1
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
2
|
Imam S, Dar P, Aziz SW, Zahid ZA, Sarwar H, Karim T, Faisal S, Haseeb I, Naqvi AS, Shah R, Haque A, Salim N, Jaume JC. Immune Cell Plasticity Allows for Resetting of Phenotype From Effector to Regulator With Combined Inhibition of Notch/eIF5A Pathways. Front Cell Dev Biol 2021; 9:777805. [PMID: 34881246 PMCID: PMC8645838 DOI: 10.3389/fcell.2021.777805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/04/2021] [Indexed: 01/23/2023] Open
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic β-cells caused by an altered immune balance in the pancreatic microenvironment. In humans as well as in mouse models, T cells are well recognized as key orchestrators of T1D, which is characterized by T helper (Th) 1 and Th17 cell bias and/or low/defective T-regulatory cells (Treg), and culminates in cytotoxic T-cell (CTL)-mediated destruction of β-cells. Refitting of immune cells toward the non-inflammatory phenotype in the pancreas may represent a way to prevent/treat T1D. Recently we developed a unique spontaneous humanized mouse model of type 1 diabetes, wherein mouse MHC-II molecules were replaced by human DQ8, and β-cells were made to express human glutamic acid decarboxylase (GAD) 65 auto-antigen. The mice spontaneously developed T1D resembling the human disease. Humanized T1D mice showed hyperglycemic (250-300 mg/dl) symptoms by the 4th week of life. The diabetogenic T cells (CD4, CD8) present in our model are GAD65 antigen-specific in nature. Intermolecular antigen spreading recorded during 3rd-6th week of age is like that observed in the human preclinical period of T1D. In this paper, we tested our hypothesis in our spontaneous humanized T1D mouse model. We targeted two cell-signaling pathways and their inhibitions: eIF5A pathway inhibition influences T helper cell dynamics toward the non-inflammatory phenotype and Notch signaling inhibition enrich Tregs and targets auto-reactive CTLs, rescues the pancreatic islet structure, and increases the functionality of β-cells in terms of insulin production. We report that inhibition of (eIF5A + Notch) signaling mediates suppression of diabetogenic T cells by inducing plasticity in CD4 + T cells co-expressing IL-17 and IFNγ (IL-17 + IFNγ +) toward the Treg cells phenotype.
Collapse
Affiliation(s)
- Shahnawaz Imam
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Pervaiz Dar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States.,Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, India
| | - Saba Wasim Aziz
- Department of Internal Medicine, Division of Endocrinology, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Zeeshan A Zahid
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Haider Sarwar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States.,Windsor University School of Medicine, Cayon, West Indies
| | - Tamanna Karim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Sarah Faisal
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,College of Art and Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Ibrahim Haseeb
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Ahmed S Naqvi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Ottawa Hills High School, Ottawa, OH, United States
| | - Rayyan Shah
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Sylvania Northview High School, Toledo, OH, United States
| | - Amna Haque
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Austin College, Sherman, TX, United States
| | - Nancy Salim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| | - Juan C Jaume
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo, Toledo, OH, United States
| |
Collapse
|
3
|
Abstract
Animal models provide the link between in vitro research and the first in-man application during clinical trials. They provide substantial information in preclinical studies for the assessment of new therapeutic interventions in advance of human clinical trials. However, each model has its advantages and limitations in the ability to imitate specific pathomechanisms. Therefore, the selection of an animal model for the evaluation of a specific research question or evaluation of a novel therapeutic strategy requires a precise analysis. Transplantation research is a discipline that largely benefits from the use of animal models with mouse and pig models being the most frequently used models in organ transplantation research. A suitable animal model should reflect best the situation in humans, and the researcher should be aware of the similarities as well as the limitations of the chosen model. Small animal models with rats and mice are contributing to the majority of animal experiments with the obvious advantages of these models being easy handling, low costs, and high reproductive rates. However, unfortunately, they often do not translate to clinical use. Large animal models, especially in transplantation medicine, are an important element for establishing preclinical models that do often translate to the clinic. Nevertheless, they can be costly, present increased regulatory requirements, and often are of high ethical concern. Therefore, it is crucial to select the right animal model from which extrapolations and valid conclusions can be obtained and translated into the human situation. This review provides an overview in the models frequently used in organ transplantation research.
Collapse
|
4
|
Allen F, Maillard I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front Cell Dev Biol 2021; 9:649205. [PMID: 34124039 PMCID: PMC8194077 DOI: 10.3389/fcell.2021.649205] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the Notch signaling pathway has been investigated as a therapeutic target for the treatment of cancers, and more recently in the context of immune and inflammatory disorders. Notch is an evolutionary conserved pathway found in all metazoans that is critical for proper embryonic development and for the postnatal maintenance of selected tissues. Through cell-to-cell contacts, Notch orchestrates cell fate decisions and differentiation in non-hematopoietic and hematopoietic cell types, regulates immune cell development, and is integral to shaping the amplitude as well as the quality of different types of immune responses. Depriving some cancer types of Notch signals has been shown in preclinical studies to stunt tumor growth, consistent with an oncogenic function of Notch signaling. In addition, therapeutically antagonizing Notch signals showed preclinical potential to prevent or reverse inflammatory disorders, including autoimmune diseases, allergic inflammation and immune complications of life-saving procedures such allogeneic bone marrow and solid organ transplantation (graft-versus-host disease and graft rejection). In this review, we discuss some of these unique approaches, along with the successes and challenges encountered so far to target Notch signaling in preclinical and early clinical studies. Our goal is to emphasize lessons learned to provide guidance about emerging strategies of Notch-based therapeutics that could be deployed safely and efficiently in patients with immune and inflammatory disorders.
Collapse
Affiliation(s)
- Frederick Allen
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Maillard
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
5
|
Rosik J, Szostak B, Machaj F, Pawlik A. The Role of CTLA4 and Its Polymorphisms in Solid Organ and Haematopoietic Stem Cell Transplantation. Int J Mol Sci 2021; 22:ijms22063081. [PMID: 33802937 PMCID: PMC8002677 DOI: 10.3390/ijms22063081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
HLA matching, transplantation technique, or underlying disease greatly influences the probability of long-term transplantation success. It has been hypothesised that genetic variation affecting antigen presentation also contributes to the outcomes of both solid organ transplantation and allogeneic haematopoietic stem cell transplantation (AHSCT). Those genes, along with those responsible for innate and adaptive immunity, have become targets of investigation. In this review, we focus on the role of CTLA4 in the process of acute graft rejection and summarise the progress in our understanding of its role in predicting the outcome. We present the results of the latest studies investigating the link between CTLA4 gene variability and AHSCT, as well as organ transplantation outcomes. While some studies found a link between +49 A/G and −318 C/T and transplantation outcomes, comprehensive meta-analyses have failed to present any association. The most recent field reviews suggest that the −1772 T/C (rs733618) CC genotype is weakly associated with a lower risk of acute graft rejection, while +49 A/G might be clinically meaningful when investigated in the context of combinations with other polymorphisms. Studies verifying associations between 12 CTLA4 gene SNPs and AHSCT outcomes present inexplicit results. Some of the most commonly studied polymorphisms in this context include +49 A/G (rs231775) and CT60 A/G (rs3087243). The results signify that, in order to understand the role of CTLA4 and its gene polymorphisms in transplantology, further studies must be conducted.
Collapse
|
6
|
Maryamchik E, Gallagher KME, Preffer FI, Kadauke S, Maus MV. New directions in chimeric antigen receptor T cell [CAR-T] therapy and related flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2020; 98:299-327. [PMID: 32352629 DOI: 10.1002/cyto.b.21880] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor (CAR) T cells provide a promising approach to the treatment of hematologic malignancies and solid tumors. Flow cytometry is a powerful analytical modality, which plays an expanding role in all stages of CAR T therapy, from lymphocyte collection, to CAR T cell manufacturing, to in vivo monitoring of the infused cells and evaluation of their function in the tumor environment. Therefore, a thorough understanding of the new directions is important for designing and implementing CAR T-related flow cytometry assays in the clinical and investigational settings. However, the speed of new discoveries and the multitude of clinical and preclinical trials make it challenging to keep up to date in this complex field. In this review, we summarize the current state of CAR T therapy, highlight the areas of emergent research, discuss applications of flow cytometry in modern cell therapy, and touch upon several considerations particular to CAR detection and assessing the effectiveness of CAR T therapy.
Collapse
Affiliation(s)
- Elena Maryamchik
- Department of Pathology and Laboratory Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Frederic I Preffer
- Clinical Cytometry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stephan Kadauke
- Department of Pathology and Laboratory Medicine, Cell and Gene Therapy Laboratory, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Cellular Immunotherapy Program, Department of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Perkey E, Maurice De Sousa D, Carrington L, Chung J, Dils A, Granadier D, Koch U, Radtke F, Ludewig B, Blazar BR, Siebel CW, Brennan TV, Nolz J, Labrecque N, Maillard I. GCNT1-Mediated O-Glycosylation of the Sialomucin CD43 Is a Sensitive Indicator of Notch Signaling in Activated T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:1674-1688. [PMID: 32060138 DOI: 10.4049/jimmunol.1901194] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/10/2020] [Indexed: 01/05/2023]
Abstract
Notch signaling is emerging as a critical regulator of T cell activation and function. However, there is no reliable cell surface indicator of Notch signaling across activated T cell subsets. In this study, we show that Notch signals induce upregulated expression of the Gcnt1 glycosyltransferase gene in T cells mediating graft-versus-host disease after allogeneic bone marrow transplantation in mice. To determine if Gcnt1-mediated O-glycosylation could be used as a Notch signaling reporter, we quantified the core-2 O-glycoform of CD43 in multiple T cell subsets during graft-versus-host disease. Pharmacological blockade of Delta-like Notch ligands abrogated core-2 O-glycosylation in a dose-dependent manner after allogeneic bone marrow transplantation, both in donor-derived CD4+ and CD8+ effector T cells and in Foxp3+ regulatory T cells. CD43 core-2 O-glycosylation depended on cell-intrinsic canonical Notch signals and identified CD4+ and CD8+ T cells with high cytokine-producing ability. Gcnt1-deficient T cells still drove lethal alloreactivity, showing that core-2 O-glycosylation predicted, but did not cause, Notch-dependent T cell pathogenicity. Using core-2 O-glycosylation as a marker of Notch signaling, we identified Ccl19-Cre+ fibroblastic stromal cells as critical sources of Delta-like ligands in graft-versus-host responses irrespective of conditioning intensity. Core-2 O-glycosylation also reported Notch signaling in CD8+ T cell responses to dendritic cell immunization, Listeria infection, and viral infection. Thus, we uncovered a role for Notch in controlling core-2 O-glycosylation and identified a cell surface marker to quantify Notch signals in multiple immunological contexts. Our findings will help refine our understanding of the regulation, cellular source, and timing of Notch signals in T cell immunity.
Collapse
Affiliation(s)
- Eric Perkey
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Dave Maurice De Sousa
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec H1T 2M4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Léolène Carrington
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Jooho Chung
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Alexander Dils
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - David Granadier
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Ute Koch
- École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Freddy Radtke
- École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | | | | | - Jeffrey Nolz
- Oregon Health and Sciences University, Portland, OR 97239; and
| | - Nathalie Labrecque
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montreal, Quebec H1T 2M4, Canada; .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3T 1J4, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104;
| |
Collapse
|
8
|
Abstract
The evolutionarily conserved Notch signalling pathway regulates the differentiation and function of mature T lymphocytes with major context-dependent consequences in host defence, autoimmunity and alloimmunity. The emerging effects of Notch signalling in T cell responses build upon a more established role for Notch in T cell development. Here, we provide a critical review of this burgeoning literature to make sense of what has been learned so far and highlight the experimental strategies that have been most useful in gleaning physiologically relevant information. We outline the functional consequences of Notch signalling in mature T cells in addition to key specific Notch ligand–receptor interactions and downstream molecular signalling pathways. Our goal is to help clarify future directions for this expanding body of work and the best approaches to answer important open questions.
Collapse
Affiliation(s)
- Joshua D Brandstadter
- Division of Hematology-Oncology, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
9
|
Magee CN, Murakami N, Borges TJ, Shimizu T, Safa K, Ohori S, Cai S, Uffing A, Azzi J, Elyaman W, Charbonnier LM, Liu K, Toprak D, Visner G, Chatila TA, Siebel CW, Najafian N, Riella LV. Notch-1 Inhibition Promotes Immune Regulation in Transplantation Via Regulatory T Cell-Dependent Mechanisms. Circulation 2019; 140:846-863. [PMID: 31266349 DOI: 10.1161/circulationaha.119.040563] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Transplantation is the treatment of choice for many patients with end-stage organ disease. Despite advances in immunosuppression, long-term outcomes remain suboptimal, hampered by drug toxicity and immune-mediated injury, the leading cause of late graft loss. The development of therapies that promote regulation while suppressing effector immunity is imperative to improve graft survival and minimize conventional immunosuppression. Notch signaling is a highly conserved pathway pivotal to T-cell differentiation and function, rendering it a target of interest in efforts to manipulate T cell-mediated immunity. METHODS We investigated the pattern of Notch-1 expression in effector and regulatory T cells (Tregs) in both murine and human recipients of a solid-organ transplant. Using a selective human anti-Notch-1 antibody (aNotch-1), we examined the effect of Notch-1 receptor inhibition in full major histocompatibility complex-mismatch murine cardiac and lung transplant models, and in a humanized skin transplant model. On the basis of our findings, we further used a genetic approach to investigate the effect of selective Notch-1 inhibition in Tregs. RESULTS We observed an increased proportion of Tregs expressing surface and intracellular (activated) Notch-1 in comparison with conventional T cells, both in mice with transplants and in the peripheral blood of patients with transplants. In the murine cardiac transplant model, peritransplant administration of aNotch-1 (days 0, 2, 4, 6, 8, and 10) significantly prolonged allograft survival in comparison with immunoglobulin G-treated controls. Similarly, aNotch-1 treatment improved both histological and functional outcomes in the murine lung transplant model. The use of aNotch-1 resulted in a reduced proportion of both splenic and intragraft conventional T cells, while increasing the proportion of Tregs. Furthermore, Tregs isolated from aNotch-1-treated mice showed enhanced suppressive function on a per-cell basis, confirmed with selective Notch-1 deletion in Tregs (Foxp3EGFPCreNotch1fl/fl). Notch-1 blockade inhibited the mammalian target of rapamycin pathway and increased the phosphorylation of STAT5 (signal transducer and activator of transcription 5) in murine Tregs. Notch-1low Tregs isolated from human peripheral blood exhibited more potent suppressive capacity than Notch-1high Tregs. Last, the combination of aNotch-1 with costimulation blockade induced long-term tolerance in a cardiac transplant model, and this tolerance was dependent on CTLA-4 (cytotoxic T-lymphocyte-associated antigen-4) signaling. CONCLUSIONS Our data reveal a promising, clinically relevant approach for immune modulation in transplantation by selectively targeting Notch-1.
Collapse
Affiliation(s)
- Ciara N Magee
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.).,University College London Department of Renal Medicine, Centre for Transplantation, Royal Free Hospital, United Kingdom (C.N.M.)
| | - Naoka Murakami
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Thiago J Borges
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Tetsunosuke Shimizu
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Kassem Safa
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Shunsuke Ohori
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Songjie Cai
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Audrey Uffing
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Jamil Azzi
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Wassim Elyaman
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY (W.E.)
| | - Louis-Marie Charbonnier
- Division of Immunology (L.-M.C., T.A.C.), Children's Hospital Boston, Harvard Medical School, MA
| | - Kaifeng Liu
- Pulmonary and Respiratory Diseases Division (K.L., G.V.), Children's Hospital Boston, Harvard Medical School, MA
| | - Demet Toprak
- Department of Pediatrics, Seattle Children's Hospital, WA (D.T.)
| | - Gary Visner
- Pulmonary and Respiratory Diseases Division (K.L., G.V.), Children's Hospital Boston, Harvard Medical School, MA
| | - Talal A Chatila
- Division of Immunology (L.-M.C., T.A.C.), Children's Hospital Boston, Harvard Medical School, MA
| | - Christian W Siebel
- Department of Molecular Biology, Genentech Inc, South San Francisco, CA (C.W.S.)
| | - Nader Najafian
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| | - Leonardo V Riella
- Transplantation Research Center, Brigham & Women's Hospital, Harvard Medical School, Boston, MA (C.N.M., N.M., T.J.B., T.S., K.S., S.O., S.C., A.U., J.A., N.N., L.V.R.)
| |
Collapse
|
10
|
Chung J, Radojcic V, Perkey E, Parnell TJ, Niknafs Y, Jin X, Friedman A, Labrecque N, Blazar BR, Brennan TV, Siebel CW, Maillard I. Early Notch Signals Induce a Pathogenic Molecular Signature during Priming of Alloantigen-Specific Conventional CD4 + T Cells in Graft-versus-Host Disease. THE JOURNAL OF IMMUNOLOGY 2019; 203:557-568. [PMID: 31182480 DOI: 10.4049/jimmunol.1900192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
Graft-versus-host disease (GVHD) is the most serious complication of allogeneic hematopoietic cell transplantation. Notch signals delivered during the first 48 h after transplantation drive proinflammatory cytokine production in conventional T cells (Tconv) and inhibit the expansion of regulatory T cells (Tregs). Short-term Notch inhibition induces long-term GVHD protection. However, it remains unknown whether Notch blockade blunts GVHD through its effects on Tconv, Tregs, or both and what early Notch-regulated molecular events occur in alloantigen-specific T cells. To address these questions, we engineered T cell grafts to achieve selective Notch blockade in Tconv versus Tregs and evaluated their capacity to trigger GVHD in mice. Notch blockade in Tconv was essential for GVHD protection as GVHD severity was similar in the recipients of wild-type Tconv combined with Notch-deprived versus wild-type Tregs. To identify the impact of Notch signaling on the earliest steps of T cell activation in vivo, we established a new acute GVHD model mediated by clonal alloantigen-specific 4C CD4+ Tconv. Notch-deprived 4C T cells had preserved early steps of activation, IL-2 production, proliferation, and Th cell polarization. In contrast, Notch inhibition dampened IFN-γ and IL-17 production, diminished mTORC1 and ERK1/2 activation, and impaired transcription of a subset of Myc-regulated genes. The distinct Notch-regulated signature had minimal overlap with known Notch targets in T cell leukemia and developing T cells, highlighting the specific impact of Notch signaling in mature T cells. Our findings uncover a unique molecular program associated with the pathogenic effects of Notch in T cells at the earliest stages of GVHD.
Collapse
Affiliation(s)
- Jooho Chung
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Vedran Radojcic
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.,Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT 84112
| | - Eric Perkey
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Timothy J Parnell
- Huntsman Cancer Institute Bioinformatic Analysis Shared Resource, University of Utah, Salt Lake City, UT 84112
| | - Yashar Niknafs
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xi Jin
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Ann Friedman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Nathalie Labrecque
- Centre de Recherche Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, Quebec H1T 2M4, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec H3T IJ4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3T IJ4, Canada
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | - Todd V Brennan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | - Ivan Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109; .,Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109.,Division of Hematology-Oncology, Department of Internal Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
11
|
Norum HM, Michelsen AE, Lekva T, Arora S, Otterdal K, Olsen MB, Kong XY, Gude E, Andreassen AK, Solbu D, Karason K, Dellgren G, Gullestad L, Aukrust P, Ueland T. Circulating delta-like Notch ligand 1 is correlated with cardiac allograft vasculopathy and suppressed in heart transplant recipients on everolimus-based immunosuppression. Am J Transplant 2019; 19:1050-1060. [PMID: 30312541 DOI: 10.1111/ajt.15141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/02/2018] [Accepted: 10/07/2018] [Indexed: 02/06/2023]
Abstract
Cardiac allograft vasculopathy (CAV) causes heart failure after heart transplantation (HTx), but its pathogenesis is incompletely understood. Notch signaling, possibly modulated by everolimus (EVR), is essential for processes involved in CAV. We hypothesized that circulating Notch ligands would be dysregulated after HTx. We studied circulating delta-like Notch ligand 1 (DLL1) and periostin (POSTN) and CAV in de novo HTx recipients (n = 70) randomized to standard or EVR-based, calcineurin inhibitor-free immunosuppression and in maintenance HTx recipients (n = 41). Compared to healthy controls, plasma DLL1 and POSTN were elevated in de novo (P < .01; P < .001) and maintenance HTx recipients (P < .001; P < .01). Use of EVR was associated with a treatment effect for DLL1. For de novo HTx recipients, a change in DLL1 correlated with a change in CAV at 1 (P = .021) and 3 years (P = .005). In vitro, activation of T cells increased DLL1 secretion, attenuated by EVR. In vitro data suggest that also endothelial cells and vascular smooth muscle cells (VSMCs) could contribute to circulating DLL1. Immunostaining of myocardial specimens showed colocalization of DLL1 with T cells, endothelial cells, and VSMCs. Our findings suggest a role of DLL1 in CAV progression, and that the beneficial effect of EVR on CAV could reflect a suppressive effect on DLL1. Trial registration numbers-SCHEDULE trial: ClinicalTrials.gov NCT01266148; NOCTET trial: ClinicalTrials.gov NCT00377962.
Collapse
Affiliation(s)
- Hilde M Norum
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Medical Faculty, University of Oslo, Oslo, Norway.,Division of Emergencies and Critical Care, Department for Research and Development, Oslo University Hospital, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Medical Faculty, University of Oslo, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Satish Arora
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Center for Heart Failure Research, Medical Faculty, University of Oslo, Oslo, Norway
| | - Kari Otterdal
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Medical Faculty, University of Oslo, Oslo, Norway
| | - Einar Gude
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Arne K Andreassen
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Kristjan Karason
- Sahlgrenska University Hospital, Transplant Institute, Gothenburg, Sweden.,Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Göran Dellgren
- Sahlgrenska University Hospital, Transplant Institute, Gothenburg, Sweden.,Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lars Gullestad
- Institute of Clinical Medicine, Medical Faculty, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Medical Faculty, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, Medical Faculty, University of Oslo, Oslo, Norway.,K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| |
Collapse
|
12
|
Qin Y, Sun B, Zhang F, Wang Y, Shen B, Liu Y, Guo Y, Fan Y, Qiu J. Sox7 is involved in antibody-dependent endothelial cell activation and renal allograft injury via the Jagged1-Notch1 pathway. Exp Cell Res 2019; 375:20-27. [PMID: 30639059 DOI: 10.1016/j.yexcr.2019.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/28/2018] [Accepted: 01/08/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Antibody-mediated rejection (AMR) can cause graft loss and reduces long-term graft survival after kidney transplantation. Human leukocyte antigen (HLA) and/or non-HLA antibodies play a key role in the pathogenesis of AMR by targeting the allograft epithelium via complement activation and complement-independent mechanisms. However, the precise mechanisms of AMR remain unclear and treatment is still limited. METHODS In this study, we investigated the role of the endothelial-associated transcription factor Sox7 in AMR, using the anti-HLA antibody W6/32, shRNA-mediated Sox7 knockdown, and by manipulating the Notch pathway. We used an in vitro human kidney glomerular endothelial cells (HKGECs) model and an in vivo MHC-mismatched kidney transplantation model. RESULTS Sox7 expression was upregulated and the Jagged1-Notch1 pathway was activated in HKGECs after W6/32 activation. W6/32 antibodies increased the expression of adhesion molecules (VCAM-1, ICAM-1), inflammatory cytokines (IL-6, TNF-α), and chemokines (CXCL8, CXCL10), and Sox7 knockdown and inhibition of the Notch pathway by DAPT significantly reduced these effects. Jagged1 overexpression rescued the inhibitory effects of Sox7 knockdown. In addition, Sox7 knockdown attenuated acute allograft kidney injury in mice and reduced the expression of adhesion molecules and Jagged1-Notch1 signaling after transplantation. CONCLUSIONS Our findings suggest that Sox7 plays an important role in mediating HLA I antibody-dependent endothelial cell activation and acute kidney allograft rejection via the Jagged1-Notch1 pathway. Manipulating Sox7 in donor organs may represent a useful treatment for AMR in solid organ transplantation.
Collapse
Affiliation(s)
- Yan Qin
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Bo Sun
- Shanghai Center for Drug Evaluation & Inspection, Shanghai 201203, China
| | - Fang Zhang
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yong Wang
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Bing Shen
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yong Liu
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yifeng Guo
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Yu Fan
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Jianxin Qiu
- Department of Kidney Transplantation & Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China.
| |
Collapse
|
13
|
Radojcic V, Paz K, Chung J, Du J, Perkey ET, Flynn R, Ivcevic S, Zaiken M, Friedman A, Yan M, Pletneva MA, Sarantopoulos S, Siebel CW, Blazar BR, Maillard I. Notch signaling mediated by Delta-like ligands 1 and 4 controls the pathogenesis of chronic GVHD in mice. Blood 2018; 132:2188-2200. [PMID: 30181175 PMCID: PMC6238189 DOI: 10.1182/blood-2018-03-841155] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic graft-versus-host disease (cGVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT) and remains an area of unmet clinical need with few treatment options available. Notch blockade prevents acute GVHD in multiple mouse models, but the impact of Notch signaling on cGVHD remains unknown. Using genetic and antibody-mediated strategies of Notch inhibition, we investigated the role of Notch signaling in complementary mouse cGVHD models that mimic several aspects of human cGVHD in search of candidate therapeutics. In the B10.D2→BALB/c model of sclerodermatous cGVHD, Delta-like ligand 4 (Dll4)-driven Notch signaling was essential for disease development. Antibody-mediated Dll4 inhibition conferred maximum benefits when pursued early in a preventative fashion, with anti-Dll1 enhancing early protection. Notch-deficient alloantigen-specific T cells showed no early defects in proliferation or helper polarization in vivo but subsequently exhibited markedly decreased cytokine secretion and enhanced accumulation of FoxP3+ regulatory T cells. In the B6→B10.BR major histocompatibility complex-mismatched model with multi-organ system cGVHD and prominent bronchiolitis obliterans (BO), but not skin manifestations, absence of Notch signaling in T cells provided long-lasting disease protection that was replicated by systemic targeting of Dll1, Dll4, or both Notch ligands, even during established disease. Notch inhibition decreased target organ damage and germinal center formation. Moreover, decreased BO-cGVHD was observed upon inactivation of Notch1 and/or Notch2 in T cells. Systemic targeting of Notch2 alone was safe and conferred therapeutic benefits. Altogether, Notch ligands and receptors regulate key pathogenic steps in cGVHD and emerge as novel druggable targets to prevent or treat different forms of cGVHD.
Collapse
Affiliation(s)
- Vedran Radojcic
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Katelyn Paz
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Jooho Chung
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Jing Du
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Eric T Perkey
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI
| | - Ryan Flynn
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Sanja Ivcevic
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Michael Zaiken
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Ann Friedman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Minhong Yan
- Department of Discovery Oncology, Genentech, South San Francisco, CA
| | | | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC; and
| | | | - Bruce R Blazar
- Division of Blood and Marrow Transplant, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
14
|
Black CK, Termanini KM, Aguirre O, Hawksworth JS, Sosin M. Solid organ transplantation in the 21 st century. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:409. [PMID: 30498736 PMCID: PMC6230860 DOI: 10.21037/atm.2018.09.68] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/29/2018] [Indexed: 12/20/2022]
Abstract
Solid organ transplantation (SOT) has emerged from an experimental approach in the 20th century to now being an established and practical definitive treatment option for patients with end-organ dysfunction. The evolution of SOT has seen the field progress rapidly over the past few decades with incorporation of a variety of solid organs-liver, kidney, pancreas, heart, and lung-into the donor pool. New advancements in surgical technique have allowed for more efficient and refined multi-organ procurements with minimal complications and decreased ischemic injury events. Additionally, immunosuppression therapy has also seen advancements with the expansion of immunosuppressive protocols to dampen the host immune response and improve short and long-term graft survival. However, the field of SOT faces new barriers, most importantly the expanding demand for SOT that is outpacing the current supply. Allocation protocols have been developed in an attempt to address these concerns. Other avenues for SOT are also being explored to increase the donor pool, including split-liver donor transplants, islet cell implantation for pancreas transplants, and xenotransplantation. The future of SOT is bright with exciting new research being explored to overcome current obstacles.
Collapse
Affiliation(s)
- Cara K. Black
- Georgetown University School of Medicine, Washington, DC, USA
| | | | - Oswaldo Aguirre
- MedStar Georgetown University Hospital, MedStar Georgetown Transplant Institute, Washington, DC, USA
| | - Jason S. Hawksworth
- MedStar Georgetown University Hospital, MedStar Georgetown Transplant Institute, Washington, DC, USA
| | - Michael Sosin
- Hansjörg Wyss Department of Plastic Surgery, NYU Langone Health, New York, NY, USA
| |
Collapse
|
15
|
Perkey E, Maillard I. New Insights into Graft-Versus-Host Disease and Graft Rejection. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:219-245. [PMID: 29099650 DOI: 10.1146/annurev-pathol-020117-043720] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Allogeneic transplantation of foreign organs or tissues has lifesaving potential, but can lead to serious complications. After solid organ transplantation, immune-mediated rejection mandates the use of prolonged global immunosuppression and limits the life span of transplanted allografts. After bone marrow transplantation, donor-derived immune cells can trigger life-threatening graft-versus-host disease. T cells are central mediators of alloimmune complications and the target of most existing therapeutic interventions. We review recent progress in identifying multiple cell types in addition to T cells and new molecular pathways that regulate pathogenic alloreactivity. Key discoveries include the cellular subsets that function as potential sources of alloantigens, the cross talk of innate lymphoid cells with damaged epithelia and with the recipient microbiome, the impact of the alarmin interleukin-33 on alloreactivity, and the role of Notch ligands expressed by fibroblastic stromal cells in alloimmunity. While refining our understanding of transplantation immunobiology, these findings identify new therapeutic targets and new areas of investigation.
Collapse
Affiliation(s)
- Eric Perkey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA;
| | - Ivan Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, USA; .,Department of Internal Medicine, Division of Hematology-Oncology, and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Medicine, Division of Hematology-Oncology, and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
16
|
Neal LM, Qiu Y, Chung J, Xing E, Cho W, Malachowski AN, Sandy-Sloat AR, Osterholzer JJ, Maillard I, Olszewski MA. T Cell-Restricted Notch Signaling Contributes to Pulmonary Th1 and Th2 Immunity during Cryptococcus neoformans Infection. THE JOURNAL OF IMMUNOLOGY 2017; 199:643-655. [PMID: 28615417 DOI: 10.4049/jimmunol.1601715] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 05/17/2017] [Indexed: 12/31/2022]
Abstract
Cryptococcus neoformans is a ubiquitous, opportunistic fungal pathogen but the cell signaling pathways that drive T cell responses regulating antifungal immunity are incompletely understood. Notch is a key signaling pathway regulating T cell development, and differentiation and functional responses of mature T cells in the periphery. The targeting of Notch signaling within T cells has been proposed as a potential treatment for alloimmune and autoimmune disorders, but it is unknown whether disturbances to T cell immunity may render these patients vulnerable to fungal infections. To elucidate the role of Notch signaling during fungal infections, we infected mice expressing the pan-Notch inhibitor dominant negative mastermind-like within mature T cells with C. neoformans Inhibition of T cell-restricted Notch signaling increased fungal burdens in the lungs and CNS, diminished pulmonary leukocyte recruitment, and simultaneously impaired Th1 and Th2 responses. Pulmonary leukocyte cultures from T cell Notch-deprived mice produced less IFN-γ, IL-5, and IL-13 than wild-type cells. This correlated with lower frequencies of IFN-γ-, IL-5-, and IL-13-producing CD4+ T cells, reduced expression of Th1 and Th2 associated transcription factors, Tbet and GATA3, and reduced production of IFN-γ by CD8+ T cells. In contrast, Th17 responses were largely unaffected by Notch signaling. The changes in T cell responses corresponded with impaired macrophage activation and reduced leukocyte accumulation, leading to diminished fungal control. These results identify Notch signaling as a previously unappreciated regulator of Th1 and Th2 immunity and an important element of antifungal defenses against cryptococcal infection and CNS dissemination.
Collapse
Affiliation(s)
- Lori M Neal
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109.,Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| | - Yafeng Qiu
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| | - Jooho Chung
- Graduate Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, MI 48109.,Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109
| | - Enze Xing
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| | - Woosung Cho
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| | | | | | - John J Osterholzer
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109.,Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| | - Ivan Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109.,Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109; and.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Michal A Olszewski
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109; .,Research Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105
| |
Collapse
|
17
|
Abstract
Solid organ and allogeneic hematopoietic cell transplantation have become standard therapeutic interventions that save patient lives and improve quality of life. Our enhanced understanding of transplantation immunobiology has refined clinical management and improved outcomes. However, organ rejection and graft-versus-host disease remain major obstacles to the broader successful application of these therapeutic procedures. Notch signaling regulates multiple aspects of adaptive and innate immunity. Preclinical studies identified Notch signaling as a promising target in autoimmune diseases, as well as after allogeneic hematopoietic cell and solid organ transplantation. Notch was found to be a central regulator of alloreactivity across clinically relevant models of transplantation. Notch inhibition in T cells prevented graft-versus-host disease and organ rejection, establishing organ tolerance by skewing CD4 T helper polarization away from a proinflammatory response toward suppressive regulatory T cells. Notch ligand blockade also dampened alloantibody deposition and prevented chronic rejection through humoral mechanisms. Toxicities of systemic Notch blockade were observed with γ-secretase inhibitors in preclinical and early clinical trials across different indications, but they did not arise upon preclinical targeting of Delta-like Notch ligands, a strategy sufficient to confer full benefits of Notch ablation in T cell alloimmunity. Because multiple clinical grade reagents have been developed to target individual Notch ligands and receptors, the benefits of Notch blockade in transplantation are calling for translation of preclinical findings into human transplantation medicine.
Collapse
|
18
|
Chung J, Ebens CL, Perkey E, Radojcic V, Koch U, Scarpellino L, Tong A, Allen F, Wood S, Feng J, Friedman A, Granadier D, Tran IT, Chai Q, Onder L, Yan M, Reddy P, Blazar BR, Huang AY, Brennan TV, Bishop DK, Ludewig B, Siebel CW, Radtke F, Luther SA, Maillard I. Fibroblastic niches prime T cell alloimmunity through Delta-like Notch ligands. J Clin Invest 2017; 127:1574-1588. [PMID: 28319044 PMCID: PMC5373885 DOI: 10.1172/jci89535] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Alloimmune T cell responses induce graft-versus-host disease (GVHD), a serious complication of allogeneic bone marrow transplantation (allo-BMT). Although Notch signaling mediated by Delta-like 1/4 (DLL1/4) Notch ligands has emerged as a major regulator of GVHD pathogenesis, little is known about the timing of essential Notch signals and the cellular source of Notch ligands after allo-BMT. Here, we have shown that critical DLL1/4-mediated Notch signals are delivered to donor T cells during a short 48-hour window after transplantation in a mouse allo-BMT model. Stromal, but not hematopoietic, cells were the essential source of Notch ligands during in vivo priming of alloreactive T cells. GVHD could be prevented by selective inactivation of Dll1 and Dll4 in subsets of fibroblastic stromal cells that were derived from chemokine Ccl19-expressing host cells, including fibroblastic reticular cells and follicular dendritic cells. However, neither T cell recruitment into secondary lymphoid organs nor initial T cell activation was affected by Dll1/4 loss. Thus, we have uncovered a pathogenic function for fibroblastic stromal cells in alloimmune reactivity that can be dissociated from their homeostatic functions. Our results reveal what we believe to be a previously unrecognized Notch-mediated immunopathogenic role for stromal cell niches in secondary lymphoid organs after allo-BMT and define a framework of early cellular and molecular interactions that regulate T cell alloimmunity.
Collapse
Affiliation(s)
- Jooho Chung
- Graduate Program in Cellular and Molecular Biology
- Life Sciences Institute, and
| | - Christen L. Ebens
- Life Sciences Institute, and
- Division of Hematology-Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric Perkey
- Graduate Program in Cellular and Molecular Biology
- Life Sciences Institute, and
| | - Vedran Radojcic
- Life Sciences Institute, and
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ute Koch
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Alexander Tong
- Medical Scientist Training Program and Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Frederick Allen
- Medical Scientist Training Program and Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sherri Wood
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jiane Feng
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | - Qian Chai
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Minhong Yan
- Genentech, South San Francisco, California, USA
| | - Pavan Reddy
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alex Y. Huang
- Medical Scientist Training Program and Division of Pediatric Hematology-Oncology, Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | - Todd V. Brennan
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - D. Keith Bishop
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | - Freddy Radtke
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sanjiv A. Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Ivan Maillard
- Life Sciences Institute, and
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Choy L, Hagenbeek TJ, Solon M, French D, Finkle D, Shelton A, Venook R, Brauer MJ, Siebel CW. Constitutive NOTCH3 Signaling Promotes the Growth of Basal Breast Cancers. Cancer Res 2017; 77:1439-1452. [DOI: 10.1158/0008-5472.can-16-1022] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 12/01/2016] [Accepted: 12/19/2016] [Indexed: 11/16/2022]
|
20
|
Murakami N, Maillard I, Riella LV. Notch Signaling and Immune Regulation in Alloimmunity. CURRENT TRANSPLANTATION REPORTS 2016; 3:294-302. [PMID: 29977738 DOI: 10.1007/s40472-016-0126-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Notch signaling plays a pivotal role in the differentiation and fate determination of T cells, B cells, dendritic cells (DCs) and innate lymphoid cells (ILCs). Recent gene-targeting and antibody approaches advanced our knowledge of the importance of Notch signaling in fine-tuning the peripheral immune response. Here we review current knowledge of the Notch pathway, focusing on solid organ transplant and graft-versus-host disease preclinical models, and discuss the potential of targeting Notch to suppress the immune response and improve transplant outcomes.
Collapse
Affiliation(s)
- Naoka Murakami
- Transplantation Research Center, Renal Division, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| | - Ivan Maillard
- Life Sciences Institute and Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Leonardo V Riella
- Transplantation Research Center, Renal Division, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
21
|
Chung J, Riella LV, Maillard I. Targeting the Notch Pathway to Prevent Rejection. Am J Transplant 2016; 16:3079-3085. [PMID: 27037759 PMCID: PMC7017453 DOI: 10.1111/ajt.13816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 02/23/2016] [Accepted: 03/24/2016] [Indexed: 01/25/2023]
Abstract
Immune rejection is mediated by a complex interplay of cellular and humoral mechanisms. Current therapeutic strategies, which rely on global immunosuppression, can result in serious complications and are not completely effective. Notch signaling is a cell-to-cell communication pathway that plays an important role during T cell development and in the regulation of peripheral immune responses. Initial work, performed mainly through gain-of-function approaches, paradoxically identified Notch as an inducer of tolerance; however, recent studies using loss-of-function approaches in mouse models of transplant rejection and graft-versus-host disease have clarified an important role for Notch as a central mediator of T cell alloreactivity. Short-term inhibition of individual Notch ligands in the peritransplant period had long-lasting protective effects. In a vascularized heart allograft model, blockade of Delta-like Notch ligands dampened both cellular and humoral rejection. In this minireview, we summarize current knowledge about the role of Notch signaling during allograft rejection and provide an overarching mechanism through which Notch acts to promote T cell pathogenicity and allograft damage. We propose that targeting elements of the Notch pathway could provide a new therapeutic approach to prevent allograft rejection.
Collapse
Affiliation(s)
- J. Chung
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI,Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - L. V. Riella
- Schuster Transplantation Research Center, Harvard Medical School, Boston, MA,Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - I. Maillard
- Life Sciences Institute, University of Michigan, Ann Arbor, MI,Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI,Corresponding author: Ivan Maillard,
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The Notch signaling pathway is known to play a pivotal role in T- and B-cell development and fate, presenting it as an attractive therapeutic target in alloimmunity. This review provides an overview of the mechanisms of Notch signaling, focusing on new insights into its diverse functions in T-cell activation, differentiation and memory subset formation, and the consequences thereof in transplantation. RECENT FINDINGS Recent evidence has shown that while not critical for early antigen-specific CD4 T-cell activation, Notch signaling regulates the survival of memory CD4 T cells via control of glycolytic metabolism; in contrast, Notch signaling is critical for the generation of short-lived CD8 effector T cells, but not memory CD8 cells. Transient, selective inhibition of various Notch receptors and ligands in models of solid organ transplantation has been shown to successfully modulate the alloimmune response, affecting the balance between effector and regulatory cells, with particular influence on the natural regulatory T-cell population. SUMMARY These studies reveal diverse roles for individual Notch receptors and ligands in peripheral immunity and indicate that selective targeting of the Notch pathway is a promising, novel approach for immune modulation in transplantation; the advent of therapeutic human antibodies to neutralize both the Notch ligands and the individual Notch receptors suggests that this approach could be efficiently developed.
Collapse
|
23
|
Donor-derived exosomes induce specific regulatory T cells to suppress immune inflammation in the allograft heart. Sci Rep 2016; 7:20077. [PMID: 26822278 PMCID: PMC4731812 DOI: 10.1038/srep20077] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/09/2015] [Indexed: 02/08/2023] Open
Abstract
To inhibit the immune inflammation in the allografts can be beneficial to organ transplantation. This study aims to induce the donor antigen specific regulatory T cells (Treg cell) inhibit the immune inflammation in the allograft heart. In this study, peripheral exosomes were purified from the mouse serum. A heart transplantation mouse model was developed. The immune inflammation of the allograft heart was assessed by histology and flow cytometry. The results showed that the donor antigen-specific T helper (Th)2 pattern inflammation was observed in the allograft hearts; the inflammation was inhibited by immunizing the recipient mice with the donor-derived exosomes. Purified peripheral exosomes contained integrin MMP1a; the latter induced CD4+ T cells to express Fork head protein-3 and transforming growth factor (TGF)-β via inhibiting the Th2 transcription factor, GATA binding protein 3, in CD4+ T cells. Administration with the donor-derived exosomes significantly prolonged the allograft heart survival. We conclude that the donor-derived peripheral exosomes have the capacity to inhibit the immune inflammation in the allograft heart via inducing specific Treg cells, implicating that administration with the donor-derived exosomes may be beneficial to cardiac transplantation.
Collapse
|
24
|
Pabois A, Pagie S, Gérard N, Laboisse C, Pattier S, Hulin P, Nedellec S, Toquet C, Charreau B. Notch signaling mediates crosstalk between endothelial cells and macrophages via Dll4 and IL6 in cardiac microvascular inflammation. Biochem Pharmacol 2016; 104:95-107. [PMID: 26826491 DOI: 10.1016/j.bcp.2016.01.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
Although short-term outcomes have improved with modern era immunosuppression, little progress has been made in long-term graft survival in cardiac transplantation. Antibody-mediated rejection (AMR) is one of the leading causes of graft failure and contributes significantly to poor long-term outcomes. Endothelial cell (EC) injury, intravascular macrophage infiltrate and microvascular inflammation are the histological features of AMR. Nevertheless, mechanisms of AMR remain unclear and treatment is still limited. Here, we investigated the mechanisms underlying vascular and inflammatory cell network involved in AMR at endothelial and macrophage levels, using endomyocardial transplant biopsies and EC/monocyte cocultures. First, we found that AMR associates with changes in Notch signaling at endothelium/monocyte interface including loss of endothelial Notch4 and the acquisition of the Notch ligand Dll4 in both cell types. We showed that endothelial Dll4 induces macrophage polarization into a pro-inflammatory fate (CD40(high)CD64(high)CD200R(low) HLA-DR(low)CD11b(low)) eliciting the production of IL-6. Dll4 and IL-6 are both Notch-dependent and are required for macrophage polarization through selective down and upregulation of M2- and M1-type markers, respectively. Overall, these findings highlight the impact of the graft's endothelium on macrophage recruitment and differentiation upon AMR via Notch signaling. We identified Dll4 and IL-6 as coregulators of vascular inflammation in cardiac transplantation and as potential targets for immunotherapy.
Collapse
Affiliation(s)
- Angélique Pabois
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Sylvain Pagie
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France
| | - Nathalie Gérard
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France
| | | | - Sabine Pattier
- Service de transplantation cardiaque, CHU de Nantes, Nantes F44000, France
| | - Philippe Hulin
- LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France; Plateforme MicroPICell SFR Santé - IRT, Nantes, France
| | - Steven Nedellec
- LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France; Plateforme MicroPICell SFR Santé - IRT, Nantes, France
| | - Claire Toquet
- Service d'Anatomie Pathologique, CHU de Nantes, Nantes F44000, France
| | - Béatrice Charreau
- INSERM UMR1064, Centre de Recherche en Transplantation et Immunologie, LabEx IGO and LabEx Transplantex, Nantes F44000, France; CHU de Nantes, Institut de Transplantation-Urologie-Néphrologie, ITUN, Nantes F44000, France; LUNAM Université de Nantes, Faculté de Médecine, Nantes F44000, France.
| |
Collapse
|
25
|
Amsen D, Helbig C, Backer RA. Notch in T Cell Differentiation: All Things Considered. Trends Immunol 2015; 36:802-814. [PMID: 26617322 DOI: 10.1016/j.it.2015.10.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 10/16/2015] [Accepted: 10/18/2015] [Indexed: 12/24/2022]
Abstract
Differentiation of naïve T cells into effector cells is required for optimal protection against different classes of microbial pathogen and for the development of immune memory. Recent findings have revealed important roles for the Notch signaling pathway in T cell differentiation into all known effector subsets, raising the question of how this pathway controls such diverse differentiation programs. Studies in preclinical models support the therapeutic potential of manipulating the Notch pathway to alleviate immune pathology, highlighting the importance of understanding the mechanisms through which Notch regulates T cell differentiation and function. We review these findings here, and outline both unifying principles involved in Notch-mediated T cell fate decisions and cell type- and context-specific differences that may present the most suitable points for therapeutic intervention.
Collapse
Affiliation(s)
- Derk Amsen
- Department of Hematopoiesis, Sanquin and Landsteiner Laboratory at the CLB, Plesmanlaan125, 1066CX, Amsterdam, the Netherlands.
| | - Christina Helbig
- Department of Hematopoiesis, Sanquin and Landsteiner Laboratory at the CLB, Plesmanlaan125, 1066CX, Amsterdam, the Netherlands
| | - Ronald A Backer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
26
|
Koga JI, Nakano T, Dahlman JE, Figueiredo JL, Zhang H, Decano J, Khan OF, Niida T, Iwata H, Aster JC, Yagita H, Anderson DG, Ozaki CK, Aikawa M. Macrophage Notch Ligand Delta-Like 4 Promotes Vein Graft Lesion Development: Implications for the Treatment of Vein Graft Failure. Arterioscler Thromb Vasc Biol 2015; 35:2343-2353. [PMID: 26404485 DOI: 10.1161/atvbaha.115.305516] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Despite its large clinical impact, the underlying mechanisms for vein graft failure remain obscure and no effective therapeutic solutions are available. We tested the hypothesis that Notch signaling promotes vein graft disease. APPROACH AND RESULTS We used 2 biotherapeutics for Delta-like ligand 4 (Dll4), a Notch ligand: (1) blocking antibody and (2) macrophage- or endothelial cell (EC)-targeted small-interfering RNA. Dll4 antibody administration for 28 days inhibited vein graft lesion development in low-density lipoprotein (LDL) receptor-deficient (Ldlr(-/-)) mice, and suppressed macrophage accumulation and macrophage expression of proinflammatory M1 genes. Dll4 antibody treatment for 7 days after grafting also reduced macrophage burden at day 28. Dll4 silencing via macrophage-targeted lipid nanoparticles reduced lesion development and macrophage accumulation, whereas EC-targeted Dll4 small-interfering RNA produced no effects. Gain-of-function and loss-of-function studies suggested in vitro that Dll4 induces proinflammatory molecules in macrophages. Macrophage Dll4 also stimulated smooth muscle cell proliferation and migration and suppressed their differentiation. CONCLUSIONS These results suggest that macrophage Dll4 promotes lesion development in vein grafts via macrophage activation and crosstalk between macrophages and smooth muscle cells, supporting the Dll4-Notch axis as a novel therapeutic target.
Collapse
Affiliation(s)
- Jun-Ichiro Koga
- The Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Toshiaki Nakano
- The Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - James E Dahlman
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA.,Institutes for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Jose-Luiz Figueiredo
- The Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hengmin Zhang
- the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Julius Decano
- the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Omar F Khan
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA.,Institutes for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Tomiharu Niida
- The Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hiroshi Iwata
- the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA.,Institutes for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - C Keith Ozaki
- Division of Vascular and Endovascular Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- The Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,the Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|