano Viera
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Michael Ag Kern
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Caroline Graf
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Niklas Beyersdorf
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany.,Institute for Virology and Immunobiology, Würzburg University, Würzburg, Germany
| | - Manfred B Lutz
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany.,Institute for Virology and Immunobiology, Würzburg University, Würzburg, Germany
| | - Angela Riedel
- Mildred Scheel Early Career Centre, University Hospital of Würzburg, Würzburg, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection (HZI), Würzburg, Germany
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research (IZKF), Experimental Stem Cell Transplantation Laboratory, and.,Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| |
Collapse
|
7
|
Lalic H, Aurer I, Batinic D, Visnjic D, Smoljo T, Babic A. Bendamustine: A review of pharmacology, clinical use and immunological effects (Review). Oncol Rep 2022; 47:114. [PMID: 35506458 PMCID: PMC9100486 DOI: 10.3892/or.2022.8325] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 11/15/2022] Open
Abstract
Bendamustine is an alkylating agent classified into the group of nitrogen mustard analogues, synthesized almost sixty years ago. It was registered in former East Germany in 1971 and approved by the US Food and Drug Administration in 2008 for treatment of chronic lymphocytic leukemia and indolent B-cell non-Hodgkin lymphoma. Considering its beneficial properties in the therapy of relapsed or refractory hematological malignancies, synergistic effects with other antineoplastic agents and increasing recent reports on its immunomodulatory effects, bendamustine has once again gained its justified attention. The uniqueness of bendamustine-mediated effects should be observed keeping in mind its distinctive structure with structural similarities to both alkylating agents and purine analogs. In the present review, the current knowledge on the use of bendamustine in oncology, its pharmacokinetics, mechanism of action and toxicity was summarized. In addition, its immune-modulating effects that have not been fully elucidated so far are emphasized, hoping to encourage further investigations of this unique drug.
Collapse
Affiliation(s)
- Hrvoje Lalic
- Laboratory of Cell Biology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Igor Aurer
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, 10 000 Zagreb, Croatia
| | - Drago Batinic
- Department of Physiology, University of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Dora Visnjic
- Laboratory of Cell Biology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Tomislav Smoljo
- Laboratory of Cell Biology and Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10 000 Zagreb, Croatia
| | - Antonija Babic
- Department of Laboratory Immunology, Clinical Department of Laboratory Diagnostics, University Hospital Center Zagreb, 10 000 Zagreb, Croatia
| |
Collapse
|
8
|
Adoptive transfer of ex vivo expanded regulatory T-cells improves immune cell engraftment and therapy-refractory chronic GvHD. Mol Ther 2022; 30:2298-2314. [PMID: 35240319 DOI: 10.1016/j.ymthe.2022.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/09/2022] [Accepted: 02/25/2022] [Indexed: 11/22/2022] Open
Abstract
Graft-versus-Host-Disease (GvHD) is still the major non-relapse, life-limiting complication following hematopoietic stem cell transplantation. Modern pharmacologic immunosuppression is often insufficient and associated with significant side effects. Novel treatment strategies now include adoptive transfer of ex vivo expanded regulatory T-cells (Tregs), but their efficacy in chronic GvHD is unknown. We treated three children suffering from severe, therapy-refractory GvHD with polyclonally expanded Tregs generated from the original stem cell donor. Third-line maintenance immunosuppression was tapered to Cyclosporin A and low-dose steroids shortly before cell transfer. Regular follow-up included assessment of the subjective and objective clinical development, safety parameters and in-depth immune monitoring. All patients showed marked clinical improvement with substantially reduced GvHD activity. Laboratory follow-up showed a significant enhancement of the immunologic engraftment including lymphocytes and dendritic cells. Monitoring the fate of Tregs by next generation sequencing demonstrated clonal expansion. In summary, adoptive transfer of Tregs was well tolerated and able to modulate an established undesired T-cell mediated allo-response. Although no signs of overimmunosuppression were detectable, treatment of patients with invasive opportunistic infections should be undertaken with caution. Further controlled studies, are necessary to confirm these encouraging effects and eventually pave the way for adoptive Treg therapy in chronic GvHD.
Collapse
|
9
|
Nicholls J, Cao B, Le Texier L, Xiong LY, Hunter CR, Llanes G, Aguliar EG, Schroder WA, Phipps S, Lynch JP, Cao H, Heazlewood SY, Williams B, Clouston AD, Nefzger CM, Polo JM, Nilsson SK, Blazar BR, MacDonald KPA. Bone Marrow Regulatory T Cells Are a Unique Population, Supported by Niche-Specific Cytokines and Plasmacytoid Dendritic Cells, and Required for Chronic Graft-Versus-Host Disease Control. Front Cell Dev Biol 2021; 9:737880. [PMID: 34631716 PMCID: PMC8493124 DOI: 10.3389/fcell.2021.737880] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Regulatory T cell (Treg) reconstitution is essential for reestablishing tolerance and maintaining homeostasis following stem-cell transplantation. We previously reported that bone marrow (BM) is highly enriched in autophagy-dependent Treg and autophagy disruption leads to a significant Treg loss, particularly BM-Treg. To correct the known Treg deficiency observed in chronic graft-versus-host disease (cGVHD) patients, low dose IL-2 infusion has been administered, substantially increasing peripheral Treg (pTreg) numbers. However, as clinical responses were only seen in ∼50% of patients, we postulated that pTreg augmentation was more robust than for BM-Treg. We show that BM-Treg and pTreg have distinct characteristics, indicated by differential transcriptome expression for chemokine receptors, transcription factors, cell cycle control of replication and genes linked to Treg function. Further, BM-Treg were more quiescent, expressed lower FoxP3, were highly enriched for co-inhibitory markers and more profoundly depleted than splenic Treg in cGVHD mice. In vivo our data are consistent with the BM and not splenic microenvironment is, at least in part, driving this BM-Treg signature, as adoptively transferred splenic Treg that entered the BM niche acquired a BM-Treg phenotype. Analyses identified upregulated expression of IL-9R, IL-33R, and IL-7R in BM-Treg. Administration of the T cell produced cytokine IL-2 was required by splenic Treg expansion but had no impact on BM-Treg, whereas the converse was true for IL-9 administration. Plasmacytoid dendritic cells (pDCs) within the BM also may contribute to BM-Treg maintenance. Using pDC-specific BDCA2-DTR mice in which diptheria toxin administration results in global pDC depletion, we demonstrate that pDC depletion hampers BM, but not splenic, Treg homeostasis. Together, these data provide evidence that BM-Treg and splenic Treg are phenotypically and functionally distinct and influenced by niche-specific mediators that selectively support their respective Treg populations. The unique properties of BM-Treg should be considered for new therapies to reconstitute Treg and reestablish tolerance following SCT.
Collapse
Affiliation(s)
- Jemma Nicholls
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Benjamin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Laetitia Le Texier
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Laura Yan Xiong
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christopher R. Hunter
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Genesis Llanes
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Ethan G. Aguliar
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Wayne A. Schroder
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Simon Phipps
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jason P. Lynch
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Huimin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Shen Y. Heazlewood
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Brenda Williams
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | | | - Christian M. Nefzger
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jose M. Polo
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Susan K. Nilsson
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organization, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant and Cellular Therapies, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kelli P. A. MacDonald
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
10
|
Nakano R, Tran LM, Geller DA, Macedo C, Metes DM, Thomson AW. Dendritic Cell-Mediated Regulation of Liver Ischemia-Reperfusion Injury and Liver Transplant Rejection. Front Immunol 2021; 12:705465. [PMID: 34262574 PMCID: PMC8273384 DOI: 10.3389/fimmu.2021.705465] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/11/2021] [Indexed: 12/24/2022] Open
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.
Collapse
Affiliation(s)
- Ryosuke Nakano
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Lillian M. Tran
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - David A. Geller
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Liver Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Camila Macedo
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Diana M. Metes
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angus W. Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Molina MS, Hoffman EA, Stokes J, Kummet N, Smith KA, Baker F, Zúñiga TM, Simpson RJ, Katsanis E. Regulatory Dendritic Cells Induced by Bendamustine Are Associated With Enhanced Flt3 Expression and Alloreactive T-Cell Death. Front Immunol 2021; 12:699128. [PMID: 34249005 PMCID: PMC8264365 DOI: 10.3389/fimmu.2021.699128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 12/30/2022] Open
Abstract
The growth factor Flt3 ligand (Flt3L) is central to dendritic cell (DC) homeostasis and development, controlling survival and expansion by binding to Flt3 receptor tyrosine kinase on the surface of DCs. In the context of hematopoietic cell transplantation, Flt3L has been found to suppress graft-versus-host disease (GvHD), specifically via host DCs. We previously reported that the pre-transplant conditioning regimen consisting of bendamustine (BEN) and total body irradiation (TBI) results in significantly reduced GvHD compared to cyclophosphamide (CY)+TBI. Pre-transplant BEN+TBI conditioning was also associated with greater Flt3 expression among host DCs and an accumulation of pre-cDC1s. Here, we demonstrate that exposure to BEN increases Flt3 expression on both murine bone marrow-derived DCs (BMDCs) and human monocyte-derived DCs (moDCs). BEN favors development of murine plasmacytoid DCs, pre-cDC1s, and cDC2s. While humans do not have an identifiable equivalent to murine pre-cDC1s, exposure to BEN resulted in decreased plasmacytoid DCs and increased cDC2s. BEN exposure and heightened Flt3 signaling are associated with a distinct regulatory phenotype, with increased PD-L1 expression and decreased ICOS-L expression. BMDCs exposed to BEN exhibit diminished pro-inflammatory cytokine response to LPS and induce robust proliferation of alloreactive T-cells. These proliferative alloreactive T-cells expressed greater levels of PD-1 and underwent increased programmed cell death as the concentration of BEN exposure increased. Alloreactive CD4+ T-cell death may be attributable to pre-cDC1s and provides a potential mechanism by which BEN+TBI conditioning limits GvHD and yields T-cells tolerant to host antigen.
Collapse
Affiliation(s)
- Megan S Molina
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States
| | - Emely A Hoffman
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Nicole Kummet
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Molecular & Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Kyle A Smith
- Department of Physiology, University of Arizona, Tucson, AZ, United States.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Forrest Baker
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Tiffany M Zúñiga
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Richard J Simpson
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States.,The University of Arizona Cancer Center, Tucson, AZ, United States
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States.,The University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Medicine, University of Arizona, Tucson, AZ, United States.,Department of Pathology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
12
|
Lin J, Wang H, Liu C, Cheng A, Deng Q, Zhu H, Chen J. Dendritic Cells: Versatile Players in Renal Transplantation. Front Immunol 2021; 12:654540. [PMID: 34093544 PMCID: PMC8170486 DOI: 10.3389/fimmu.2021.654540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022] Open
Abstract
Dendritic cells (DCs) induce and regulate adaptive immunity through migrating and maturing in the kidney. In this procedure, they can adopt different phenotypes—rejection-associated DCs promote acute or chronic injury renal grafts while tolerogenic DCs suppress the overwhelmed inflammation preventing damage to renal functionality. All the subsets interact with effector T cells and regulatory T cells (Tregs) stimulated by the ischemia–reperfusion procedure, although the classification corresponding to different effects remains controversial. Thus, in this review, we discuss the origin, maturation, and pathological effects of DCs in the kidney. Then we summarize the roles of divergent DCs in renal transplantation: taking both positive and negative stages in ischemia–reperfusion injury (IRI), switching phenotypes to induce acute or chronic rejection, and orchestrating surface markers for allograft tolerance via alterations in metabolism. In conclusion, we prospect that multidimensional transcriptomic analysis will revolute researches on renal transplantation by addressing the elusive mononuclear phagocyte classification and providing a holistic view of DC ontogeny and subpopulations.
Collapse
Affiliation(s)
- Jinwen Lin
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Disease, Institute of Nephrology, Zhejiang University, Hangzhou, China.,The Third Grade Laboratory under the National State, Administration of Traditional Chinese Medicine, Hangzhou, China
| | - Hongyi Wang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Chenxi Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ao Cheng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Qingwei Deng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Huijuan Zhu
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Disease, Institute of Nephrology, Zhejiang University, Hangzhou, China.,The Third Grade Laboratory under the National State, Administration of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
13
|
Hong C, Jin R, Dai X, Gao X. Functional Contributions of Antigen Presenting Cells in Chronic Graft-Versus-Host Disease. Front Immunol 2021; 12:614183. [PMID: 33717098 PMCID: PMC7943746 DOI: 10.3389/fimmu.2021.614183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/11/2021] [Indexed: 12/27/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is one of the most common reasons of late non-relapse morbidity and mortality of patients with allogeneic hematopoietic stem cell transplantation (allo-HSCT). While acute GVHD is considered driven by a pathogenic T cell dominant mechanism, the pathogenesis of cGVHD is much complicated and involves participation of a variety of immune cells other than pathogenic T cells. Existing studies have revealed that antigen presenting cells (APCs) play crucial roles in the pathophysiology of cGVHD. APCs could not only present auto- and alloantigens to prime and activate pathogenic T cells, but also directly mediate the pathogenesis of cGVHD via multiple mechanisms including infiltration into tissues/organs, production of inflammatory cytokines as well as auto- and alloantibodies. The studies of this field have led to several therapies targeting different APCs with promising results. This review will focus on the important roles of APCs and their contributions in the pathophysiology of cGVHD after allo-HSCT.
Collapse
Affiliation(s)
- Chao Hong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Rong Jin
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiaoqiu Dai
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiaoming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
14
|
Schmid H, Ribeiro EM, Secker KA, Duerr-Stoerzer S, Keppeler H, Dong R, Munz T, Schulze-Osthoff K, Hailfinger S, Schneidawind C, Schneidawind D. Human invariant natural killer T cells promote tolerance by preferential apoptosis induction of conventional dendritic cells. Haematologica 2021; 107:427-436. [PMID: 33440919 PMCID: PMC8804566 DOI: 10.3324/haematol.2020.267583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Indexed: 11/20/2022] Open
Abstract
Graft-versus-host disease (GvHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. We recently showed in murine studies and in vitro human models that adoptively transferred invariant natural killer T (iNKT) cells protect from GvHD and promote graft-versus-leukemia effects. The cellular mechanisms underlying GvHD prevention by iNKT cells in humans, however, remain unknown. In order to study relevant cellular interactions, dendritic cells (DC) were either generated from monocytes or isolated directly from blood of healthy donors or GvHD patients and co-cultured in a mixed lymphocyte reaction (MLR) with T cells obtained from healthy donors or transplantation bags. Addition of culture-expanded iNKT cells to the MLR-induced DC apoptosis in a cell contact-dependent manner, thereby preventing T-cell activation and proliferation. Annexin V/propidium iodide staining and image stream assays showed that CD4+CD8–, CD4–CD8+ and double negative iNKT cells are similarly able to induce DC apoptosis. Further MLR assays revealed that conventional DC (cDC) but not plasmacytoid DC (pDC) could induce alloreactive T-cell activation and proliferation. Interestingly, cDC were also more susceptible to apoptosis induced by iNKT cells, which correlates with their higher CD1d expression, leading to a bias in favor of pDC. Remarkably, these results could also be observed in GvHD patients. We propose a new mechanism how ex vivo expanded human iNKT cells prevent alloreactivity of T cells. iNKT cells modulate T-cell responses by selective apoptosis of DC subsets, resulting in suppression of T-cell activation and proliferation while enabling beneficial immune responses through pDC.
Collapse
Affiliation(s)
- Hannes Schmid
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Emmanuelle M Ribeiro
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Kathy-Ann Secker
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Silke Duerr-Stoerzer
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Hildegard Keppeler
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Ruoyun Dong
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Timo Munz
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | | | - Stephan Hailfinger
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tuebingen
| | - Corina Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Dominik Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen.
| |
Collapse
|
15
|
Molina MS, Stokes J, Hoffman EA, Eremija J, Zeng Y, Simpson RJ, Katsanis E. Bendamustine Conditioning Skews Murine Host DCs Toward Pre-cDC1s and Reduces GvHD Independently of Batf3. Front Immunol 2020; 11:1410. [PMID: 32765499 PMCID: PMC7378358 DOI: 10.3389/fimmu.2020.01410] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/02/2020] [Indexed: 11/21/2022] Open
Abstract
Graft-versus-host disease (GvHD) remains the second leading cause of death in allogeneic hematopoietic stem cell transplantation recipients, highlighting the need for improved preventative strategies. Our laboratory has previously demonstrated in an experimental bone marrow transplantation (BMT) model that bendamustine combined with total body irradiation (BEN+TBI) is a safer alternative to cyclophosphamide with TBI (CY+TBI). The biological mechanisms of action of BEN have not been fully elucidated and likely involve multiple cell populations. Host dendritic cells (DCs) can prime naïve donor T-cells immediately following transplantation, making host DCs critical for the initiation phase of GvHD. We hypothesized that BEN+TBI conditioning favorably alters host DC composition to reduce GvHD. We demonstrate that host DCs treated with BEN+TBI induce less allogeneic T-cell proliferation than those conditioned with CY+TBI. We further show that BEN+TBI conditioning results in greater total numbers of all host DC subsets but with a more favorable composition compared to CY+TBI with significantly larger proportions of type 1 conventional DCs (cDC1), a highly regulatory DC subset capable of suppressing GvHD. Our studies using recipient Batf3 KO mice indicate that CD8α+ cDC1s are largely dispensable for the reduced GvHD following BEN+TBI conditioning. We found a higher frequency of host pre-cDC1s with BEN+TBI conditioning in both wild-type (WT) and Batf3 KO mice, which was inversely associated with GvHD. Additionally, we observed that BEN treatment results in greater expression of Flt3 receptor (CD135) on host DCs compared to CY, potentially contributing to the skewing of host DCs toward cDC1s. Further, BEN+TBI conditioning results in host cDCs with greater expression of PIR-B, an inhibitory receptor capable of preventing lethal GvHD. We conclude that BEN+TBI is a safer alternative to CY+TBI, resulting in a greater frequency of host pre-cDC1s and limiting GvHD.
Collapse
Affiliation(s)
- Megan S. Molina
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Emely A. Hoffman
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Jelena Eremija
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Yi Zeng
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
- Department of Pathology, University of Arizona, Tucson, AZ, United States
| | - Richard J. Simpson
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
- Department of Nutritional Science, University of Arizona, Tucson, AZ, United States
| | - Emmanuel Katsanis
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
- Department of Pathology, University of Arizona, Tucson, AZ, United States
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
16
|
Immunological and Clinical Impact of Manipulated and Unmanipulated DLI after Allogeneic Stem Cell Transplantation of AML Patients. J Clin Med 2019; 9:jcm9010039. [PMID: 31878060 PMCID: PMC7019914 DOI: 10.3390/jcm9010039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is the preferred curative treatment for several hematological malignancies. The efficacy of allo-SCT depends on the graft-versus-leukemia (GvL) effect. However, the prognosis of patients with relapsed acute myeloid leukemia (AML) following allo-SCT is poor. Donor lymphocyte infusion (DLI) is utilized after allo-SCT in this setting to prevent relapse, to prolong progression free survival, to establish full donor chimerism and to restore the GvL effect in patients with hematological malignancies. Thus, there are different options for the administration of DLI in AML patients. DLI is currently used prophylactically and in the setting of an overt relapse. In addition, in the minimal residual disease (MRD) setting, DLI may be a possibility to improve overall survival. However, DLI might increase the risk of severe life-threatening complications such as graft-versus-host disease (GvHD) as well as severe infections. The transfusion of lymphocytes has been tested not only for the treatment of hematological malignancies but also chronic infections. In this context, manipulated DLI in a prophylactic or therapeutic approach are an option, e.g., virus-specific DLI using different selection methods or antigen-specific DLI such as peptide-specific CD8+ cytotoxic T lymphocytes (CTLs). In addition, T cells are also genetically engineered, using both chimeric antigen receptor (CAR) genetically modified T cells and T cell receptor (TCR) genetically modified T cells. T cell therapies in general have the potential to enhance antitumor immunity, augment vaccine efficacy, and limit graft-versus-host disease after allo-SCT. The focus of this review is to discuss the different strategies to use donor lymphocytes after allo-SCT. Our objective is to give an insight into the functional effects of DLI on immunogenic antigen recognition for a better understanding of the mechanisms of DLI. To ultimately increase the GvL potency without raising the risk of GvHD at the same time.
Collapse
|
17
|
Ma YY, Zhao M, Liu Y, Zhao DF, Wang LX, Chen XP, Li L. Use of decitabine for patients with refractory or relapsed acute myeloid leukemia: a systematic review and meta-analysis. Hematology 2019; 24:507-515. [PMID: 31242832 DOI: 10.1080/16078454.2019.1632407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Yuan-yuan Ma
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Min Zhao
- Department of Nutrition, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yi Liu
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - De-feng Zhao
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Li-xin Wang
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xiao-ping Chen
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Li Li
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
18
|
Thomson AW, Metes DM, Ezzelarab MB, Raïch-Regué D. Regulatory dendritic cells for human organ transplantation. Transplant Rev (Orlando) 2019; 33:130-136. [PMID: 31130302 DOI: 10.1016/j.trre.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/09/2019] [Indexed: 12/30/2022]
Abstract
Current immunosuppressive (IS) regimens used to prevent organ allograft rejection have well-recognized side effects, that include enhanced risk of infection and certain types of cancer, metabolic disorders, cardiovascular disease, renal complications and failure to control chronic allograft rejection. The life-long dependency of patients on these IS agents reflects their inability to induce donor-specific tolerance. Extensive studies in rodent and non-human primate models have demonstrated the ability of adoptively-transferred regulatory immune cells (either regulatory myeloid cells or regulatory T cells) to promote transplant tolerance. Consequently, there is considerable interest in the potential of regulatory immune cell therapy to allow safe minimization/complete withdrawal of immunosuppression and the promotion of organ transplant tolerance in the clinic. Here, we review the properties of regulatory dendritic cells (DCreg) with a focus on the approaches being taken to generate human DCreg for clinical testing. We also document the early phase clinical trials that are underway to assess DCreg therapy in clinical organ transplantation as well as in autoimmune disorders.
Collapse
Affiliation(s)
- Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Diana M Metes
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mohamed B Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dalia Raïch-Regué
- Nephropathies Research Group, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
19
|
Wiegering V, Peter L, Frietsch M, Schlegel PG, Eyrich M. Differences of Immune Reconstitution of Dendritic Cells in Pediatric GvHD Patients After Allogenic Stem Cell Transplantation. J Pediatr Hematol Oncol 2019; 41:e101-e107. [PMID: 30557171 DOI: 10.1097/mph.0000000000001342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hematopoietic stem cell transplantation (HSCT) is a life-saving procedure for children with a variety of (non) malignant conditions. GvHD is a severe complication with high morbidity and mortality. The pathogenesis remains unclear. We studied dendritic cell (DC) reconstitution to detect potential differences, which may improve our knowledge in the development of chronic GvHD (cGvHD). PROCEDURE We examined immune reconstitution (T, B, and NK cells and dendritic cells) at defined time points in a pediatric cohort who underwent 61 allogeneic HSCTs. RESULTS Regarding DC reconstitution we found a fast reconstitution of the DC compartment negatively correlated with age. After HSCT, both myeloid DC (mDC) and plasmacytoid DC (pDC) counts recover to pre-HSCT levels within 2 months. Higher CCR7 positive cell counts were found in patients receiving TBI during engraftment and during the whole posttransplant period we found a correlation with an improved outcome. In cGVHD patients decreased total DC counts and increased pDCs were found after day+100. No relevant correlation was achieved regarding to HLA-matching, stem cell manipulation of the graft as well as HSCT-indication compared with different DC counts. DISCUSSION Pathogenesis of cGvHD remains complex. Our data suggest an influence of dendritic cells, which may contribute to the clinical picture and should be further investigated in future studies.
Collapse
Affiliation(s)
- Verena Wiegering
- Department of Pediatric Hematology and Oncology, University Hospital Würzburg, Germany
| | | | | | | | | |
Collapse
|
20
|
Yu H, Tian Y, Wang Y, Mineishi S, Zhang Y. Dendritic Cell Regulation of Graft-Vs.-Host Disease: Immunostimulation and Tolerance. Front Immunol 2019; 10:93. [PMID: 30774630 PMCID: PMC6367268 DOI: 10.3389/fimmu.2019.00093] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Graft-vs.-host disease (GVHD) remains a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Significant progresses have been made in defining the dichotomous role of dendritic cells (DCs) in the development of GVHD. Host-derived DCs are important to elicit allogeneic T cell responses, whereas certain donor-types of DCs derived from newly engrafted hematopoietic stem/progenitor cells (HSPCs) can amply this graft-vs.-host reaction. In contrast, some DCs also play non-redundant roles in mediating immune tolerance. They induce apoptotic deletion of host-reactive donor T cells while promoting expansion and function of regulatory T cells (Treg). Unfortunately, this tolerogenic effect of DCs is impaired during GVHD. Severe GVHD in patients subject to allo-HSCT is associated with significantly decreased number of circulating peripheral blood DCs during engraftment. Existing studies reveal that GVHD causes delayed reconstitution of donor DCs from engrafted HSPCs, impairs the antigen presentation function of newly generated DCs and reduces the capacity of DCs to regulate Treg. The present review will discuss the importance of DCs in alloimmunity and the mechanism underlying DC reconstitution after allo-HSCT.
Collapse
Affiliation(s)
- Hongshuang Yu
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Yuanyuan Tian
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Shin Mineishi
- Department of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States,Department of Microbiology & Immunology, Temple University, Philadelphia, PA, United States,*Correspondence: Yi Zhang
| |
Collapse
|
21
|
Targeting the niche: depleting haemopoietic stem cells with targeted therapy. Bone Marrow Transplant 2019; 54:961-968. [PMID: 30664721 DOI: 10.1038/s41409-019-0445-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/04/2018] [Accepted: 12/30/2018] [Indexed: 12/11/2022]
Abstract
Haemopoietic stem cell transplantation is an expanding procedure worldwide but is associated with significant morbidity and mortality. Depletion of resident haemopoietic stem and progenitor cells (HSPC) is required for both autologous and allogeneic haemopoietic stem cell transplantation. Current conditioning protocols utilise chemotherapy or radiation to effectively reduce HSPC but are toxic in both the short and long term. The initial trials to use monoclonal antibodies to target HSPC were limited with marginal efficacy but platforms including antibody drug conjugates and chimeric antigen receptor T cells have made targeted conditioning strategies achievable. In this review we summarise the work developing targeted conditioning that may replace or reduce alkylating agents and total body irradiation. The prospect of conditioning with significantly reduced toxicity will improve outcomes and open transplantation to patients unable to tolerate current conditioning protocols.
Collapse
|
22
|
Chang YJ, Zhao XY, Huang XJ. Strategies for Enhancing and Preserving Anti-leukemia Effects Without Aggravating Graft-Versus-Host Disease. Front Immunol 2018; 9:3041. [PMID: 30619371 PMCID: PMC6308132 DOI: 10.3389/fimmu.2018.03041] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/10/2018] [Indexed: 12/29/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curable method for the treatment of hematological malignancies. In the past two decades, the establishment of haploidentical transplant modalities make “everyone has a donor” become a reality. However, graft-versus-host disease (GVHD) and relapse remain the major two causes of death either in the human leukocyte antigen (HLA)-matched transplant or haploidentical transplant settings, both of which restrict the improvement of transplant outcomes. Preclinical mice model showed that both donor-derived T cells and natural killer (NK) cells play important role in the pathogenesis of GVHD and the effects of graft-versus-leukemia (GVL). Hence, understanding the immune mechanisms of GVHD and GVL would provide potential strategies for the control of leukemia relapse without aggravating GVHD. The purpose of the current review is to summarize the biology of GVHD and GVL responses in preclinical models and to discuss potential novel therapeutic strategies to reduce the relapse rate after allo-SCT. We will also review the approaches, including optimal donor selection and, conditioning regimens, donor lymphocyte infusion, BCR/ABL-specific CTL, and chimeric antigen receptor-modified T cells, which have been successfully used in the clinic to enhance and preserve anti-leukemia activity, especially GVL effects, without aggravating GVHD or alleviate GVHD.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
23
|
Nguyen H, Kuril S, Bastian D, Kim J, Zhang M, Vaena SG, Dany M, Dai M, Heinrichs JL, Daenthanasanmak A, Iamsawat S, Schutt S, Fu J, Wu Y, Fairlie DP, Atkinson C, Ogretmen B, Tomlinson S, Yu XZ. Complement C3a and C5a receptors promote GVHD by suppressing mitophagy in recipient dendritic cells. JCI Insight 2018; 3:121697. [PMID: 30568037 DOI: 10.1172/jci.insight.121697] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a major complication of allogeneic hematopoietic cell transplantation (HCT). DCs play critical roles in GVHD induction. Modulating autophagy represents a promising therapeutic strategy for the treatment of immunological diseases. Complement receptors C3aR/C5aR expressed on DCs regulate immune responses by translating extracellular signals into intracellular activity. In the current study, we found that C3aR/C5aR deficiency enhanced ceramide-dependent lethal mitophagy (CDLM) in DCs. Cotransfer of host-type C3aR-/-/C5aR-/- DCs in the recipients significantly improved GVHD outcome after allogeneic HCT, primarily through enhancing CDLM in DCs. C3aR/C5aR deficiency in the host hematopoietic compartment significantly reduced GVHD severity via impairing Th1 differentiation and donor T cell glycolytic activity while enhancing Treg generation. Prophylactic treatment with C3aR/C5aR antagonists effectively alleviated GVHD while maintaining the graft-versus-leukemia (GVL) effect. Altogether, we demonstrate that inhibiting C3aR/C5aR induces lethal mitophagy in DCs, which represents a potential therapeutic approach to control GVHD while preserving the GVL effect.
Collapse
Affiliation(s)
- Hung Nguyen
- Department of Microbiology and Immunology and
| | - Sandeepkumar Kuril
- Division of Pediatric Hematology/Oncology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Jisun Kim
- Department of Biochemistry and Molecular Biology and
| | | | | | - Mohammed Dany
- Department of Biochemistry and Molecular Biology and
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jessica Lauren Heinrichs
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | | | - Jianing Fu
- Columbia Center for Translational Immunology, Columbia University, New York City, New York, USA
| | - Yongxia Wu
- Department of Microbiology and Immunology and
| | - David P Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Carl Atkinson
- Department of Microbiology and Immunology and.,Department of Surgery, Transplant Immunobiology Laboratory
| | | | - Stephen Tomlinson
- Department of Microbiology and Immunology and.,Research Service, Ralph H. Johnson Veterans Affairs Medical Center, and
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology and.,Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
24
|
Wang Y, Yang X, Sun X, Rong L, Kang M, Wu P, Ji X, Lin R, Huang J, Xue Y, Fang Y. Bone marrow infiltrated Lnc-INSR induced suppressive immune microenvironment in pediatric acute lymphoblastic leukemia. Cell Death Dis 2018; 9:1043. [PMID: 30310051 PMCID: PMC6181910 DOI: 10.1038/s41419-018-1078-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 08/21/2018] [Accepted: 09/14/2018] [Indexed: 12/27/2022]
Abstract
Immune escape due to immunosuppressive microenvironments, such as those associated with regulatory T (Treg) cells is highly associated with initial occurrence and development of solid tumors or hematologic malignancies. Here, we employed high-throughput transcriptome screening to demonstrate immunosuppression-associated increases in the long noncoding (lnc) RNA lnc-insulin receptor precursor (INSR), which was corrected with INSR expression in CD4+ T cells extracted from the bone marrow of patients with childhood acute T lymphoblastic leukemia. Loss-of-function and gain-of-function assays in vitro and in vivo revealed that membrane-localized and cytoplasm-localized lnc-INSR promoted Treg distribution and decreased the percentage of cytotoxic T lymphocytes, which induced tumor growth. Through direct binding with INSR, lnc-INSR blocked the INSR ubiquitination site, causing abnormal activation of INSR and the phosphatidylinositide 3-kinase/AKT-signaling pathway. These results indicated that lnc-INSR might promote immune suppression by enhancing Treg-cell differentiation and serve as valuable therapeutic targets in the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China.
| | - Xiaoyun Yang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Xiaoyan Sun
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Liucheng Rong
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Meiyun Kang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Peng Wu
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Xiaohui Ji
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Rufeng Lin
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Jie Huang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing Medical University, 72# Guangzhou Road, Nanjing, Jiangsu Province, China.
| |
Collapse
|
25
|
So EY, Sun C, Reginato AM, Dubielecka PM, Ouchi T, Liang OD. Loss of lipid phosphatase SHIP1 promotes macrophage differentiation through suppression of dendritic cell differentiation. Cancer Biol Ther 2018; 20:201-211. [PMID: 30277839 DOI: 10.1080/15384047.2018.1523846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
SH2-containing inositol 5'-phosphatase-1 (SHIP1) deficiency in mice results in abnormal myeloid expansion, and proinflammatory conditions in the lung. However, the mechanisms involved in SHIP1-mediated regulation of myeloid differentiation remain unclear. Here we show that SHIP1 is a key regulator of early differentiation for dendritic cells (DCs). We also provide critical evidence to modify the function of SHIP1 in in vitro development of BMDCs using the recent framework of defining DCs. We found that loss of SHIP1 suppresses GM-CSF-induced formation of bone marrow-derived DC (BMDC) colonies, leading to reduced BMDC number in BM cell culture. The number of maturated BMDCs decreased in SHIP1-KO culture, due to reduction of immature BMDCs, suggesting SHIP1 is critical for lineage commitment rather than for maturation from myeloid precursors to DCs. We further showed that F4/80+/MHCIIlow BM macrophage-like cells (BMMs) were the main population of SHIP1-KO BM culture. Treatment of wild-type BM culture with 3 α-aminocholestane (3AC), a specific inhibitor for functional activity of SHIP1, caused a similar developmental defect in BMDCs as seen in SHIP1-KO cells, resulting in the absence of BMDC colony, and increased number of BMMs in BM culture. In conclusion, our results suggest that differentiation of BMDCs are markedly impaired under SHIP1 deficient condition, which causes skewed development of myeloid lineage cells manifested as pathological conditions associated with an excess of macrophage population.
Collapse
Affiliation(s)
- Eui Young So
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Changqi Sun
- b Division of Rheumatology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Anthony M Reginato
- b Division of Rheumatology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Patrycia M Dubielecka
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| | - Toru Ouchi
- c Department of Cancer Genetics , Roswell Park Cancer Institute , Buffalo , USA
| | - Olin D Liang
- a Division of Hematology and Oncology, Department of Medicine, Rhode Island Hospital , Warren Alpert Medical School of Brown University , Providence , USA
| |
Collapse
|
26
|
Thomson AW, Ezzelarab MB. Regulatory dendritic cells: profiling, targeting, and therapeutic application. Curr Opin Organ Transplant 2018; 23:538-545. [PMID: 30036199 PMCID: PMC6620776 DOI: 10.1097/mot.0000000000000565] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
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.
Collapse
Affiliation(s)
- Angus W Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
27
|
Leyendecker A, Pinheiro CCG, Amano MT, Bueno DF. The Use of Human Mesenchymal Stem Cells as Therapeutic Agents for the in vivo Treatment of Immune-Related Diseases: A Systematic Review. Front Immunol 2018; 9:2056. [PMID: 30254638 PMCID: PMC6141714 DOI: 10.3389/fimmu.2018.02056] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
Background: One of the greatest challenges for medicine is to find a safe and effective treatment for immune-related diseases. However, due to the low efficacy of the treatment available and the occurrence of serious adverse effects, many groups are currently searching for alternatives to the traditional therapy. In this regard, the use of human mesenchymal stem cells (hMSCs) represents a great promise for the treatment of a variety of immune-related diseases due to their potent immunomodulatory properties. The main objective of this study is, therefore, to present and summarize, through a systematic review of the literature, in vivo studies in which the efficacy of the administration of hMSCs for the treatment of immune-related diseases was evaluated. Methods: The article search was conducted in PubMed/MEDLINE, Scopus and Web of Science databases. Original research articles assessing the therapeutic potential of hMSCs administration for the in vivo treatment immune-related diseases, published from 1984 to December 2017, were selected and evaluated. Results: A total of 132 manuscripts formed the basis of this systematic review. Most of the studies analyzed reported positive results after hMSCs administration. Clinical effects commonly observed include an increase in the survival rates and a reduction in the severity and incidence of the immune-related diseases studied. In addition, hMSCs administration resulted in an inhibition in the proliferation and activation of CD19+ B cells, CD4+ Th1 and Th17 cells, CD8+ T cells, NK cells, macrophages, monocytes, and neutrophils. The clonal expansion of both Bregs and Tregs cells, however, was stimulated. Administration of hMSCs also resulted in a reduction in the levels of pro-inflammatory cytokines such as IFN-γ, TNF-α, IL-1, IL-2, IL-12, and IL-17 and in an increase in the levels of immunoregulatory cytokines such as IL-4, IL-10, and IL-13. Conclusions: The results obtained in this study open new avenues for the treatment of immune-related diseases through the administration of hMSCs and emphasize the importance of the conduction of further studies in this area.
Collapse
|
28
|
Teipel R, Oelschlägel U, Wetzko K, Schmiedgen M, Kramer M, Rücker-Braun E, Hölig K, von Bonin M, Heidrich K, Fuchs A, Ordemann R, Kroschinsky F, Bornhäuser M, Hütter G, Schmidt H, Ehninger G, Schetelig J, Heidenreich F. Differences in Cellular Composition of Peripheral Blood Stem Cell Grafts from Healthy Stem Cell Donors Mobilized with Either Granulocyte Colony-Stimulating Factor (G-CSF) Alone or G-CSF and Plerixafor. Biol Blood Marrow Transplant 2018; 24:2171-2177. [PMID: 29935214 DOI: 10.1016/j.bbmt.2018.06.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/13/2018] [Indexed: 11/29/2022]
Abstract
This study was conducted to characterize and compare peripheral blood stem cell grafts from healthy donors who underwent granulocyte colony-stimulating factor (G-CSF) mobilization and subsequently received 1 dose of plerixafor after insufficient stem cell yields were achieved at the first apheresis. Aliquots from 35 donors were collected from the first apheresis after mobilization with G-CSF alone and from the second apheresis after additional plerixafor administration. Samples were freshly analyzed for cellular subsets by 8-color flow cytometry. Leukapheresis samples mobilized with additional plerixafor showed a significant increase of total nucleated cells, including B cells, CD4+ and CD8+ T cells, and CD34+ hematopoietic stem and progenitor cells. Absolute numbers of plasmacytoid dendritic cells were also significantly increased, whereas no changes were detected for myeloid dendritic cells. Furthermore, absolute numbers of regulatory T cells increased, with naive CD45RA+ regulatory T cells showing the highest rise. Finally, strikingly higher numbers of myeloid-derived suppressor cells were detected in the plerixafor and G-CSF-mobilized graft. The mobilization of peripheral stem cells in healthy donors with G-CSF and plerixafor led to a significant difference in cellular graft composition compared with G-CSF alone. The clinical impact of the different cell composition for the graft recipient warrants further clinical investigation.
Collapse
Affiliation(s)
- Raphael Teipel
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Uta Oelschlägel
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Katrin Wetzko
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Maria Schmiedgen
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Michael Kramer
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Elke Rücker-Braun
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Kristina Hölig
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Malte von Bonin
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Katharina Heidrich
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Anke Fuchs
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Rainer Ordemann
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; Cellex Gesellschaft für Zellgewinnung mbH, Dresden, Germany
| | - Frank Kroschinsky
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany
| | - Martin Bornhäuser
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Dresden, Germany
| | - Gero Hütter
- Cellex Gesellschaft für Zellgewinnung mbH, Dresden, Germany
| | | | - Gerhard Ehninger
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Cellex Gesellschaft für Zellgewinnung mbH, Dresden, Germany
| | - Johannes Schetelig
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; DKMS, Clinical Trials Unit, Dresden, Germany
| | - Falk Heidenreich
- Universitätsklinikum Carl Gustav Carus der TU Dresden, Medizinische Klinik und Poliklinik I, Dresden, Germany; DKMS, Clinical Trials Unit, Dresden, Germany.
| |
Collapse
|
29
|
Thomson AW, Humar A, Lakkis FG, Metes DM. Regulatory dendritic cells for promotion of liver transplant operational tolerance: Rationale for a clinical trial and accompanying mechanistic studies. Hum Immunol 2018; 79:314-321. [PMID: 29100944 PMCID: PMC5924712 DOI: 10.1016/j.humimm.2017.10.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022]
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.
Collapse
Affiliation(s)
- Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Abhinav Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fadi G Lakkis
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Diana M Metes
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Ezzelarab MB, Lu L, Shufesky WF, Morelli AE, Thomson AW. Donor-Derived Regulatory Dendritic Cell Infusion Maintains Donor-Reactive CD4 +CTLA4 hi T Cells in Non-Human Primate Renal Allograft Recipients Treated with CD28 Co-Stimulation Blockade. Front Immunol 2018; 9:250. [PMID: 29520267 PMCID: PMC5827543 DOI: 10.3389/fimmu.2018.00250] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/29/2018] [Indexed: 12/14/2022] Open
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.
Collapse
Affiliation(s)
- Mohamed B. Ezzelarab
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Lien Lu
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - William F. Shufesky
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Adrian E. Morelli
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angus W. Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
31
|
Mohammadpour H, O'Neil R, Qiu J, McCarthy PL, Repasky EA, Cao X. Blockade of Host β2-Adrenergic Receptor Enhances Graft-versus-Tumor Effect through Modulating APCs. THE JOURNAL OF IMMUNOLOGY 2018; 200:2479-2488. [PMID: 29445008 DOI: 10.4049/jimmunol.1701752] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/24/2018] [Indexed: 11/19/2022]
Abstract
Allogeneic hematopoietic cell transplantation is a potential curative therapy for hematologic malignancies. Host APCs are pivotal to the desired graft-versus-tumor (GVT) effect. Recent studies have shown that β2-adrenergic receptor (β2AR) signaling can have an important impact on immune cell function, including dendritic cells (DCs). In this article, we demonstrate that pretreatment of host mice with a β2AR blocker significantly increases the GVT effect of donor CD8+ T cells by decreasing tumor burden without increasing graft-versus-host disease. β2AR-deficient host mice have significantly increased effector memory and central memory CD8+ T cells and improved reconstitution of T cells, including CD4+Foxp3+ regulatory T cells. Notably, β2AR deficiency induces increased CD11c+ DC development. Also, β2AR-deficient bone marrow-derived DCs induce higher CD8+ T cell proliferation and improved tumor killing in vitro. Metabolic profiling shows that β2AR deficiency renders DCs more immunogenic through upregulation of mTOR activity and reduction of STAT3 phosphorylation. Altogether, these findings demonstrate an important role for host β2AR signaling in suppressing T cell reconstitution and GVT activity.
Collapse
Affiliation(s)
- Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Rachel O'Neil
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263.,Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201
| | - Jingxin Qiu
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263; and
| | - Philip L McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263
| | - Xuefang Cao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263; .,Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
32
|
Mathew JM, Ansari MJ, Gallon L, Leventhal JR. Cellular and functional biomarkers of clinical transplant tolerance. Hum Immunol 2018; 79:322-333. [PMID: 29374560 DOI: 10.1016/j.humimm.2018.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/16/2022]
Abstract
Development of tolerance protocols requires assays or biomarkers that distinguish tolerant recipients from non-tolerant ones to be established. In addition, a thorough understanding of the plausible mechanisms associated with clinical transplant tolerance is necessary to take the field forward. Unlike the majority of molecular signature analyses utilized by others, the emphasis of this article is on the cellular and functional biomarkers of induced transplant tolerance. Immunity to an organ transplant is very complex, comprised of two broad categories - innate and acquired or adaptive immune responses. Innate immunity can be avoided by eliminating or preventing ischemic injuries to the donor organ and tolerance at the level of adaptive immunity can be induced by infusions of a number of cellular products. Since adaptive immune response consists of inflammatory hypersensitivity, cellular (cytotoxic and helper) and humoral aspects, all these need to be measured, and the recipients who demonstrate donor-specific unresponsiveness in all can be considered tolerant or candidates for immunosuppression minimization and/or withdrawal. The mechanisms by which these agents bring about transplant tolerance include regulation, anergy, exhaustion, senescence and deletion of the recipient immune cells. Another proven mechanism of tolerance is full or mixed donor chimerism. However, it should be cautioned that non-deletional tolerance can be reversed.
Collapse
Affiliation(s)
- James M Mathew
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA; Department of Microbiology-Immunology, Northwestern University, Chicago, IL, USA.
| | - Mohammed Javeed Ansari
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA; Department of Medicine-Nephrology, Northwestern University, Chicago, IL, USA
| | - Lorenzo Gallon
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA; Department of Medicine-Nephrology, Northwestern University, Chicago, IL, USA
| | - Joseph R Leventhal
- Department of Surgery - Comprehensive Transplant Center, Northwestern University, Chicago, IL, USA
| |
Collapse
|
33
|
High PD-L1/CD86 MFI ratio and IL-10 secretion characterize human regulatory dendritic cells generated for clinical testing in organ transplantation. Cell Immunol 2017; 323:9-18. [PMID: 29217299 DOI: 10.1016/j.cellimm.2017.08.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/24/2022]
Abstract
Human regulatory dendritic cells (DCreg) were generated from CD14 immunobead-purified or elutriated monocytes in the presence of vitamin D3 and IL-10. They exhibited similar, low levels of costimulatory CD80 and CD86, but comparatively high levels of co-inhibitory programed death ligand-1 (PD-L1) and IL-10 production compared to control immature DC (iDC). Following Toll-like receptor 4 ligation, unlike control iDC, DCreg resisted phenotypic and functional maturation and further upregulated PD-L1:CD86 expression. Whereas LPS-stimulated control iDC (mature DC; matDC) secreted pro-inflammatory tumor necrosis factor but no IL-10, the converse was observed for LPS-stimulated DCreg. DCreg weakly stimulated naïve and memory allogeneic CD4+ and CD8+ T cell proliferation and IFNγ, IL-17A and perforin/granzyme B production in MLR. Their stimulatory function was enhanced however, by blocking PD-1 ligation. High-throughput T cell receptor (TCR) sequencing revealed that, among circulating T cell subsets, memory CD8+ T cells contained the most alloreactive TCR clonotypes and that, while matDC expanded these alloreactive memory CD8 TCR clonotypes, DCreg induced more attenuated responses. These findings demonstrate the feasibility of generating highly-purified GMP-grade DCreg for systemic infusion, their influence on the alloreactive T cell response, and a key mechanistic role of the PD1 pathway.
Collapse
|
34
|
Zhang C, Liao W, Liu F, Zhu X, He X, Hu A. Immune roles of dendritic cells in stem cell transplantation. Clin Transplant 2017; 31. [PMID: 28833479 DOI: 10.1111/ctr.13090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2017] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells and initial stimulators for immune response. DCs can shape their functions based on their immune states, which are crucial for the balance of immunity and tolerance to preserve homeostasis. In the immune response involved in stem cell transplantation, DCs also play important roles in inducing immune tolerance and antitumor immunity. After the rapid development of stem cell transplantation technology in recent years, the risks of graft rejection, tumor recurrence, and tumorigenicity are still present after stem cell transplantation. It is important to understand the mechanisms of DC-mediated immune tolerance and stimulation during stem cell transplantation. In this review, we will summarize and analyze the regulatory mechanisms of DCs in stem cell transplantation and their application in clinical settings. It may help to promote the innovation in basic theories and therapeutic approaches of stem cell transplantation.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of General Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenwei Liao
- Department of General Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Furong Liu
- Department of General Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaofeng Zhu
- Department of General Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoshun He
- Department of General Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anbin Hu
- Department of General Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
35
|
Radu CA, Fischer S, Diehm Y, Hetzel O, Neubrech F, Dittmar L, Kleist C, Gebhard MM, Terness P, Kneser U, Kiefer J. The combination of mitomycin-induced blood cells with a temporary treatment of ciclosporin A prolongs allograft survival in vascularized composite allotransplantation. Langenbecks Arch Surg 2017; 403:83-92. [PMID: 28823033 DOI: 10.1007/s00423-017-1616-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 08/10/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Vascularized composite allotransplantation (VCA) is a rapidly expanding field of transplantation and provides a potential treatment for complex tissue defects. Peripheral blood mononuclear cells (PBMCs) shortly incubated with the antibiotic and chemotherapeutic agent mitomycin C (MMC) can suppress allogeneic T cell response and control allograft rejection in various organ transplantation models. MMC-incubated PBMCs (MICs) are currently being tested in a phase I clinical trial in kidney transplant patients. Previous studies with MICs in a complex VCA model showed the immunomodulatory potential of these cells. The aim of this study is to optimize and evaluate the use of MICs in combination with a standard immunosuppressive drug in VCA. METHODS Fully mismatched rats were used as hind limb donors [Lewis (RT11)] and recipients [Brown-Norway (RT1n)]. Sixty allogeneic hind limb transplantations were performed in six groups. Group A received donor-derived MICs combined with a temporary ciclosporin A (CsA) treatment. Group B received MICs in combination with a temporarily administered reduced dose of CsA. Group C served as a control and received a standard CsA dose temporarily without an additional administration of MICs, whereas Group D was solely medicated with a reduced CsA dose. Group E received no immunosuppressive therapy, neither CsA nor MICs. Group F was given a continuous standard immunosuppressive regimen consisting of CsA and prednisolone. The endpoint of the study was the onset of allograft rejection which was assessed clinically and histologically. RESULTS In group A and B, the rejection-free interval of the allograft was significantly prolonged to an average of 23.1 ± 1.7 and 24.7 ± 1.8 days compared to the corresponding control groups (p < 0.01). Rejection in groups C, D, and E was noted after 14.3 ± 1.1, 7.8 ± 0.7, and 6.9 ± 0.6 days. No rejection occurred in control group F during the follow-up period of 100 days. No adverse events have been noted. CONCLUSION The findings of this study show that the combination of MICs with a temporary CsA treatment significantly prolongs the rejection-free interval in a complex VCA model. The combination of MICs with CsA showed no adverse events such as graft-versus-host disease. MICs, which are generated by a simple and reliable in vitro technique, represent a potential therapeutic tool for prolonging allograft survival through immunomodulation.
Collapse
Affiliation(s)
- Christian Andreas Radu
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Sebastian Fischer
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Yannick Diehm
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Otto Hetzel
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Florian Neubrech
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Laura Dittmar
- Transplantation Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Christian Kleist
- Transplantation Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany.,Department of Nuclear Medicine, University of Heidelberg, Heidelberg, Germany
| | - Martha Maria Gebhard
- Department of Experimental Surgery, University of Heidelberg, Heidelberg, Germany
| | - Peter Terness
- Transplantation Immunology, Institute for Immunology, University of Heidelberg, Heidelberg, Germany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany
| | - Jurij Kiefer
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, Plastic- and Hand Surgery, University of Heidelberg, Ludwig-Guttmann-Str. 13, D-67071, Ludwigshafen, Germany.
| |
Collapse
|
36
|
Role of the intestinal mucosa in acute gastrointestinal GVHD. Blood 2017; 128:2395-2402. [PMID: 27856471 DOI: 10.1182/blood-2016-06-716738] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022] Open
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
|
37
|
Ezzelarab M, Raich-Regue D, Lu L, Zahorchak A, Perez-Gutierrez A, Humar A, Wijkstrom M, Minervini M, Wiseman R, Cooper D, Morelli A, Thomson A. Renal Allograft Survival in Nonhuman Primates Infused With Donor Antigen-Pulsed Autologous Regulatory Dendritic Cells. Am J Transplant 2017; 17:1476-1489. [PMID: 28009481 PMCID: PMC5444942 DOI: 10.1111/ajt.14182] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/30/2016] [Accepted: 12/15/2016] [Indexed: 01/25/2023]
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.
Collapse
Affiliation(s)
- M.B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - D. Raich-Regue
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. Perez-Gutierrez
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A. Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. Wijkstrom
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M. Minervini
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - R.W. Wiseman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI
| | - D.K.C. Cooper
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.E. Morelli
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - A.W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Angus W. Thomson, PhD, DSc, University of Pittsburgh School of Medicine, 200 Lothrop Street, W1540 BST, Pittsburgh, PA 15261, Phone: (412) 624-6392,
| |
Collapse
|
38
|
Gaignage M, Marillier RG, Uyttenhove C, Dauguet N, Saxena A, Ryffel B, Michiels T, Coutelier JP, Van Snick J. Mouse nidovirus LDV infection alleviates graft versus host disease and induces type I IFN-dependent inhibition of dendritic cells and allo-responsive T cells. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:200-213. [PMID: 28474504 PMCID: PMC5418140 DOI: 10.1002/iid3.157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/20/2017] [Accepted: 02/08/2017] [Indexed: 11/09/2022]
Abstract
Introduction Viruses have developed multiple mechanisms to alter immune reactions. In 1969, it was reported that lactate dehydrogenase‐elevating virus (LDV), a single stranded positive sense mouse nidovirus, delays skin allograft rejection and inhibits spleen alterations in graft versus host disease (GVHD). As the underlying mechanisms have remained unresolved and given the need for new therapies of this disease, we reassessed the effects of the virus on GVHD and tried to uncover its mode of action. Methods GVHD was induced by transfer of parent (B6) spleen cells to non‐infected or LDV‐infected B6D2F1 recipients. In vitro mixed‐lymhocyte culture (MLC) reactions were used to test the effects of the virus on antigen‐presenting cells (APC) and responder T cells. Results LDV infection resulted in a threefold increase in survival rate with reduced weight loss and liver inflammation but with the establishment of permanent chimerism that correlated with decreased interleukine (IL)‐27 and interferon (IFN)γ plasma levels. Infected mice showed a transient elimination of splenic CD11b+ and CD8α+ conventional dendritic cells (cDCs) required for allogeneic CD4 and CD8 T cell responses in vitro. This drop of APC numbers was not observed with APCs derived from toll‐like receptor (TLR)7‐deficient mice. A second effect of the virus was a decreased T cell proliferation and IFNγ production during MLC without detectable changes in Foxp3+ regulatory T cell (Tregs) numbers. Both cDC and responder T cell inhibition were type I IFN dependent. Although the suppressive effects were very transient, the GVHD inhibition was long‐lasting. Conclusion A type I IFN‐dependent suppression of DC and T cells just after donor spleen cell transplantation induces permanent chimerism and donor cell implantation in a parent to F1 spleen cell transplantation model. If this procedure can be extended to full allogeneic bone marrow transplantation, it could open new therapeutic perspectives for hematopoietic stem cell transplantation (HSCT).
Collapse
Affiliation(s)
- Mélanie Gaignage
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Reece G Marillier
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Nicolas Dauguet
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Anubha Saxena
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), University of Orleans, Orleans, France.,Institute of Infectious Disease and Molecular Medicine, RSA, University of Cape Town, Cape Town, South Africa
| | - Thomas Michiels
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Jacques Van Snick
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels Branch, Brussels, Belgium
| |
Collapse
|
39
|
Antithymocyte Globulin Induces a Tolerogenic Phenotype in Human Dendritic Cells. Int J Mol Sci 2016; 17:ijms17122081. [PMID: 27973435 PMCID: PMC5187881 DOI: 10.3390/ijms17122081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/23/2016] [Accepted: 12/05/2016] [Indexed: 11/17/2022] Open
Abstract
Antithymocyte globulin (ATG) is used in the prevention of graft-versus-host disease during allogeneic hematopoietic stem cell transplantation. It is generally accepted that ATG mediates its immunosuppressive effect primarily via depletion of T cells. Here, we analyzed the impact of ATG-Fresenius (now Grafalon®) on human monocyte-derived dendritic cells (DC). ATG induced a semi-mature phenotype in DC with significantly reduced expression of CD14, increased expression of HLA-DR, and intermediate expression of CD54, CD80, CD83, and CD86. ATG-DC showed an increase in IL-10 secretion but no IL-12 production. In line with this tolerogenic phenotype, ATG caused a significant induction of indoleamine 2,3-dioxygenase expression and a concomitant increase in levels of tryptophan metabolites in the supernatants of DC. Further, ATG-DC did not induce the proliferation of allogeneic T cells in a mixed lymphocyte reaction but actively suppressed the T cell proliferation induced by mature DC. These data suggest that besides its well-known effect on T cells, ATG modulates the phenotype of DC in a tolerogenic way, which might constitute an essential part of its immunosuppressive action in vivo.
Collapse
|
40
|
Peled JU, Hanash AM, Jenq RR. Role of the intestinal mucosa in acute gastrointestinal GVHD. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:119-127. [PMID: 27913470 PMCID: PMC5575743 DOI: 10.1182/asheducation-2016.1.119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Intestinal graft-versus-host disease (GVHD) remains a significant obstacle to the success of allogeneic hematopoietic cell transplantation. The intestinal mucosa comprises the inner lining of the intestinal tract and maintains close proximity with commensal microbes that reside within the intestinal lumen. Recent advances have significantly improved our understanding of the interactions between the intestinal mucosa and the enteric microbiota. Changes in host mucosal tissue and commensals posttransplant have been actively investigated, and provocative insights into mucosal immunity and the enteric microbiota are now being translated into clinical trials of novel approaches for preventing and treating acute GVHD. In this review, we summarize recent findings related to aspects of the intestinal mucosa during acute GVHD.
Collapse
Affiliation(s)
- Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Weill Cornell Medical College, New York, NY
| | - Alan M Hanash
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Robert R Jenq
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
41
|
Zhou H, Zheng C, Zhu X, Tang B, Tong J, Zhang X, Zhang L, Liu H, Sun Z. Decitabine prior to salvaged unrelated cord blood transplantation for refractory or relapsed childhood acute leukemia. Pediatr Transplant 2016; 20:1117-1124. [PMID: 27620713 DOI: 10.1111/petr.12805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/17/2016] [Indexed: 01/24/2023]
Abstract
No clinical studies have investigated the role of decitabine as a part of the myeloablative conditioning regimen prior to UCBT for refractory or relapsed childhood AL in patients in NR status. The aim of this study was to identify the potential benefits of decitabine as a prior therapy before salvaged unrelated UCBT for refractory or relapsed childhood AL. Eight consecutive patients with childhood refractory/relapsed AL were enrolled in our study between 2013 and 2014. All patients were in NR status before the time of transplant and had features associated with poor outcomes, such as CNSL, MDS-AML, high WBC count at diagnosis, and hypodiploid status (FLT3+/ITD+). Additionally, all patients had one of the following disease statuses: PIF, multiple relapse, or early relapse. All transplants were performed with decitabine as part of the myeloablative conditioning regimen, which was decitabine+Flu/Bu/CY±BCNU or decitabine+Ara-c/BU/CY2±BCNU. A total of seven patients (7 of 8) achieved neutrophil engraftment and platelet engraftment, and one patient experienced primary graft failure. All eight patients (100%) developed PES at a median of 7 days. Three patients developed stage II-IV acute GVHD at a median of 18 days. Additionally, three patients developed chronic GVHD, but it was not extensive in any of those three patients. The median follow-up time after CBT was 19.9 months (range, 9.2-30.7 months). The estimated probability of OS was 75%. Two patients (2 of 8) experienced a testis relapse, and two patients (2 of 8) died. Our experience suggests that the additional application of decitabine as part of the myeloablative conditioning regimen prior to UCBT for refractory or relapsed childhood AL among patients who are not in remission is safe and might be an effective treatment option.
Collapse
Affiliation(s)
- Haixia Zhou
- Shandong University School of Medicine, Jinan, China
| | - Changcheng Zheng
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Xiaoyu Zhu
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Baolin Tang
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Juan Tong
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Xuhan Zhang
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Lei Zhang
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Huilan Liu
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Zimin Sun
- Shandong University School of Medicine, Jinan, China.,Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| |
Collapse
|
42
|
DLL4 + dendritic cells: Key regulators of Notch Signaling in effector T cell responses. Pharmacol Res 2016; 113:449-457. [PMID: 27639599 DOI: 10.1016/j.phrs.2016.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 01/07/2023]
Abstract
Dendritic cells (DCs) are critical regulators of adaptive immune responses. DCs can elicit primary T cell responses at low DC:T cell ratios through their expression of high levels of antigen-presenting molecules and costimulatory molecules. DCs are important for induction of functionally diverse T cell subsets such as CD4+ T helper (Th)1 and Th17 cells and effector CD8+ T cells able to reside in epithelial tissues. Recent studies begin illuminating the underlying mechanism by which DCs regulate specialized T cell subsets. DCs are composed of subsets that differ in their phenotype, localization and function. DCs expressing high levels of DLL4 (DLL4+ DCs), which is a member of Notch ligand family, are newly discovered cells that have greater ability than DLL4- DCs to promote the generation of Th1 and Th17 CD4+ T cells. DLL4 derived from DLL4+ DCs is also important for promoting the differentiation and expansion of effector CD8+ T cells. Experimental studies have demonstrated that selective deletion of DLL4 in DCs causes impaired antitumor immunity. In contrast, blocking DLL4 leads to dramatic reduction of inflammatory T cell responses and their-mediated tissue damage. We will discuss emerging functional specialization within the DLL4+ DC compartment, DLL4+ DC biology and the impact of pharmacological modulation of DLL4 to control inflammatory disorders.
Collapse
|
43
|
Urso K, Alvarez D, Cremasco V, Tsang K, Grauel A, Lafyatis R, von Andrian UH, Ermann J, Aliprantis AO. IL4RA on lymphatic endothelial cells promotes T cell egress during sclerodermatous graft versus host disease. JCI Insight 2016; 1:e88057. [PMID: 27547823 PMCID: PMC4988402 DOI: 10.1172/jci.insight.88057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/07/2016] [Indexed: 01/06/2023] Open
Abstract
Systemic sclerosis (SSc) is a potentially fatal autoimmune disorder with limited therapeutic options. Sclerodermatous graft versus host disease (sclGvHD), induced by transfer of B10.D2 splenocytes into BALB/c Rag2-/- mice, models an inflammatory subset of SSc characterized by a prominent IL13-induced gene expression signature in the skin. Host mice deficient in IL4RA, a subunit of the type II IL4/IL13 receptor, are protected from sclGvHD. While IL4RA has a well-established role in Th2 differentiation and alternative macrophage activation, we report here a previously unappreciated function for IL4RA in lymphatic endothelial cells (LECs): regulation of activated T cell egress. Seven days after splenocyte transfer, Il4ra-/- hosts had increased numbers of activated graft CD4+ T cells in skin draining lymph nodes (dLNs) but fewer T cells in efferent lymph, blood, and skin. Sphingosine-1 phosphate (S1P), master regulator of lymphocyte egress from LNs, was lower in dLNs of Il4ra-/- hosts with a corresponding decrease of S1P kinase 1 (Sphk1) expression in LECs. Bypassing the efferent lymphatics via i.v. injection of CD4+ T cells from dLNs of Il4ra-/- sclGvHD mice restored clinical GvHD in secondary Il4ra-/- recipients. These results identify a role for IL4RA and suggest that modulation of lymphocyte egress from LNs may be effective in SSc and GvHD.
Collapse
Affiliation(s)
- Katia Urso
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School
| | - David Alvarez
- Department of Microbiology and Immunobiology, Harvard Medical School
| | - Viviana Cremasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute
| | - Kelly Tsang
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School
| | - Angelo Grauel
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute
| | - Robert Lafyatis
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ulrich H. von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School
- Ragon Institute of Massachusetts General Hospital (MGH), MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Joerg Ermann
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School
| | - Antonios O. Aliprantis
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital and Harvard Medical School
| |
Collapse
|
44
|
Li Y, Chen D, Wang X, Tong J, Li K, Jing Y, Li G. The Effect of Traditional Chinese Formula Danchaiheji on the Differentiation of Regulatory Dendritic Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:9179470. [PMID: 27525028 PMCID: PMC4976157 DOI: 10.1155/2016/9179470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 01/03/2023]
Abstract
Recently, regulatory dendritic cells (DCregs), a newly described dendritic cell subset with potent immunomodulatory function, have attracted increased attention for their utility in treating immune response-related diseases, such as graft-versus-host disease, hypersensitivity, and autoimmune diseases. Danchaiheji (DCHJ) is a traditional Chinese formula that has been used for many years in the clinic. However, whether DCHJ can program dendritic cells towards a regulatory phenotype and the underlying mechanism behind this process remain unknown. Herein, we investigate the effects of traditional Chinese DCHJ on DCregs differentiation and a mouse model of skin transplantation. The current study demonstrates that DCHJ can induce dendritic cells to differentiate into DCregs, which are represented by high CD11b and low CD86 and HLA-DR expression as well as the secretion of IL-10 and TGF-β. In addition, DCHJ inhibited DC migration and T cell proliferation, which correlated with increased IDO expression. Furthermore, DCHJ significantly prolonged skin graft survival time in a mouse model of skin transplantation without any liver or kidney toxicity. The traditional Chinese formula DCHJ has the potential to be a potent immunosuppressive agent with high efficiency and nontoxicity.
Collapse
Affiliation(s)
- Yingxi Li
- Department of Genetics, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| | - Dan Chen
- Department of Pharmacology, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| | - Xiaodong Wang
- Department of Pharmacology, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| | - Jingzhi Tong
- Department of Pharmacology, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| | - Keqiu Li
- Department of Genetics, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| | - Yaqing Jing
- Department of Genetics, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| | - Guang Li
- Department of Genetics, Basic Medical College, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
45
|
Alloantigen presentation and graft-versus-host disease: fuel for the fire. Blood 2016; 127:2963-70. [PMID: 27030390 DOI: 10.1182/blood-2016-02-697250] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/05/2016] [Indexed: 12/16/2022] Open
Abstract
Allogeneic stem cell transplantation (SCT) is a unique procedure, primarily in patients with hematopoietic malignancies, involving chemoradiotherapy followed by the introduction of donor hematopoietic and immune cells into an inflamed and lymphopenic environment. Interruption of the process by which recipient alloantigen is presented to donor T cells to generate graft-versus-host disease (GVHD) represents an attractive therapeutic strategy to prevent morbidity and mortality after SCT and has been increasingly studied in the last 15 years. However, the immune activation resulting in GVHD has no physiological equivalent in nature; alloantigen is ubiquitous, persists indefinitely, and can be presented by multiple cell types at numerous sites, often on incompatible major histocompatibility complex, and occurs in the context of intense inflammation early after SCT. The recognition that alloantigen presentation is also critical to the development of immunological tolerance via both deletional and regulatory mechanisms further adds to this complexity. Finally, GVHD itself appears capable of inhibiting the presentation of microbiological antigens by donor dendritic cells late after SCT that is mandatory for the establishment of effective pathogen-specific immunity. Here, we review our current understanding of alloantigen, its presentation by various antigen-presenting cells, subsequent recognition by donor T cells, and the potential of therapeutic strategies interrupting this disease-initiating process to modify transplant outcome.
Collapse
|
46
|
Seldon TA, Pryor R, Palkova A, Jones ML, Verma ND, Findova M, Braet K, Sheng Y, Fan Y, Zhou EY, Marks JD, Munro T, Mahler SM, Barnard RT, Fromm PD, Silveira PA, Elgundi Z, Ju X, Clark GJ, Bradstock KF, Munster DJ, Hart DNJ. Immunosuppressive human anti-CD83 monoclonal antibody depletion of activated dendritic cells in transplantation. Leukemia 2016; 30:692-700. [PMID: 26286117 DOI: 10.1038/leu.2015.231] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 07/27/2015] [Indexed: 02/05/2023]
Abstract
Current immunosuppressive/anti-inflammatory agents target the responding effector arm of the immune response and their nonspecific action increases the risk of infection and malignancy. These effects impact on their use in allogeneic haematopoietic cell transplantation and other forms of transplantation. Interventions that target activated dendritic cells (DCs) have the potential to suppress the induction of undesired immune responses (for example, graft versus host disease (GVHD) or transplant rejection) and to leave protective T-cell immune responses intact (for example, cytomegalovirus (CMV) immunity). We developed a human IgG1 monoclonal antibody (mAb), 3C12, specific for CD83, which is expressed on activated but not resting DC. The 3C12 mAb and an affinity improved version, 3C12C, depleted CD83(+) cells by CD16(+) NK cell-mediated antibody-dependent cellular cytotoxicity, and inhibited allogeneic T-cell proliferation in vitro. A single dose of 3C12C prevented human peripheral blood mononuclear cell-induced acute GVHD in SCID mouse recipients. The mAb 3C12C depleted CMRF-44(+)CD83(bright) activated DC but spared CD83(dim/-) DC in vivo. It reduced human T-cell activation in vivo and maintained the proportion of CD4(+) FoxP3(+) CD25(+) Treg cells and also viral-specific CD8(+) T cells. The anti-CD83 mAb, 3C12C, merits further evaluation as a new immunosuppressive agent in transplantation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/genetics
- Antigens, CD/immunology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Proliferation/drug effects
- Cytotoxicity, Immunologic/drug effects
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Female
- Gene Expression
- Graft Rejection/immunology
- Graft Rejection/mortality
- Graft Rejection/pathology
- Graft Rejection/prevention & control
- Graft vs Host Disease/immunology
- Graft vs Host Disease/mortality
- Graft vs Host Disease/pathology
- Graft vs Host Disease/prevention & control
- Humans
- Immunoglobulins/genetics
- Immunoglobulins/immunology
- Immunosuppressive Agents/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/transplantation
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, SCID
- Survival Analysis
- Transplantation, Heterologous
- CD83 Antigen
Collapse
Affiliation(s)
- T A Seldon
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
| | - R Pryor
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - A Palkova
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - M L Jones
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - N D Verma
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - M Findova
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - K Braet
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
| | - Y Sheng
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
| | - Y Fan
- Anesthesia, Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, CA, USA
| | - E Y Zhou
- Anesthesia, Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, CA, USA
| | - J D Marks
- Anesthesia, Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, CA, USA
| | - T Munro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - S M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - R T Barnard
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - P D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - P A Silveira
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Z Elgundi
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - X Ju
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - G J Clark
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - K F Bradstock
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - D J Munster
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
| | - D N J Hart
- DC Program, Mater Medical Research Institute, Brisbane, Queensland, Australia
- Co-operative Research Centre for Biomarker Translation, Melbourne, Victoria, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
- Dendritic Cell Research, ANZAC Research Institute, Concord, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Fu J, Wu Y, Nguyen H, Heinrichs J, Schutt S, Liu Y, Liu C, Jin J, Anasetti C, Yu XZ. T-bet Promotes Acute Graft-versus-Host Disease by Regulating Recipient Hematopoietic Cells in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:3168-79. [PMID: 26903480 DOI: 10.4049/jimmunol.1501020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/21/2016] [Indexed: 01/16/2023]
Abstract
Beyond its critical role in T cells, T-bet regulates the functions of APCs including dendritic cells and B cells, as well as NK cells. Given that recipient APCs are essential for priming allogeneic T cells and recipient NK or T cells are able to reject allogeneic donor cells, we evaluated the role of T-bet on the host in acute graft-versus-host disease (GVHD) using murine models of allogeneic bone marrow transplantation. T-bet(-/-) recipients developed significantly milder GVHD than their wild type counterparts in MHC-mismatched or CD4-dependent minor histocompatibility Ag-mismatched models. Allogeneic donor T cells, in particular, CD4 subset, significantly reduced IFN-γ production, proliferation and migration, and caused less injury in liver and gut of T-bet(-/-) recipients. We further observed that T-bet on recipient hematopoietic cells was primarily responsible for the donor T cell response and pathogenicity in GVHD. T-bet(-/-) dendritic cells expressed higher levels of Trail, whereas they produced lower levels of IFN-γ and IL-12/23 p40, as well as chemokine CXCL9, resulting in significantly higher levels of apoptosis, less priming, and infiltration of donor T cells. Meanwhile, NK cells in T-bet(-/-) hosts partially contribute to the decreased donor T cell proliferation. Furthermore, although T-bet on hematopoietic cells was required for GVHD development, it was largely dispensable for the graft-versus-leukemia effect. Taken together with our previous findings, we propose that T-bet is a potential therapeutic target for the control of GVHD through regulating donor T cells and recipient hematopoietic cells.
Collapse
Affiliation(s)
- Jianing Fu
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33612; Department of Immunology, Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Hung Nguyen
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Jessica Heinrichs
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Steven Schutt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Yuejun Liu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Chen Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32611
| | - Junfei Jin
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425; Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541001 Guangxi, China; and
| | - Claudio Anasetti
- Department of Immunology, Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425; Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
48
|
Norelli M, Camisa B, Bondanza A. Modeling Human Graft-Versus-Host Disease in Immunocompromised Mice. Methods Mol Biol 2016; 1393:127-32. [PMID: 27033222 DOI: 10.1007/978-1-4939-3338-9_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) from an allogeneic donor is an effective form of cancer immunotherapy, especially for acute leukemias. HSCT is however frequently complicated by the occurrence of graft-versus-host disease (GVHD). Immunocompromised mice infused with human T cells often develop a clinical syndrome resembling human GVHD (xenogeneic or X-GVHD). Herein, we describe a method for inducing X-GVHD in a highly reproducible manner. Given the human nature of immune effectors, this xenogeneic model can be routinely adopted for screening the efficacy of new treatments for GVHD.
Collapse
Affiliation(s)
- Margherita Norelli
- Innovative Immunotherapies Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, Milano, 21032, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Barbara Camisa
- Innovative Immunotherapies Unit, IRCCS San Raffaele Hospital, Via Olgettina 60, Milano, 21032, Italy
| | - Attilio Bondanza
- Vita-Salute San Raffaele University, Milano, Italy.
- Innovative Immunotherapies Unit, IRCCS San Raffaele Hospital Scientific Institute, Via Olgettina 60, Milano, 21032, Italy.
| |
Collapse
|
49
|
Meng L, Bai Z, He S, Mochizuki K, Liu Y, Purushe J, Sun H, Wang J, Yagita H, Mineishi S, Fung H, Yanik GA, Caricchio R, Fan X, Crisalli LM, Hexner EO, Reshef R, Zhang Y, Zhang Y. The Notch Ligand DLL4 Defines a Capability of Human Dendritic Cells in Regulating Th1 and Th17 Differentiation. THE JOURNAL OF IMMUNOLOGY 2015; 196:1070-80. [PMID: 26712946 DOI: 10.4049/jimmunol.1501310] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 11/19/2015] [Indexed: 01/07/2023]
Abstract
Notch signaling regulates multiple helper CD4(+) T cell programs. We have recently demonstrated that dendritic cells (DCs) expressing the Notch ligand DLL4 are critical for eliciting alloreactive T cell responses and induction of graft-versus-host disease in mice. However, the human counterpart of murine DLL4(+) DCs has yet to be examined. We report the identification of human DLL4(+) DCs and their critical role in regulating Th1 and Th17 differentiation. CD1c(+) DCs and plasmacytoid DCs (pDCs) from the peripheral blood (PB) of healthy donors did not express DLL4. In contrast, patients undergoing allogeneic hematopoietic stem cell transplantation had a 16-fold more DLL4(+)CD1c(+) DCs than healthy donors. Upon activation of TLR signaling, healthy donor-derived CD1c(+) DCs dramatically upregulated DLL4, as did pDCs to a lesser extent. Activated DLL4(+) DCs were better able to promote Th1 and Th17 differentiation than unstimulated PB DCs. Blocking DLL4 using a neutralizing Ab decreased Notch signaling in T cells stimulated with DLL4(+) DCs, and it reduced the generation of Th1 and Th17 cells. Both NF-κB and STAT3 were crucial for inducing DLL4 in human DCs. Interestingly, STAT3 directly activated DLL4 transcription and inhibiting STAT3 alone was sufficient to reduce DLL4 in activated PB DCs. Thus, DLL4 is a unique functional molecule of human circulating DCs critical for directing Th1 and Th17 differentiation. These findings identify a pathway for therapeutic intervention for inflammatory disorders in humans, such as graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, autoimmunity, and tumor immunity.
Collapse
Affiliation(s)
- Lijun Meng
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200231, China; Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140; Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140
| | - Zhenjiang Bai
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140; Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Soochow University, Suzhou 215003, China
| | - Shan He
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140; Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140
| | - Kazuhiro Mochizuki
- Department of Pediatric Oncology, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yongnian Liu
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140
| | - Janaki Purushe
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140; Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140
| | - Hongxing Sun
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140; Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140
| | - Jian Wang
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Soochow University, Suzhou 215003, China
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Shin Mineishi
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294
| | - Henry Fung
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111
| | - Gregory A Yanik
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Roberto Caricchio
- Department of Medicine, Temple University School of Medicine, Philadelphia, PA 19148
| | - Xiaoxuan Fan
- Flow Cytometry Core Facility, Temple University School of Medicine, Temple University, Philadelphia, PA 19148; and
| | - Lisa M Crisalli
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Elizabeth O Hexner
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Ran Reshef
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Yanyun Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai 200231, China;
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140; Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140;
| |
Collapse
|
50
|
Plasmacytoid dendritic cells in allogeneic hematopoietic cell transplantation: benefit or burden? Bone Marrow Transplant 2015; 51:333-43. [PMID: 26642333 DOI: 10.1038/bmt.2015.301] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/25/2015] [Accepted: 10/27/2015] [Indexed: 11/09/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) bridge innate and adaptive immune responses and have important roles in hematopoietic engraftment, GvHD and graft-versus-leukemia responses following allogeneic hematopoietic cell transplantation (HCT). In addition, pDCs mediate antiviral immunity, particularly as they are the body's primary cellular source of type I interferon. Given their pleiotropic roles, pDCs have emerged as cells that critically impact transplant outcomes, including overall survival. In this article, we will review the pre-clinical and clinical literature, supporting the crucial roles that pDCs assume as key immune effector cells during HCT.
Collapse
|