1
|
Weijler AM, Wekerle T. Combining Treg Therapy With Donor Bone Marrow Transplantation: Experimental Progress and Clinical Perspective. Transplantation 2024; 108:1100-1108. [PMID: 37789519 DOI: 10.1097/tp.0000000000004814] [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] [Indexed: 10/05/2023]
Abstract
Donor-specific tolerance remains a goal in transplantation because it could improve graft survival and reduce morbidity. Cotransplantation of donor hematopoietic cells to achieve chimerism is a promising approach for tolerance induction, which was successfully tested in clinical trials. However, current protocols are associated with side effects related to the myelosuppressive recipient conditioning, which makes it difficult to introduce them as standard therapy. More recently, adoptive cell therapy with polyclonal or donor-specific regulatory T cells (Treg) proved safe and feasible in several transplant trials, but it is unclear whether it can induce tolerance on its own. The combination of both approaches-Treg therapy and hematopoietic cell transplantation-leads to chimerism and tolerance without myelosuppressive treatment in murine models. Treg therapy promotes engraftment of allogeneic hematopoietic cells, reducing conditioning requirements and enhancing regulatory mechanisms maintaining tolerance. This review discusses possible modes of action of transferred Treg in experimental chimerism models and describes translational efforts investigating the potent synergy of Treg and chimerism.
Collapse
Affiliation(s)
- Anna Marianne Weijler
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
2
|
Spicer JA, Huttunen KM, Jose J, Dimitrov I, Akhlaghi H, Sutton VR, Voskoboinik I, Trapani J. Small Molecule Inhibitors of Lymphocyte Perforin as Focused Immunosuppressants for Infection and Autoimmunity. J Med Chem 2022; 65:14305-14325. [PMID: 36263926 DOI: 10.1021/acs.jmedchem.2c01338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
New drugs that precisely target the immune mechanisms critical for cytotoxic T lymphocyte (CTL) and natural killer (NK) cell driven pathologies are desperately needed. In this perspective, we explore the cytolytic protein perforin as a target for therapeutic intervention. Perforin plays an indispensable role in CTL/NK killing and controls a range of immune pathologies, while being encoded by a single copy gene with no redundancy of function. An immunosuppressant targeting this protein would provide the first-ever therapy focused specifically on one of the principal cell death pathways contributing to allotransplant rejection and underpinning multiple autoimmune and postinfectious diseases. No drugs that selectively block perforin-dependent cell death are currently in clinical use, so this perspective will review published novel small molecule inhibitors, concluding with in vivo proof-of-concept experiments performed in mouse models of perforin-mediated immune pathologies that provide a potential pathway toward a clinically useful therapeutic agent.
Collapse
Affiliation(s)
- Julie A Spicer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1142, New Zealand
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jiney Jose
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1142, New Zealand
| | - Ivo Dimitrov
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1142, New Zealand
| | - Hedieh Akhlaghi
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Vivien R Sutton
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ilia Voskoboinik
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Joseph Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
3
|
del Rio ML, Perez-Simon JA, Rodriguez-Barbosa JI. Differential Engraftment of Parental A20 PD-L1 WT and PD-L1 KO Leukemia Cells in Semiallogeneic Recipients in the Context of PD-L1/PD-1 Interaction and NK Cell-Mediated Hybrid Resistance. Front Immunol 2022; 13:887348. [PMID: 35795681 PMCID: PMC9251058 DOI: 10.3389/fimmu.2022.887348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022] Open
Abstract
The contribution of natural killer (NK) cells to tumor rejection in the context of programmed death-ligand 1/programmed death 1 (PD-L1/PD-1) blockade is a matter of intense debate. To elucidate the role of PD-L1 expression on tumor cells and the functional consequences of engaging PD-1 receptor on cytotoxic cells, PD-L1 expression was genetically inactivated and WT or PD-L1-deficient parental tumor cells were adoptively transferred intravenously into F1 recipients. The engraftment of PD-L1-deficient A20 tumor cells in the spleen and liver of F1 recipients was impaired compared with A20 PD-L1 WT tumor counterparts. To elucidate the mechanism responsible for this differential tumor engraftment and determine the relevance of the role of the PD-L1/PD-1 pathway in the interplay of tumor cells/NK cells, a short-term competitive tumor implantation assay in the peritoneal cavity of semiallogeneic F1 recipients was designed. The results presented herein showed that NK cells killed target tumor cells with similar efficiency regardless of PD-L1 expression, whereas PD-L1 expression on A20 tumor cells conferred significant tumor protection against rejection by CD8 T cells confirming the role of the co-inhibitory receptor PD-1 in the modulation of their cytotoxic activity. In summary, PD-L1 expression on A20 leukemia tumor cells modulates CD8 T-cell-mediated responses to tumor-specific antigens but does not contribute to inhibit NK cell-mediated hybrid resistance, which correlates with the inability to detect PD-1 expression on NK cells neither under steady-state conditions nor under inflammatory conditions.
Collapse
Affiliation(s)
- Maria-Luisa del Rio
- Transplantation Immunobiology and Immunotherapy Section, Institute of Molecular Biology, University of Leon, Leon, Spain
- CIBERONC Consortium, Accion Estrategica en Salud, Spain
| | - Jose-Antonio Perez-Simon
- CIBERONC Consortium, Accion Estrategica en Salud, Spain
- Department of Hematology, University Hospital Virgen del Rocio/Institute of Biomedicine [Instituto de Biomedicina de Sevilla (IBIS)/Centro Superior de Investigaciones Científicas (CSIC)/Centro de Investigación Biomédica en Red Cáncer (CIBERONC)], Seville, Spain
| | - Jose-Ignacio Rodriguez-Barbosa
- Transplantation Immunobiology and Immunotherapy Section, Institute of Molecular Biology, University of Leon, Leon, Spain
- CIBERONC Consortium, Accion Estrategica en Salud, Spain
| |
Collapse
|
4
|
Gartlan KH, Jaiswal JK, Bull MR, Akhlaghi H, Sutton VR, Alexander KA, Chang K, Hill GR, Miller CK, O'Connor PD, Jose J, Trapani JA, Charman SA, Spicer JA, Jamieson SMF. Preclinical Activity and Pharmacokinetic/Pharmacodynamic Relationship for a Series of Novel Benzenesulfonamide Perforin Inhibitors. ACS Pharmacol Transl Sci 2022; 5:429-439. [PMID: 35711815 DOI: 10.1021/acsptsci.2c00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Indexed: 01/25/2023]
Abstract
Perforin is a key effector of lymphocyte-mediated cell death pathways and contributes to transplant rejection of immunologically mismatched grafts. We have developed a novel series of benzenesulfonamide (BZS) inhibitors of perforin that can mitigate graft rejection during allogeneic bone marrow/stem cell transplantation. Eight such perforin inhibitors were tested for their murine pharmacokinetics, plasma protein binding, and their ability to block perforin-mediated lysis in vitro and to block the rejection of major histocompatibility complex (MHC)-mismatched mouse bone marrow cells. All compounds showed >99% binding to plasma proteins and demonstrated perforin inhibitory activity in vitro and in vivo. A lead compound, compound 1, that showed significant increases in allogeneic bone marrow preservation was evaluated for its plasma pharmacokinetics and in vivo efficacy at multiple dosing regimens to establish a pharmacokinetic/pharmacodynamic (PK/PD) relationship. The strongest PK/PD correlation was observed between perforin inhibition in vivo and time that total plasma concentrations remained above 900 μM, which correlates to unbound concentrations similar to 3× the unbound in vitro IC90 of compound 1. This PK/PD relationship will inform future dosing strategies of BZS perforin inhibitors to maintain concentrations above 3× the unbound IC90 for as long as possible to maximize efficacy and enhance progression toward clinical evaluation.
Collapse
Affiliation(s)
- Kate H Gartlan
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Jagdish K Jaiswal
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Matthew R Bull
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Hedieh Akhlaghi
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia
| | - Vivien R Sutton
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Kylie A Alexander
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Karshing Chang
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Christian K Miller
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jiney Jose
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Julie A Spicer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
5
|
Isakov N. Histocompatibility and Reproduction: Lessons from the Anglerfish. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010113. [PMID: 35054506 PMCID: PMC8780861 DOI: 10.3390/life12010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 11/16/2022]
Abstract
Reproduction in certain deep-sea anglerfishes involves the permanent attachment of dwarf males to much larger females and fusion of their tissues leading to the establishment of a shared circulatory system. This unusual phenomenon of sexual parasitism enables anglerfishes to maximize reproductive success in the vast and deep oceans, where females and males otherwise rarely meet. An even more surprising phenomenon relates to the observation that joining of genetically disparate male and female anglerfishes does not evoke a strong anti-graft immune rejection response, which occurs in vertebrates following allogeneic parabiosis. Recent studies demonstrated that the evolutionary processes that led to the unique mating strategy of anglerfishes coevolved with genetic changes that resulted in loss of functional genes encoding critical components of the adaptive immune system. These genetic alterations enabled anglerfishes to tolerate the histoincompatible tissue antigens of their mate and prevent the occurrence of reciprocal graft rejection responses. While the exact mechanisms by which anglerfishes defend themselves against pathogens have not yet been deciphered, it is speculated that during evolution, anglerfishes adopted new immune strategies that compensate for the loss of B and T lymphocyte functions and enable them to resist infection by pathogens.
Collapse
Affiliation(s)
- Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| |
Collapse
|
6
|
Henden AS, Koyama M, Robb RJ, Forero A, Kuns RD, Chang K, Ensbey KS, Varelias A, Kazakoff SH, Waddell N, Clouston AD, Giri R, Begun J, Blazar BR, Degli-Esposti MA, Kotenko SV, Lane SW, Bowerman KL, Savan R, Hugenholtz P, Gartlan KH, Hill GR. IFN-λ therapy prevents severe gastrointestinal graft-versus-host disease. Blood 2021; 138:722-737. [PMID: 34436524 PMCID: PMC8667051 DOI: 10.1182/blood.2020006375] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Immunopathology and intestinal stem cell (ISC) loss in the gastrointestinal (GI) tract is the prima facie manifestation of graft-versus-host disease (GVHD) and is responsible for significant mortality after allogeneic bone marrow transplantation (BMT). Approaches to prevent GVHD to date focus on immune suppression. Here, we identify interferon-λ (IFN-λ; interleukin-28 [IL-28]/IL-29) as a key protector of GI GVHD immunopathology, notably within the ISC compartment. Ifnlr1-/- mice displayed exaggerated GI GVHD and mortality independent of Paneth cells and alterations to the microbiome. Ifnlr1-/- intestinal organoid growth was significantly impaired, and targeted Ifnlr1 deficiency exhibited effects intrinsic to recipient Lgr5+ ISCs and natural killer cells. PEGylated recombinant IL-29 (PEG-rIL-29) treatment of naive mice enhanced Lgr5+ ISC numbers and organoid growth independent of both IL-22 and type I IFN and modulated proliferative and apoptosis gene sets in Lgr5+ ISCs. PEG-rIL-29 treatment improved survival, reduced GVHD severity, and enhanced epithelial proliferation and ISC-derived organoid growth after BMT. The preservation of ISC numbers in response to PEG-rIL-29 after BMT occurred both in the presence and absence of IFN-λ-signaling in recipient natural killer cells. IFN-λ is therefore an attractive and rapidly testable approach to prevent ISC loss and immunopathology during GVHD.
Collapse
Affiliation(s)
- Andrea S Henden
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Renee J Robb
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Adriana Forero
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Rachel D Kuns
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Karshing Chang
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kathleen S Ensbey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Antiopi Varelias
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Stephen H Kazakoff
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicole Waddell
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Rabina Giri
- Mater Research Institute, The University of Queensland-Translational Research Institute, Brisbane, QLD, Australia
| | - Jakob Begun
- Mater Research Institute, The University of Queensland-Translational Research Institute, Brisbane, QLD, Australia
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Mariapia A Degli-Esposti
- Centre for Experimental Immunology, Lions Eye Institute, Perth, WA, Australia
- Infection and Immunity Program, Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sergei V Kotenko
- Center for Immunity and Inflammation, New Jersey Medical School, and
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ
| | - Steven W Lane
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kate L Bowerman
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia; and
| | - Ram Savan
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia; and
| | - Kate H Gartlan
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Division of Medical Oncology, The University of Washington, Seattle, WA
| |
Collapse
|
7
|
Cheong M, Gartlan KH, Lee JS, Tey SK, Zhang P, Kuns RD, Andoniou CE, Martins JP, Chang K, Sutton VR, Kelly G, Varelias A, Vuckovic S, Markey KA, Boyle GM, Smyth MJ, Engwerda CR, MacDonald KPA, Trapani JA, Degli-Esposti MA, Koyama M, Hill GR. ASC Modulates CTL Cytotoxicity and Transplant Outcome Independent of the Inflammasome. Cancer Immunol Res 2020; 8:1085-1098. [PMID: 32444423 DOI: 10.1158/2326-6066.cir-19-0653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 02/10/2020] [Accepted: 05/14/2020] [Indexed: 11/16/2022]
Abstract
The adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) is known to facilitate caspase-1 activation, which is essential for innate host immunity via the formation of the inflammasome complex, a multiprotein structure responsible for processing IL1β and IL18 into their active moieties. Here, we demonstrated that ASC-deficient CD8+ T cells failed to induce severe graft-versus-host disease (GVHD) and had impaired capacity for graft rejection and graft-versus-leukemia (GVL) activity. These effects were inflammasome independent because GVHD lethality was not altered in recipients of caspase-1/11-deficient T cells. We also demonstrated that ASC deficiency resulted in a decrease in cytolytic function, with a reduction in granzyme B secretion and CD107a expression by CD8+ T cells. Altogether, our findings highlight that ASC represents an attractive therapeutic target for improving outcomes of clinical transplantation.
Collapse
Affiliation(s)
- Melody Cheong
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,School of Natural Sciences, Griffith University, Nathan, Queensland, Australia
| | - Kate H Gartlan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Jason S Lee
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Siok-Keen Tey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,The Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Ping Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rachel D Kuns
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Christopher E Andoniou
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia.,Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jose Paulo Martins
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Karshing Chang
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Vivien R Sutton
- Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - Greg Kelly
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Slavica Vuckovic
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Institute of Haematology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Kate A Markey
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Glen M Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | - Joseph A Trapani
- Cancer Cell Death Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - Mariapia A Degli-Esposti
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia.,Infection and Immunity Program and Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Motoko Koyama
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia. .,Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
8
|
Spicer JA, Miller CK, O'Connor PD, Jose J, Giddens AC, Jaiswal JK, Jamieson SMF, Bull MR, Denny WA, Akhlaghi H, Trapani JA, Hill GR, Chang K, Gartlan KH. Inhibition of the Cytolytic Protein Perforin Prevents Rejection of Transplanted Bone Marrow Stem Cells in Vivo. J Med Chem 2020; 63:2229-2239. [PMID: 31525966 DOI: 10.1021/acs.jmedchem.9b00881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Perforin is a key effector protein in the vertebrate immune system and is secreted by cytotoxic T lymphocytes and natural killer cells to help eliminate virus-infected and transformed target cells. The ability to modulate perforin activity in vivo could be extremely useful, especially in the context of bone marrow stem cell transplantation where early rejection of immunologically mismatched grafts is driven by the recipient's natural killer cells, which overwhelmingly use perforin to kill their targets. Bone marrow stem cell transplantation is a potentially curative treatment for both malignant and nonmalignant disorders, but when the body recognizes the graft as foreign, it is rejected by this process, often with fatal consequences. Here we report optimization of a previously identified series of benzenesulfonamide-based perforin inhibitors for their physicochemical and pharmacokinetic properties, resulting in the identification of 16, the first reported small molecule able to prevent rejection of transplanted bone marrow stem cells in vivo by blocking perforin function.
Collapse
Affiliation(s)
- Julie A Spicer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - Christian K Miller
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - Jiney Jose
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - Anna C Giddens
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jagdish K Jaiswal
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Matthew R Bull
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, A New Zealand Centre for Research Excellence, Auckland 1010, New Zealand
| | - Hedieh Akhlaghi
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Geoff R Hill
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Karshing Chang
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| | - Kate H Gartlan
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Queensland 4006, Australia
| |
Collapse
|
9
|
NK cell recognition of hematopoietic cells by SLAM-SAP families. Cell Mol Immunol 2019; 16:452-459. [PMID: 30911116 DOI: 10.1038/s41423-019-0222-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/01/2019] [Indexed: 01/07/2023] Open
Abstract
The signaling lymphocyte activation molecule (SLAM) family of receptors (SFRs) are ubiquitously expressed on immune cells, and they regulate multiple immune events by recruiting SH2 (Src homology 2) domain-containing SAP family adapters, including SAP and its homologs, Ewing's sarcoma-associated transcript 2 (EAT-2) and EAT-2 related transducer (ERT). In human patients with X-linked lymphoproliferative (XLP) disease, which is caused by SAP mutations, SFRs alternatively bind other inhibitory SH2 domain-containing molecules to suppress immune cell activation and development. NK cells express multiple SFRs and all SAP family adapters. In recent decades, SFRs have been found to be critical for enhancing NK cell activation in response to abnormal hematopoietic cells in SAP-family-intact NK cells; however, SFRs might suppress NK cell activation in SAP-family-deficient mice or patients with XLP1. In this paper, we review how these two distinct SFR signaling pathways orchestrate NK cell activation and inhibition and highlight the importance of SFR regulation of NK cell biology and their physiological status and pathological relevance in patients with XLP1.
Collapse
|
10
|
Mahr B, Pilat N, Granofszky N, Wiletel M, Muckenhuber M, Maschke S, Hock K, Wekerle T. Hybrid resistance to parental bone marrow grafts in nonlethally irradiated mice. Am J Transplant 2019; 19:591-596. [PMID: 30346652 PMCID: PMC6492153 DOI: 10.1111/ajt.15146] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 01/25/2023]
Abstract
Resistance to parental bone marrow (BM) grafts in F1 hybrid recipients is due to natural killer (NK) cell-mediated rejection triggered through "missing self" recognition. "Hybrid resistance" has usually been investigated in lethally irradiated F1 recipients in conjunction with pharmacological activation of NK cells. Here, we investigated BM-directed NK-cell alloreactivity in settings of reduced conditioning. Nonlethally irradiated (1-3 Gy) or nonirradiated F1 (C57BL6 × BALB/c) recipient mice received titrated doses (5-20 x 106 ) of unseparated parental BALB/c BM without pharmacological NK cell activation. BM successfully engrafted in all mice and multilineage donor chimerism persisted long-term (24 weeks), even in the absence of irradiation. Chimerism was associated with the rearrangement of the NK-cell receptor repertoire suggestive of reduced reactivity to BALB/c. Chimerism levels were lower after transplantation with parental BALB/c than with syngeneic F1 BM, indicating partial NK-mediated rejection of parental BM. Activation of NK cells with polyinosinic-polycytidylic acid sodium salt poly(I:C), reduced parental chimerism in nonirradiated BM recipients but did not prevent hematopoietic stem cell engraftment. In contrast, equal numbers of parental lymph node cells were completely rejected. Hence, hybrid resistance leads to incomplete rejection of parental BM under reduced conditioning settings.
Collapse
Affiliation(s)
- Benedikt Mahr
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Nina Pilat
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Nicolas Granofszky
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Mario Wiletel
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Moritz Muckenhuber
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Svenja Maschke
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Karin Hock
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Thomas Wekerle
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| |
Collapse
|
11
|
Mahr B, Pilat N, Maschke S, Granofszky N, Schwarz C, Unger L, Hock K, Farkas AM, Klaus C, Regele H, Wekerle T. Regulatory T Cells Promote Natural Killer Cell Education in Mixed Chimeras. Am J Transplant 2017; 17:3049-3059. [PMID: 28489338 DOI: 10.1111/ajt.14342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 01/25/2023]
Abstract
Therapeutic administration of regulatory T cells (Tregs) leads to engraftment of conventional doses of allogeneic bone marrow (BM) in nonirradiated recipient mice conditioned with costimulation blockade and mammalian target of rapamycin inhibition. The mode of action responsible for this Treg effect is poorly understood but may encompass the control of costimulation blockade-resistant natural killer (NK) cells. We show that transient NK cell depletion at the time of BM transplantation led to BM engraftment and persistent chimerism without Treg transfer but failed to induce skin graft tolerance. In contrast, the permanent absence of anti-donor NK reactivity in mice grafted with F1 BM was associated with both chimerism and tolerance comparable to Treg therapy, implying that NK cell tolerization is a critical mechanism of Treg therapy. Indeed, NK cells of Treg-treated BM recipients reshaped their receptor repertoire in the presence of donor MHC in a manner suggesting attenuated donor reactivity. These results indicate that adoptively transferred Tregs prevent BM rejection, at least in part, by suppressing NK cells and promote tolerance by regulating the appearance of NK cells expressing activating receptors to donor class I MHC.
Collapse
Affiliation(s)
- B Mahr
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - N Pilat
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - S Maschke
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - N Granofszky
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - C Schwarz
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - L Unger
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - K Hock
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - A M Farkas
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - C Klaus
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - H Regele
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - T Wekerle
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
da Silva MB, da Cunha FF, Terra FF, Camara NOS. Old game, new players: Linking classical theories to new trends in transplant immunology. World J Transplant 2017; 7:1-25. [PMID: 28280691 PMCID: PMC5324024 DOI: 10.5500/wjt.v7.i1.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
The evolutionary emergence of an efficient immune system has a fundamental role in our survival against pathogenic attacks. Nevertheless, this same protective mechanism may also establish a negative consequence in the setting of disorders such as autoimmunity and transplant rejection. In light of the latter, although research has long uncovered main concepts of allogeneic recognition, immune rejection is still the main obstacle to long-term graft survival. Therefore, in order to define effective therapies that prolong graft viability, it is essential that we understand the underlying mediators and mechanisms that participate in transplant rejection. This multifaceted process is characterized by diverse cellular and humoral participants with innate and adaptive functions that can determine the type of rejection or promote graft acceptance. Although a number of mediators of graft recognition have been described in traditional immunology, recent studies indicate that defining rigid roles for certain immune cells and factors may be more complicated than originally conceived. Current research has also targeted specific cells and drugs that regulate immune activation and induce tolerance. This review will give a broad view of the most recent understanding of the allogeneic inflammatory/tolerogenic response and current insights into cellular and drug therapies that modulate immune activation that may prove to be useful in the induction of tolerance in the clinical setting.
Collapse
|
13
|
Minor Antigen Disparities Impede Induction of Long Lasting Chimerism and Tolerance through Bone Marrow Transplantation with Costimulation Blockade. J Immunol Res 2016; 2016:8635721. [PMID: 27872868 PMCID: PMC5107841 DOI: 10.1155/2016/8635721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/27/2016] [Accepted: 10/10/2016] [Indexed: 02/08/2023] Open
Abstract
Mixed chimerism and tolerance can be successfully induced in rodents through allogeneic bone marrow transplantation (BMT) with costimulation blockade (CB), but varying success rates have been reported with distinct models and protocols. We therefore investigated the impact of minor antigen disparities on the induction of mixed chimerism and tolerance. C57BL/6 (H2b) mice received nonmyeloablative total body irradiation (3 Gy), costimulation blockade (anti-CD40L mAb and CTLA4Ig), and 2 × 107 bone marrow cells (BMC) from either of three donor strains: Balb/c (H2d) (MHC plus multiple minor histocompatibility antigen (mHAg) mismatched), B10.D2 (H2d) or B10.A (H2a) (both MHC mismatched, but mHAg matched). Macrochimerism was followed over time by flow cytometry and tolerance was tested by skin grafting. 20 of 21 recipients of B10.D2 BMC but only 13 of 18 of Balb/c BMC and 13 of 20 of B10.A BMC developed stable long-term multilineage chimerism (p < 0.05 for each donor strain versus B10.D2). Significantly superior donor skin graft survival was observed in successfully established long-term chimeras after mHAg matched BMT compared to mHAg mismatched BMT (p < 0.05). Both minor and major antigen disparities pose a substantial barrier for the induction of chimerism while the maintenance of tolerance after nonmyeloablative BMT and costimulation blockade is negatively influenced by minor antigen disparities.
Collapse
|
14
|
Kadri N, Wagner AK, Ganesan S, Kärre K, Wickström S, Johansson MH, Höglund P. Dynamic Regulation of NK Cell Responsiveness. Curr Top Microbiol Immunol 2016; 395:95-114. [PMID: 26658943 DOI: 10.1007/82_2015_485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Natural killer (NK) cells deliver cytotoxic granules and immunomodulatory cytokines in response to transformed and virally infected cells. NK cell functions are under the control of a large number of germline-encoded receptors that recognize various ligands on target cells, but NK cells also respond to cytokines in the surrounding environment. The interaction between NK cell receptors and their ligands delivers either inhibitory or activating signals. The cytokine milieu further shapes NK cell responses, either directly or by influencing the way inhibitory or activating signals are perceived by NK cells. In this review, we discuss how NK cell function is controlled by inhibitory receptors and MHC-I molecules, how activating receptors contribute to NK cell education, and finally, how cytokines secreted by the surrounding cells affect NK cell responsiveness. Inputs at these three levels involve different cell types and are seamlessly integrated to form a functional NK cell population.
Collapse
Affiliation(s)
- Nadir Kadri
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Arnika Kathleen Wagner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Sridharan Ganesan
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 141 86, Stockholm, Sweden
| | - Klas Kärre
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Stina Wickström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Maria H Johansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Petter Höglund
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, 141 86, Stockholm, Sweden.
| |
Collapse
|
15
|
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: 0.2] [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
|
16
|
Yu X, Lu L, Liu Z, Yang T, Gong X, Ning Y, Jiang Y. Brain-derived neurotrophic factor modulates immune reaction in mice with peripheral nerve xenotransplantation. Neuropsychiatr Dis Treat 2016; 12:685-94. [PMID: 27099498 PMCID: PMC4820192 DOI: 10.2147/ndt.s98387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has been demonstrated to play an important role in survival, differentiation, and neurite outgrowth for many types of neurons. This study was designed to identify the role of BDNF during peripheral nerve xenotransplantation. MATERIALS AND METHODS A peripheral nerve xenotransplantation from rats to mice was performed. Intracellular cytokines were stained for natural killer (NK) cells, natural killer T (NKT) cells, T cells, and B cells and analyzed by flow cytometry in the spleen of the recipient mouse. Serum levels of related cytokines were quantified by cytometric bead array. RESULTS Splenic NK cells significantly increased in the xenotransplanted mice (8.47±0.88×10(7) cells/mL) compared to that in the control mice (4.66±0.78×10(7) cells/mL, P=0.0003), which significantly reduced in the presence of BDNF (4.85±0.87×10(7) cells/mL, P=0.0004). In contrast, splenic NKT cell number was significantly increased in the mice with xenotransplantation plus BDNF (XT + BDNF) compared to that of control group or of mice receiving xenotransplantation only (XT only). Furthermore, the number of CD3+ T cells, CD3+CD4+ T cells, CD3+CD4- T cells, interferon-γ-producing CD3+CD4+ T cells, and interleukin (IL)-17-producing CD3+CD4+ T cells, as well as CD3-CD19+ B cells, was significantly higher in the spleen of XT only mice compared to the control mice (P<0.05), which was significantly reduced by BDNF (P<0.05). The number of IL-4-producing CD3+CD4+ T cells and CD3+CD4+CD25+Foxp3+ T cells was significantly higher in the spleen of XT + BDNF mice than that in the spleen of XT only mice (P<0.05). Serum levels of IL-6, TNF-α, interferon-γ, and IL-17 were decreased, while IL-4 and IL-10 were stimulated by BDNF following xenotransplantation. CONCLUSION BDNF reduced NK cells but increased NKT cell accumulation in the spleen of xenotransplanted mice. BDNF modulated the number of splenic T cells and its subtype cells in the mice following xenotransplantation. These findings suggest that BDNF inhibits rejection of peripheral nerve following xenotransplantation by regulating innate as well as adaptive immune reaction.
Collapse
Affiliation(s)
- Xin Yu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Laijin Lu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhigang Liu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Teng Yang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xu Gong
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yubo Ning
- Department of Orthopedics, Ningshi Orthopedics Hospital of Tonghua, Tonghua, People's Republic of China
| | - Yanfang Jiang
- Department of Central Laboratory, The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
17
|
Alhajjat AM, Strong BS, Lee AE, Turner LE, Wadhwani RK, Ortaldo JR, Heusel JW, Shaaban AF. Prenatal Allospecific NK Cell Tolerance Hinges on Instructive Allorecognition through the Activating Receptor during Development. THE JOURNAL OF IMMUNOLOGY 2015; 195:1506-16. [PMID: 26136432 DOI: 10.4049/jimmunol.1500463] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/11/2015] [Indexed: 12/24/2022]
Abstract
Little is known about how the prenatal interaction between NK cells and alloantigens shapes the developing NK cell repertoire toward tolerance or immunity. Specifically, the effect on NK cell education arising from developmental corecognition of alloantigens by activating and inhibitory receptors with shared specificity is uncharacterized. Using a murine prenatal transplantation model, we examined the manner in which this seemingly conflicting input affects NK cell licensing and repertoire formation in mixed hematopoietic chimeras. We found that prenatal NK cell tolerance arose from the elimination of phenotypically hostile NK cells that express an allospecific activating receptor without coexpressing any allospecific inhibitory receptors. Importantly, the checkpoint for the system appeared to occur centrally within the bone marrow during the final stage of NK cell maturation and hinged on the instructive recognition of allogeneic ligand by the activating receptor rather than through the inhibitory receptor as classically proposed. Residual nondeleted hostile NK cells expressing only the activating receptor exhibited an immature, anergic phenotype, but retained the capacity to upregulate inhibitory receptor expression in peripheral sites. However, the potential for this adaptive change to occur was lost in developmentally mature chimeras. Collectively, these findings illuminate the intrinsic process in which developmental allorecognition through the activating receptor regulates the emergence of durable NK cell tolerance and establishes a new paradigm to fundamentally guide future investigations of prenatal NK cell-allospecific education.
Collapse
Affiliation(s)
- Amir M Alhajjat
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Beverly S Strong
- Department of Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Amanda E Lee
- Department of Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Lucas E Turner
- Department of Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Ram K Wadhwani
- Department of Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - John R Ortaldo
- Experimental Therapeutics Section, National Cancer Institute, Frederick, MD 21702
| | - Jonathan W Heusel
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110; and Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Aimen F Shaaban
- Department of Surgery, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45229;
| |
Collapse
|
18
|
Use of Alefacept for Preconditioning in Multiply Transfused Pediatric Patients with Nonmalignant Diseases. Biol Blood Marrow Transplant 2015; 21:1845-52. [PMID: 26095669 DOI: 10.1016/j.bbmt.2015.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022]
Abstract
Transfusion-related alloimmunization is a potent barrier to the engraftment of allogeneic hematopoietic stem cells in patients with nonmalignant diseases (NMDs). Memory T cells, which drive alloimmunization, are relatively resistant to commonly used conditioning agents. Alefacept, a recombinant leukocyte function antigen-3/IgG1 fusion protein, targets CD2 and selectively depletes memory versus naive T cells. Three multiply transfused pediatric patients with NMD received a short course of high-dose i.v. alefacept (.25 mg/kg/dose on days -40 and -9 and .5 mg/kg/dose on days -33, -26, -19, and -12) before undergoing unrelated allogeneic transplant in the setting of reduced-intensity pretransplant conditioning and calcineurin inhibitor-based post-transplant graft-versus-host disease (GVHD) prophylaxis. Alefacept infusions were well tolerated in all patients. Peripheral blood flow cytometry was performed at baseline and during and after alefacept treatment. As expected, after the 5 weekly alefacept doses, each patient demonstrated selective loss of CD2(hi)/CCR7(-)/CD45RA(-) effector memory (Tem) and CD2(hi)/CCR7(+)/CD45RA(-) central memory (Tcm) CD4(+) and CD8(+) T cells with relative preservation of the CD2(lo) Tem and Tcm subpopulations. In addition, depletion of CD2(+) natural killer (NK) cells also occurred. Neutrophil recovery was rapid, and all 3 patients had 100% sorted (CD3/CD33) peripheral blood donor chimerism by day +100. Immune reconstitution (by absolute neutrophil, monocyte, and lymphocyte counts) was comparable with a cohort of historical control patients. All 3 patients developed GVHD but are all now off immune suppression and >2 years post-transplant with stable full-donor engraftment. These results suggest that alefacept at higher dosing can deplete both memory T cells and NK cells and that incorporating CD2-targeted depletion into a reduced-intensity transplant regimen is feasible and safe in heavily transfused patients.
Collapse
|
19
|
Nalle SC, Kwak HA, Edelblum KL, Joseph NE, Singh G, Khramtsova GF, Mortenson ED, Savage PA, Turner JR. Recipient NK cell inactivation and intestinal barrier loss are required for MHC-matched graft-versus-host disease. Sci Transl Med 2015; 6:243ra87. [PMID: 24990882 DOI: 10.1126/scitranslmed.3008941] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have shown a correlation between pretransplant conditioning intensity, intestinal barrier loss, and graft-versus-host disease (GVHD) severity. However, because irradiation and other forms of pretransplant conditioning have pleiotropic effects, the precise role of intestinal barrier loss in GVHD pathogenesis remains unclear. We developed GVHD models that allowed us to isolate the specific contributions of distinct pretransplant variables. Intestinal damage was required for the induction of minor mismatch [major histocompatibility complex (MHC)-matched] GVHD, but was not necessary for major mismatch GVHD, demonstrating fundamental pathogenic distinctions between these forms of disease. Moreover, recipient natural killer (NK) cells prevented minor mismatch GVHD by limiting expansion and target organ infiltration of alloreactive T cells via a perforin-dependent mechanism, revealing an immunoregulatory function of MHC-matched recipient NK cells in GVHD. Minor mismatch GVHD required MyD88-mediated Toll-like receptor 4 (TLR4) signaling on donor cells, and intestinal damage could be bypassed by parenteral lipopolysaccharide (LPS) administration, indicating a critical role for the influx of bacterial components triggered by intestinal barrier loss. In all, the data demonstrate that pretransplant conditioning plays a dual role in promoting minor mismatch GVHD by both depleting recipient NK cells and inducing intestinal barrier loss.
Collapse
Affiliation(s)
- Sam C Nalle
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - H Aimee Kwak
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Karen L Edelblum
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Nora E Joseph
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | | | - Eric D Mortenson
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Peter A Savage
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| | - Jerrold R Turner
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
20
|
Dvorak CC, Horn BN, Puck JM, Adams S, Veys P, Czechowicz A, Cowan MJ. A trial of alemtuzumab adjunctive therapy in allogeneic hematopoietic cell transplantation with minimal conditioning for severe combined immunodeficiency. Pediatr Transplant 2014; 18:609-16. [PMID: 24977928 PMCID: PMC4134761 DOI: 10.1111/petr.12310] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2014] [Indexed: 12/31/2022]
Abstract
For infants with SCID the ideal conditioning regimen before allogeneic HCT would omit cytotoxic chemotherapy to minimize short- and long-term complications. We performed a prospective pilot trial with alemtuzumab monotherapy to overcome NK-cell mediated immunologic barriers to engraftment. We enrolled four patients who received CD34-selected haploidentical cells, two of whom failed to engraft donor T cells. The two patients who engrafted had delayed T-cell reconstitution, despite rapid clearance of circulating alemtuzumab. Although well-tolerated, alemtuzumab failed to overcome immunologic barriers to donor engraftment. Furthermore, alemtuzumab may slow T-cell development in patients with SCID in the setting of a T-cell depleted graft.
Collapse
Affiliation(s)
- Christopher C. Dvorak
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplant, University of California San Francisco Benioff Children's Hospital
| | - Biljana N. Horn
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplant, University of California San Francisco Benioff Children's Hospital
| | - Jennifer M. Puck
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplant, University of California San Francisco Benioff Children's Hospital
| | - Stuart Adams
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, London, UK
| | - Paul Veys
- Centre for Immunodeficiency, Molecular Immunology Unit, UCL Institute of Child Health, London, UK
| | - Agnieszka Czechowicz
- Division of Blood & Marrow Transplantation, Stanford University School of Medicine
| | - Morton J. Cowan
- Division of Pediatric Allergy, Immunology, and Blood and Marrow Transplant, University of California San Francisco Benioff Children's Hospital
| |
Collapse
|
21
|
Nakata J, Nakano K, Okumura A, Mizutani Y, Kinoshita H, Iwai M, Hasegawa K, Morimoto S, Fujiki F, Tatsumi N, Nakajima H, Nakae Y, Nishida S, Tsuboi A, Oji Y, Oka Y, Sugiyama H, Kumanogoh A, Hosen N. In vivo eradication of MLL/ENL leukemia cells by NK cells in the absence of adaptive immunity. Leukemia 2013; 28:1316-25. [DOI: 10.1038/leu.2013.374] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 10/26/2013] [Accepted: 12/10/2013] [Indexed: 12/18/2022]
|
22
|
In vivo T cell costimulation blockade with abatacept for acute graft-versus-host disease prevention: a first-in-disease trial. Biol Blood Marrow Transplant 2013; 19:1638-49. [PMID: 24047754 DOI: 10.1016/j.bbmt.2013.09.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 09/04/2013] [Indexed: 01/20/2023]
Abstract
We performed a first-in-disease trial of in vivo CD28:CD80/86 costimulation blockade with abatacept for acute graft-versus-host disease (aGVHD) prevention during unrelated-donor hematopoietic cell transplantation (HCT). All patients received cyclosporine/methotrexate plus 4 doses of abatacept (10 mg/kg/dose) on days -1, +5, +14, +28 post-HCT. The feasibility of adding abatacept, its pharmacokinetics, pharmacodynamics, and its impact on aGVHD, infection, relapse, and transplantation-related mortality (TRM) were assessed. All patients received the planned abatacept doses, and no infusion reactions were noted. Compared with a cohort of patients not receiving abatacept (the StdRx cohort), patients enrolled in the study (the ABA cohort) demonstrated significant inhibition of early CD4(+) T cell proliferation and activation, affecting predominantly the effector memory (Tem) subpopulation, with 7- and 10-fold fewer proliferating and activated CD4(+) Tem cells, respectively, at day+28 in the ABA cohort compared with the StdRx cohort (P < .01). The ABA patients demonstrated a low rate of aGVHD, despite robust immune reconstitution, with 2 of 10 patients diagnosed with grade II-IV aGVHD before day +100, no deaths from infection, no day +100 TRM, and with 7 of 10 evaluable patients surviving (median follow-up, 16 months). These results suggest that costimulation blockade with abatacept can significantly affect CD4(+) T cell proliferation and activation post-transplantation, and may be an important adjunct to standard immunoprophylaxis for aGVHD in patients undergoing unrelated-donor HCT.
Collapse
|
23
|
Smelt MJ, Faas MM, de Haan BJ, de Haan A, Vaage JT, de Vos P. The role of alloresponsive Ly49+ NK cells in rat islet allograft failure in the presence and absence of cytomegalovirus. Cell Transplant 2013; 23:1381-94. [PMID: 23866824 DOI: 10.3727/096368913x670930] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There are still many factors to discover to explain the low success rates of islet allografts. In this study, we demonstrate that specific subpopulations of alloreactive NK cells may be involved in the failure of islet allografts. By performing allotransplantation in rats (n = 13), we observed peripheral expansion and infiltration of alloreactive Ly49i2(+) NK cells in the grafts. An effective strategy in rats to enhance the expansion of Ly49i2(+) NK cells is performing a rat cytomegalovirus infection (n = 6). Cytomegalovirus infection was associated with an early expansion of the Ly49i2(+) NK cells and accelerated islet graft failure. The Ly49i2(+) NK cells are both alloreactive and involved in virus clearance. The expansion of this subpopulation could not be blocked by cyclosporin A immunosuppression. Also alloreactive KLRH1(+) NK cells infiltrated the grafts, but nonalloreactive NKR-P1B(+) cells were not observed in the islet allografts. Perforin staining of the infiltrating NK cells demonstrated the cytotoxic capacity of these cells. Our data suggest a role for this NK subpopulation in rat islet allograft destruction.
Collapse
Affiliation(s)
- Maaike J Smelt
- Department of Pathology and Medical Biology, Division of Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | |
Collapse
|
24
|
Xu H, Yan J, Zhu Z, Hussain LR, Huang Y, Ding C, Bozulic LD, Wen Y, Ildstad ST. A critical role for the TLR4/TRIF pathway in allogeneic hematopoietic cell rejection by innate immune cells. Cell Transplant 2012; 22:2367-80. [PMID: 23146386 DOI: 10.3727/096368912x658881] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We show for the first time that signaling through the TLR4/TRIF pathway plays a critical role in allogeneic bone marrow cell (BMC) rejection. This appears to be unique to BMCs as organ allografts are rejected mainly via MyD88 signaling. Using T- or T-/B-cell-deficient mice, we found that BMC allorejection occurred early before T-cell activation and was T- and B-cell independent, suggesting an effector role for innate immune cells in BMC rejection. We further demonstrated the innate immune signaling in BMC allorejection by showing superior engraftment in mice deficient in TRIF or TLR4 but not in MyD88 or TLR3. The restored cytotoxicity in TRIF-deficient recipients transferred with wild-type F4/80(+) or NK1.1(+) cells suggests TRIF signaling dependence on macrophages or NK cells in early BMC rejection. Production of the proinflammatory cytokine IL-6 and TRIF relevant chemokine MCP-1 was significantly increased early after bone marrow transplantation. In vivo specific depletion of macrophages or NK innate immune cells in combination with anti-CD154/rapamycin resulted in additive-enhanced allogeneic engraftment. The requirement for irradiation was completely eliminated when both macrophages and NK cells were depleted in combination with anti-CD154/rapamycin to target T- and B-cells, supporting the hypothesis that two barriers involving innate and adaptive immunity exist in mediating the rejection of allogeneic BMCs. In summary, our results clearly demonstrate a previously unappreciated role for innate immunity in BMC allorejection via signaling through a unique MyD88-independent TLR4/TRIF mechanism. These findings may have direct clinical impact on strategies for conditioning recipients for stem cell transplantation.
Collapse
Affiliation(s)
- Hong Xu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Page A, Srinivasan S, Singh K, Russell M, Hamby K, Deane T, Sen S, Stempora L, Leopardi F, Price AA, Strobert E, Reimann KA, Kirk AD, Larsen CP, Kean LS. CD40 blockade combines with CTLA4Ig and sirolimus to produce mixed chimerism in an MHC-defined rhesus macaque transplant model. Am J Transplant 2012; 12:115-25. [PMID: 21929643 PMCID: PMC3259212 DOI: 10.1111/j.1600-6143.2011.03737.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In murine models, T-cell costimulation blockade of the CD28:B7 and CD154:CD40 pathways synergistically promotes immune tolerance after transplantation. While CD28 blockade has been successfully translated to the clinic, translation of blockade of the CD154:CD40 pathway has been less successful, in large part due to thromboembolic complications associated with anti-CD154 antibodies. Translation of CD40 blockade has also been slow, in part due to the fact that synergy between CD40 blockade and CD28 blockade had not yet been demonstrated in either primate models or humans. Here we show that a novel, nondepleting CD40 monoclonal antibody, 3A8, can combine with combined CTLA4Ig and sirolimus in a well-established primate bone marrow chimerism-induction model. Prolonged engraftment required the presence of all three agents during maintenance therapy, and resulted in graft acceptance for the duration of immunosuppressive treatment, with rejection resulting upon immunosuppression withdrawal. Flow cytometric analysis revealed that upregulation of CD95 expression on both CD4+ and CD8+ T cells correlated with rejection, suggesting that CD95 may be a robust biomarker of graft loss. These results are the first to demonstrate prolonged chimerism in primates treated with CD28/mTOR blockade and nondepletional CD40 blockade, and support further investigation of combined costimulation blockade targeting the CD28 and CD40 pathways.
Collapse
Affiliation(s)
- Andrew Page
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Swetha Srinivasan
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Karnail Singh
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Maria Russell
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Kelly Hamby
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Taylor Deane
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Sharon Sen
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Linda Stempora
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Frank Leopardi
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | | | - Elizabeth Strobert
- The Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322
| | | | - Allan D. Kirk
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Christian P. Larsen
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| | - Leslie S. Kean
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
- Aflac Cancer Center and Blood Disorders Service, Department of Pediatrics and The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
26
|
Nonobese diabetic natural killer cells: a barrier to allogeneic chimerism that can be reduced by rapamycin. Transplantation 2011; 92:977-84. [PMID: 21956197 DOI: 10.1097/tp.0b013e3182313e70] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Induction of allogeneic hematopoietic chimerism is a promising strategy to induce tolerance to donor islets for treating type 1 diabetes. Successful induction of chimerism requires overcoming host alloimmunity. In diabetes-prone nonobese diabetic (NOD) mice, this is challenging due to their general tolerance resistance. Although the adaptive alloimmunity of NOD mice is a known barrier to allogeneic chimerism, whether NOD natural killer (NK) cells are an additional barrier has not been examined. Because NOD NK cells exhibit functional defects, they may not inhibit chimerism generation. METHODS Antibody depletion of NK cells in vivo, or transplantation of F1 hybrid donor cells to eliminate the "missing-self" trigger of NK cells, was preformed to test the NK-mediated rejection of donor bone marrow cells. We also studied the capacity of rapamycin to block the NK cell response against allogeneic cells in vivo. RESULTS Depleting NK cells or rendering them unresponsive to the donor greatly improved the level of chimerism obtained in NOD mice. Rapamycin significantly reduced the resistance to allogeneic chimerism mounted by NOD NK cells; however, it was much less effective than NK cell depletion by antibodies. CONCLUSIONS Contrary to the view that NOD NK cells are defective, we found these cells to be a substantial barrier to allogeneic chimerism in the presence or absence of adaptive immunity. Moreover, rapamycin will need to be combined with other approaches to fully overcome the NK cell barrier.
Collapse
|
27
|
Yu GP, Nadeau KC, Berk DR, de Saint Basile G, Lambert N, Knapnougel P, Roberts J, Kavanau K, Dunn E, Stiehm ER, Lewis DB, Umetsu DT, Puck JM, Cowan MJ. Genotype, phenotype, and outcomes of nine patients with T-B+NK+ SCID. Pediatr Transplant 2011; 15:733-41. [PMID: 21883749 PMCID: PMC3196791 DOI: 10.1111/j.1399-3046.2011.01563.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There are few reports of clinical presentation, genotype, and HCT outcomes for patients with T-B+NK+ SCID. Between 1981 and 2007, eight of 84 patients with SCID who received and/or were followed after HCT at UCSF had the T-B+NK+ phenotype. One additional patient with T-B+NK+ SCID was identified as the sibling of a patient treated at UCSF. Chart reviews were performed. Molecular analyses of IL7R, IL2RG, JAK3, and the genes encoding the CD3 T-cell receptor components δ (CD3D), ε (CD3E), and ζ (CD3Z) were carried out. IL7R mutations were documented in four patients and CD3D mutations in two others. Three patients had no defects found. Only two of nine patients had an HLA-matched related HCT donor. Both survived, and neither developed GVHD. Five of seven recipients of haploidentical grafts survived. Although the majority of reported cases of T-B+NK+ SCID are caused by defects in IL7R, CD3 complex defects were also found in this series and should be considered when evaluating patients with T-B+NK+ SCID. Additional genes, mutations in which account for T-B+NK+ SCID, remain to be found. Better approaches to early diagnosis and HCT treatment are needed for patients lacking an HLA-matched related donor.
Collapse
Affiliation(s)
- Grace P Yu
- Division of Immunology and Allergy, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital at Stanford
| | - Kari C Nadeau
- Division of Immunology and Allergy, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital at Stanford
| | - David R Berk
- Departments of Medicine and Pediatrics, Divisions of Dermatology, Washington University School of Medicine
| | - Geneviève de Saint Basile
- Inserm, U768, Paris, F-75015 France,Université Paris Descartes, IRNEM (IFR95), Paris, F-75015 France,AP-HP, Hôpital Necker Enfants-Malades, Unité d'Immunologie-Hématologie Pédiatrique, Paris, F-75015 France
| | - Nathalie Lambert
- AP-HP, Hôpital Necker Enfants-Malades, Unité d'Immunologie-Hématologie Pédiatrique, Paris, F-75015 France
| | | | - Joseph Roberts
- Department of Pediatrics and Immunology, Duke University Medical Center
| | - Kristina Kavanau
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco Children's Hospital
| | - Elizabeth Dunn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco Children's Hospital
| | - E. Richard Stiehm
- Divison of Immunology, Allergy and Rheumatology, Department of Pediatrics, Mattel Children's Hospital at the University of California Los Angeles
| | - David B Lewis
- Division of Immunology and Allergy, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital at Stanford
| | - Dale T Umetsu
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital Boston
| | - Jennifer M Puck
- Department of Pediatrics, Institute for Human Genetics, University of California San Francisco Children's Hospital
| | - Morton J Cowan
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of California San Francisco Children's Hospital
| |
Collapse
|
28
|
Espinoza JL, Takami A, Onizuka M, Sao H, Akiyama H, Miyamura K, Okamoto S, Inoue M, Kanda Y, Ohtake S, Fukuda T, Morishima Y, Kodera Y, Nakao S. NKG2D gene polymorphism has a significant impact on transplant outcomes after HLA-fully-matched unrelated bone marrow transplantation for standard risk hematologic malignancies. Haematologica 2011; 94:1427-34. [PMID: 19794085 DOI: 10.3324/haematol.2009.008318] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND NKG2D, an activating and co-stimulatory receptor expressed on natural killer cells and T cells, plays pivotal roles in immunity to microbial infections as well as in cancer immunosurveillance. This study examined the impact of donor and recipient polymorphisms in the NKG2D gene on the clinical outcomes of patients undergoing allogeneic T-cell-replete myeloablative bone marrow transplantation using an HLA-matched unrelated donor. DESIGN AND METHODS The NKG2D polymorphism was retrospectively analyzed in a total 145 recipients with hematologic malignancies and their unrelated donors. The patients underwent transplantation following myeloablative conditioning; the recipients and donors were matched through the Japan Marrow Donor Program. RESULTS In patients with standard-risk disease, the donor NKG2D-HNK1 haplotype, a haplotype expected to induce greater natural killer cell activity, was associated with significantly improved overall survival (adjusted hazard ratio, 0.44; 95% confidence interval, 0.23 to 0.85; p=0.01) as well as transplant related mortality (adjusted hazard ratio, 0.42; 95% confidence interval, 0.21 to 0.86; p=0.02), but had no impact on disease relapse or the development of grade II-IV acute graft-versus-host disease or chronic graft-versus-host disease. The NKG2D polymorphism did not significantly influence the transplant outcomes in patients with high-risk disease. CONCLUSIONS These data suggest an association between the donor HNK1 haplotype and better clinical outcome among recipients, with standard-risk disease, of bone marrow transplants from HLA-matched unrelated donors.
Collapse
Affiliation(s)
- J Luis Espinoza
- Department of Hematology & Oncology, Kanazawa University Hospital, 13-1 Takaramachi, Kanazawa, 920-8641, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Elimination of the chemotherapy resistant subpopulation of 4T1 mouse breast cancer by haploidentical NK cells cures the vast majority of mice. Breast Cancer Res Treat 2011; 130:773-81. [PMID: 21274621 DOI: 10.1007/s10549-011-1355-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 01/13/2011] [Indexed: 12/16/2022]
Abstract
Metastatic breast cancer is currently incurable despite initial responsiveness, assumingly due to the presence of chemoresistant subpopulations that can be characterized as label retaining cells (LRC). In the 4T1 mouse breast cancer model, we previously achieved cure after Cyclophosphamide and Total Body Irradiation (CY + TBI) followed by haploidentical bone marrow and spleen transplantation (BMSPLT). CY + TBI without transplantation induced only transient impaired tumor growth indicating a critical role of donor immune cells. As it remained unknown if the 4T1 model resembles human disease with respect to the presence of subpopulations of chemoresistant LRC, we now demonstrate this is indeed the case. Chemoresistance of 4T1 LRC was demonstrated by in vitro co-incubation of fluorescently labeled 4T1 cells in limiting dilution with cyclophosphamide, doxorubicin or cisplatinum, after which only LRC containing colonies remained. LRC also remain in vivo after treatment with CY + TBI. Succeeding experiments set up to identify the haploidentical effector cell responsible for cure and, therefore, for the elimination of chemoresistant LRC designate donor NK cells crucial for the anti-tumor effect. NK cell depletion of the haploidentical graft fully abrogated the anti-tumor effect. Increased disease-free survival retained after transplantation of haploidentical bone marrow and NK cell-enriched spleen cell grafts, even in the absence of donor T-cells or of donor bone marrow. Tumor growth analysis indicates the anti-tumor effect being immediate (days). Based on these data, it is worthwhile to explore alloreactive adoptive NK cell therapy as consolidation for patients with metastasized breast cancer.
Collapse
|
30
|
Gill RG. NK cells: elusive participants in transplantation immunity and tolerance. Curr Opin Immunol 2010; 22:649-54. [PMID: 20952173 DOI: 10.1016/j.coi.2010.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 09/17/2010] [Accepted: 09/17/2010] [Indexed: 10/18/2022]
Abstract
NK cells constitute an innate MHC class I-reactive lymphoid population that rapidly responds to infection, injury, or cell distress. In the transplant field, NK cells have most often been associated with pro-inflammatory immunity resulting in the exacerbation of allograft injury. Despite this general view of NK cell reactivity, it has been challenging to assign unambiguous obligate roles for NK cells in the allograft response. While recent reports continue to provide evidence supporting a role for NK cells in promoting both acute and chronic rejection, there are also a growing number of studies that illustrate an alternative role for NK cells in promoting allograft survival and tolerance. This review addresses the plasticity of NK responses in transplantation by suggesting specific 'checkpoints' whereby NK cells can either enhance or inhibit the allograft response in vivo.
Collapse
|
31
|
The requirement for NKG2D in NK cell-mediated rejection of parental bone marrow grafts is determined by MHC class I expressed by the graft recipient. Blood 2010; 116:5208-16. [PMID: 20736452 DOI: 10.1182/blood-2010-05-285031] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Natural killer (NK) cells provide a unique barrier to semiallogeneic bone marrow (BM) transplantation. In the setting where the parents donate to the F1 offspring, rejection of parental bone marrow occurs. This "hybrid resistance" is completely NK cell dependent, as T cells in the F1 recipient tolerate parental grafts. Previously, we demonstrated that rejection of BALB/c parental BM by (BALB/c × C57BL/6) F1-recipient NK cells is dependent on the NKG2D-activating receptor, whereas rejection of parental C57BL/6 BM does not require NKG2D. BALB/c and B6 mice possess different NKG2D ligand genes and express these ligands differently on reconstituting BM cells. Herein, we show that the requirement for NKG2D in rejection depends on the major histocompatibility complex haplotype of donor cells and not the differences in the expression of NKG2D ligands. NKG2D stimulation of NK cell-mediated rejection was required to overcome inhibition induced by H-2D(d) when it engaged an inhibitory Ly49 receptor, whereas rejection of parental BM expressing the ligand, H-2K(b), did not require NKG2D. Thus, interactions between the inhibitory receptors on F1 NK cells and parental major histocompatibility complex class I ligands determine whether activation via NKG2D is required to achieve the threshold for rejection of parental BM grafts.
Collapse
|
32
|
Dvorak CC, Cowan MJ. Radiosensitive severe combined immunodeficiency disease. Immunol Allergy Clin North Am 2010; 30:125-42. [PMID: 20113890 DOI: 10.1016/j.iac.2009.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Inherited defects in components of the nonhomologous end-joining DNA repair mechanism produce a T-B-NK+ severe combined immunodeficiency disease (SCID) characterized by heightened sensitivity to ionizing radiation. Patients with the radiosensitive form of SCID may also have increased short- and long-term sensitivity to the alkylator-based chemotherapy regimens that are traditionally used for conditioning before allogeneic hematopoietic cell transplantation (HCT). Known causes of radiosensitive SCID include deficiencies of Artemis, DNA ligase IV, DNA-dependent protein kinase catalytic subunit, and Cernunnos-XLF, all of which have been treated with HCT. Because of these patients' sensitivity to certain forms of chemotherapy, the approach to donor selection and the type of conditioning regimen used for a patient with radiosensitive SCID requires careful consideration. Significantly more research needs to be done to determine the long-term outcomes of patients with radiosensitive SCID after HCT and to discover novel nontoxic approaches to HCT that might benefit those patients with intrinsic radiosensitivity and chemosensitivity as well as potentially all patients undergoing an HCT.
Collapse
Affiliation(s)
- Christopher C Dvorak
- Division of Pediatric Blood and Marrow Transplantation, University of California, San Francisco, 505 Parnassus Avenue, M-659, San Francisco, CA 94143-1278, USA
| | | |
Collapse
|
33
|
van der Touw W, Bromberg JS. Natural killer cells and the immune response in solid organ transplantation. Am J Transplant 2010; 10:1354-8. [PMID: 20353480 DOI: 10.1111/j.1600-6143.2010.03086.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Natural killer (NK) cells have been characterized classically for their cytotoxicity against pathogen infected or stressed cells as well as for their role in monitoring the expression of self MHC I. However, the participation of NK cells in solid organ transplantation (SOT) is poorly defined due to conflicting clinical and animal model data. Preclinical models have shown that NK cells exacerbate T-cell allogeneic responses during rejection, but can also promote tolerance induction under immunosuppressive conditions. Further, while protocols such as costimulatory blockade effectively induce tolerance by blocking T-cell activation and promoting Treg generation, how such regimens regulate other innate and adaptive immune cells, including NK cells, is incomplete. This review examines NK cells and the regulation of their effector functions in SOT.
Collapse
Affiliation(s)
- W van der Touw
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
34
|
Pilat N, Baranyi U, Klaus C, Jaeckel E, Mpofu N, Wrba F, Golshayan D, Muehlbacher F, Wekerle T. Treg-therapy allows mixed chimerism and transplantation tolerance without cytoreductive conditioning. Am J Transplant 2010; 10:751-762. [PMID: 20148810 PMCID: PMC2856406 DOI: 10.1111/j.1600-6143.2010.03018.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Establishment of mixed chimerism through transplantation of allogeneic donor bone marrow (BM) into sufficiently conditioned recipients is an effective experimental approach for the induction of transplantation tolerance. Clinical translation, however, is impeded by the lack of feasible protocols devoid of cytoreductive conditioning (i.e. irradiation and cytotoxic drugs/mAbs). The therapeutic application of regulatory T cells (Tregs) prolongs allograft survival in experimental models, but appears insufficient to induce robust tolerance on its own. We thus investigated whether mixed chimerism and tolerance could be realized without the need for cytoreductive treatment by combining Treg therapy with BM transplantation (BMT). Polyclonal recipient Tregs were cotransplanted with a moderate dose of fully mismatched allogeneic donor BM into recipients conditioned solely with short-course costimulation blockade and rapamycin. This combination treatment led to long-term multilineage chimerism and donor-specific skin graft tolerance. Chimeras also developed humoral and in vitro tolerance. Both deletional and nondeletional mechanisms contributed to maintenance of tolerance. All tested populations of polyclonal Tregs (FoxP3-transduced Tregs, natural Tregs and TGF-beta induced Tregs) were effective in this setting. Thus, Treg therapy achieves mixed chimerism and tolerance without cytoreductive recipient treatment, thereby eliminating a major toxic element impeding clinical translation of this approach.
Collapse
Affiliation(s)
- N Pilat
- Division of Transplantation, Department of Surgery, Medical University of ViennaAustria
| | - U Baranyi
- Division of Transplantation, Department of Surgery, Medical University of ViennaAustria
| | - C Klaus
- Division of Transplantation, Department of Surgery, Medical University of ViennaAustria
| | - E Jaeckel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH)Hannover, Germany
| | - N Mpofu
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH)Hannover, Germany
| | - F Wrba
- Institute of Clinical Pathology, Medical University of ViennaAustria
| | - D Golshayan
- Transplantation Centre and Transplantation Immunopathology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne UniversityLausanne, Switzerland
| | - F Muehlbacher
- Division of Transplantation, Department of Surgery, Medical University of ViennaAustria
| | - T Wekerle
- Division of Transplantation, Department of Surgery, Medical University of ViennaAustria,* Corresponding author: Thomas Wekerle,
| |
Collapse
|
35
|
Tabayoyong WB, Salas JG, Bonde S, Zavazava N. HOXB4-transduced embryonic stem cell-derived Lin-c-kit+ and Lin-Sca-1+ hematopoietic progenitors express H60 and are targeted by NK cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:5449-57. [PMID: 19828634 DOI: 10.4049/jimmunol.0901807] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Embryonic stem (ES) cells are a novel source of cells, especially hematopoietic progenitor cells that can be used to treat degenerative diseases in humans. However, there is a need to determine how ES cell-derived progenitors are regulated by both the adaptive and innate immune systems post transplantation. In this study, we demonstrate that hematopoietic progenitor cells (HPCs) derived from mouse ES cells ectopically expressing HOXB4 fail to engraft long-term in the presence of NK cells. In particular, the H60-expressing Lin(-)c-kit(+) and Lin(-)Sca-1(+) subpopulations were preferentially deleted in Rag2(-/-), but not in Rag2(-/-)gamma(c)(-/-) mice. Up-regulation of class I expression on HPCs prevented their lysis by NK cells, and Ab-mediated depletion of NK cells restored long-term HPC engraftment. In contrast to the notion that ES-derived cells are immune-privileged, we show in this study that NK cells form a formidable barrier to the long-term engraftment of ES cell-derived hematopoietic progenitors.
Collapse
Affiliation(s)
- William B Tabayoyong
- Medical Scientist Training Program and Immunology Graduate Program, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
36
|
Ascon M, Ascon DB, Liu M, Cheadle C, Sarkar C, Racusen L, Hassoun HT, Rabb H. Renal ischemia-reperfusion leads to long term infiltration of activated and effector-memory T lymphocytes. Kidney Int 2008; 75:526-35. [PMID: 19092796 DOI: 10.1038/ki.2008.602] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
It is well-established that significant ischemia-reperfusion injury during kidney transplantation results in increased incidence of long-term fibrosis and rejection. To test for a role of T cell infiltration and activation following ischemic injury, we induced both bilateral and unilateral renal ischemia in mice, followed by reperfusion, and then isolated mononuclear cells. Analysis of these cells by flow cytometry showed that 2 weeks after bilateral ischemia there was a significant increase of CD8(+) T cells. Furthermore, both CD4(+) and CD8(+) T cells infiltrated the injured kidney 6 weeks after unilateral ischemia. These T cells had increased expression of CD69(+) and CD44(hi)CD62L(-), markers of activation and effector-memory, respectively. CD4(+)NK1.1(+) and CD19(+) B cells were decreased in percentage both 6 and 11 weeks after bilateral or unilateral injury. There was a significant upregulation of IL-1beta, IL-6, TNF-alpha, IFN-gamma, MIP-2, and RANTES expression, measured by real-time PCR, 6 weeks after unilateral renal ischemia, further indicating T cell activation. Depletion of CD4(+) and CD8(+) T cells before ischemia caused less medullary damage and reduced kidney IFN-gamma expression, whereas their depletion following ischemia increased kidney IL-1beta; however, depletion of these cells had no effect on histological damage to the kidney. Our study demonstrates that moderate or severe kidney ischemia induces long-term T lymphocyte infiltration and cytokine/chemokine upregulation, leading to kidney structural changes.
Collapse
Affiliation(s)
- Miguel Ascon
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Dvorak CC, Hung GY, Horn B, Dunn E, Oon CY, Cowan MJ. Megadose CD34(+) cell grafts improve recovery of T cell engraftment but not B cell immunity in patients with severe combined immunodeficiency disease undergoing haplocompatible nonmyeloablative transplantation. Biol Blood Marrow Transplant 2008; 14:1125-1133. [PMID: 18804042 DOI: 10.1016/j.bbmt.2008.07.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Accepted: 07/15/2008] [Indexed: 11/27/2022]
Abstract
To determine whether T cell engraftment and recovery of B cell immunity could be improved, we prospectively treated 15 children with severe combined immunodeficiency disease (SCID) with megadoses of haplocompatible CD34(+) cells and a fixed number of CD3(+) cells without previous myeloablative chemotherapy. Evidence of T cell engraftment was seen in 73% of patients (95% confidence interval [CI] = 48%-90%). Engraftment was more likely in patients with X-linked SCID and in those with evidence of maternal engraftment at the time of diagnosis. In patients with T cell engraftment, the median time to development of a CD4 count > 200 cells/mm(3) and a phytohemagglutinin response > 50% of control was 1.2 and 4.9 months, respectively. Clearance of preexisting infections occurred after a median of 2.8 months. B cell function developed in 33% of engrafted patients (95% CI = 14%-61%). The 1-year event-free survival (EFS) rate was 60% (95% CI = 36%-80%), and the overall survival (OS) rate was 87% (95% CI = 61%-98%), with a median follow-up of 39 months. The use of megadoses of CD34(+) cells with a fixed number of CD3(+) cells in nonmyeloablative hematopoietic stem cell transplantation (HSCT) in patients with SCID is associated with excellent engraftment, T cell recovery, and OS; however, B cell function does not recover in most patients.
Collapse
Affiliation(s)
- Christopher C Dvorak
- Division of Pediatric Blood and Marrow Transplant, University of California San Francisco Children's Hospital, San Francisco, CA
| | - Giun-Yi Hung
- Department of Pediatrics, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taipai, Taiwan
| | - Biljana Horn
- Division of Pediatric Blood and Marrow Transplant, University of California San Francisco Children's Hospital, San Francisco, CA
| | - Elizabeth Dunn
- Division of Pediatric Blood and Marrow Transplant, University of California San Francisco Children's Hospital, San Francisco, CA
| | - Ching-Ying Oon
- Division of Pediatric Blood and Marrow Transplant, University of California San Francisco Children's Hospital, San Francisco, CA
| | - Morton J Cowan
- Division of Pediatric Blood and Marrow Transplant, University of California San Francisco Children's Hospital, San Francisco, CA.
| |
Collapse
|
38
|
Agrawal S, Tripathi P, Naik S. Roles and mechanism of natural killer cells in clinical and experimental transplantation. Expert Rev Clin Immunol 2008; 4:79-91. [DOI: 10.1586/1744666x.4.1.79] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|