1
|
Rawat K, Soucy SM, Kolling FW, Diaz KM, King WT, Tewari A, Jakubzick CV. Natural Antibodies Alert the Adaptive Immune System of the Presence of Transformed Cells in Early Tumorigenesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1252-1259. [PMID: 36028292 PMCID: PMC9515310 DOI: 10.4049/jimmunol.2200447] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/01/2022] [Indexed: 11/07/2022]
Abstract
Recent studies have revealed a critical role for natural Abs (NAbs) in antitumor immune responses. However, the role of NAbs in cancer immunosurveillance remains unexplored, mainly because of the lack of in vivo models that mimic the early recognition and elimination of transforming cells. In this article, we propose a role for NAbs in alerting the immune system against precancerous neoantigen-expressing cells immediately after they escape intrinsic tumor suppression mechanisms. We identify four distinct reproducible, trackable, MHC-matched neoantigen-expressing cell models that do not form tumors as the end point. This amplified readout in the critical window prior to tumor formation allows investigation of new mediators of cancer immunosurveillance. We found that neoantigen-expressing cells adoptively transferred in NAb-deficient mice persisted, whereas they were eliminated in wild-type mice, indicating that the circulating NAb repertoire alerts the immune system to the presence of transformed cells. Moreover, immunity is mounted against immunogenic and nonimmunogenic neoantigens contained in the NAb-tagged cells, regardless of whether the NAb directly recognizes the neoantigens. Beyond these neoantigen-expressing model systems, we observed a significantly greater tumor burden in chemically and virally induced tumor models in NAb-deficient mice compared with wild-type mice. Restoration of the NAb repertoire in NAb-deficient mice elicited the recognition and elimination of neoantigen-expressing cells and cancer. These data show that NAbs are required and sufficient for elimination of transformed cells early in tumorigenesis. These models can now be used to investigate how NAbs stimulate immunity via recognition receptors to eliminate precancerous cells.
Collapse
Affiliation(s)
- Kavita Rawat
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and
| | - Shannon M Soucy
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Hanover, NH
| | - Fred W Kolling
- Department of Biomedical Data Science, Dartmouth Geisel School of Medicine, Hanover, NH
| | - Kiara Manohar Diaz
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and
| | - William T King
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and
| | - Anita Tewari
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH; and
| |
Collapse
|
2
|
The Role of microRNAs in NK Cell Development and Function. Cells 2021; 10:cells10082020. [PMID: 34440789 PMCID: PMC8391642 DOI: 10.3390/cells10082020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
The clinical use of natural killer (NK) cells is at the forefront of cellular therapy. NK cells possess exceptional antitumor cytotoxic potentials and can generate significant levels of proinflammatory cytokines. Multiple genetic manipulations are being tested to augment the anti-tumor functions of NK cells. One such method involves identifying and altering microRNAs (miRNAs) that play essential roles in the development and effector functions of NK cells. Unique miRNAs can bind and inactivate mRNAs that code for cytotoxic proteins. MicroRNAs, such as the members of the Mirc11 cistron, downmodulate ubiquitin ligases that are central to the activation of the obligatory transcription factors responsible for the production of inflammatory cytokines. These studies reveal potential opportunities to post-translationally enhance the effector functions of human NK cells while reducing unwanted outcomes. Here, we summarize the recent advances made on miRNAs in murine and human NK cells and their relevance to NK cell development and functions.
Collapse
|
3
|
Khalil M, Wang D, Hashemi E, Terhune SS, Malarkannan S. Implications of a 'Third Signal' in NK Cells. Cells 2021; 10:cells10081955. [PMID: 34440725 PMCID: PMC8393955 DOI: 10.3390/cells10081955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Innate and adaptive immune systems are evolutionarily divergent. Primary signaling in T and B cells depends on somatically rearranged clonotypic receptors. In contrast, NK cells use germline-encoded non-clonotypic receptors such as NCRs, NKG2D, and Ly49H. Proliferation and effector functions of T and B cells are dictated by unique peptide epitopes presented on MHC or soluble humoral antigens. However, in NK cells, the primary signals are mediated by self or viral proteins. Secondary signaling mediated by various cytokines is involved in metabolic reprogramming, proliferation, terminal maturation, or memory formation in both innate and adaptive lymphocytes. The family of common gamma (γc) cytokine receptors, including IL-2Rα/β/γ, IL-7Rα/γ, IL-15Rα/β/γ, and IL-21Rα/γ are the prime examples of these secondary signals. A distinct set of cytokine receptors mediate a ‘third’ set of signaling. These include IL-12Rβ1/β2, IL-18Rα/β, IL-23R, IL-27R (WSX-1/gp130), IL-35R (IL-12Rβ2/gp130), and IL-39R (IL-23Rα/gp130) that can prime, activate, and mediate effector functions in lymphocytes. The existence of the ‘third’ signal is known in both innate and adaptive lymphocytes. However, the necessity, context, and functional relevance of this ‘third signal’ in NK cells are elusive. Here, we define the current paradigm of the ‘third’ signal in NK cells and enumerate its clinical implications.
Collapse
Affiliation(s)
- Mohamed Khalil
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Dandan Wang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Scott S. Terhune
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (S.S.T.); (S.M.)
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (S.S.T.); (S.M.)
| |
Collapse
|
4
|
Hashemi E, Malarkannan S. Tissue-Resident NK Cells: Development, Maturation, and Clinical Relevance. Cancers (Basel) 2020; 12:cancers12061553. [PMID: 32545516 PMCID: PMC7352973 DOI: 10.3390/cancers12061553] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells belong to type 1 innate lymphoid cells (ILC1) and are essential in killing infected or transformed cells. NK cells mediate their effector functions using non-clonotypic germ-line-encoded activation receptors. The utilization of non-polymorphic and conserved activating receptors promoted the conceptual dogma that NK cells are homogeneous with limited but focused immune functions. However, emerging studies reveal that NK cells are highly heterogeneous with divergent immune functions. A distinct combination of several activation and inhibitory receptors form a diverse array of NK cell subsets in both humans and mice. Importantly, one of the central factors that determine NK cell heterogeneity and their divergent functions is their tissue residency. Decades of studies provided strong support that NK cells develop in the bone marrow. However, evolving evidence supports the notion that NK cells also develop and differentiate in tissues. Here, we summarize the molecular basis, phenotypic signatures, and functions of tissue-resident NK cells and compare them with conventional NK cells.
Collapse
Affiliation(s)
- Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA;
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA;
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence:
| |
Collapse
|
5
|
Choi EY, Choi K, Nam G, Kim W, Chung M. H60: A Unique Murine Hematopoietic Cell-Restricted Minor Histocompatibility Antigen for Graft-versus-Leukemia Effect. Front Immunol 2020; 11:1163. [PMID: 32587590 PMCID: PMC7297985 DOI: 10.3389/fimmu.2020.01163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/12/2020] [Indexed: 11/17/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an important treatment for many types of hematological malignancies. Matching of donor and recipient for the major histocompatibility complex (MHC) improves the HSCT reconstitution, but donor-derived T cells reactive to non-MHC encoded minor histocompatibility antigens (MiHAs) can induce graft-versus-host disease (GVHD) while also being needed for graft-versus-leukemia (GVL) effects. MiHAs are allelically variant self-peptides presented conventionally on MHC molecules, but are alloantigenic in transplantation settings. Immunodominant MiHAs are most strongly associated with GVHD and GVL. There is need for mouse paradigms to understand these contradictory effects. H60 is a highly immunodominant mouse MiHA with hematopoietic cell-restricted expression. Immunodominance of H60 is tightly associated with its allelic nature (presence vs. absence of the transcripts), and the qualitative (TCR diversity) and quantitative (frequency) traits of the reactive T cells. The identity as a hematopoietic cell-restricted antigen (HRA) of H60 assists the appearance of the immunodominace in allo-HSCT circumstances, and generation of GVL effects without induction of serious GVHD after adoptive T cell transfer. Also it allows the low avidity T cells to escape thymic negative selection and exert GVL effect in the periphery, which is a previously unevaluated finding related to HRAs. In this review, we describe the molecular features and immunobiology in detail through which H60 selectively exerts its potent GVL effect. We further describe how lessons learned can be extrapolated to human allo-HCST.
Collapse
Affiliation(s)
- Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Human Environment Interface Biology, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyungho Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, South Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Giri Nam
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Woojin Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Minho Chung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
6
|
Nanbakhsh A, Srinivasamani A, Holzhauer S, Riese MJ, Zheng Y, Wang D, Burns R, Reimer MH, Rao S, Lemke A, Tsaih SW, Flister MJ, Lao S, Dahl R, Thakar MS, Malarkannan S. Mirc11 Disrupts Inflammatory but Not Cytotoxic Responses of NK Cells. Cancer Immunol Res 2019; 7:1647-1662. [PMID: 31515257 DOI: 10.1158/2326-6066.cir-18-0934] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/14/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cells generate proinflammatory cytokines that are required to contain infections and tumor growth. However, the posttranscriptional mechanisms that regulate NK cell functions are not fully understood. Here, we define the role of the microRNA cluster known as Mirc11 (which includes miRNA-23a, miRNA-24a, and miRNA-27a) in NK cell-mediated proinflammatory responses. Absence of Mirc11 did not alter the development or the antitumor cytotoxicity of NK cells. However, loss of Mirc11 reduced generation of proinflammatory factors in vitro and interferon-γ-dependent clearance of Listeria monocytogenes or B16F10 melanoma in vivo by NK cells. These functional changes resulted from Mirc11 silencing ubiquitin modifiers A20, Cbl-b, and Itch, allowing TRAF6-dependent activation of NF-κB and AP-1. Lack of Mirc11 caused increased translation of A20, Cbl-b, and Itch proteins, resulting in deubiquitylation of scaffolding K63 and addition of degradative K48 moieties on TRAF6. Collectively, our results describe a function of Mirc11 that regulates generation of proinflammatory cytokines from effector lymphocytes.
Collapse
Affiliation(s)
- Arash Nanbakhsh
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Anupallavi Srinivasamani
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Sandra Holzhauer
- Laboratory of Lymphocyte Signaling, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Matthew J Riese
- Laboratory of Lymphocyte Signaling, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin.,Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Yongwei Zheng
- Laboratory of B Cell Biology, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Demin Wang
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Laboratory of B Cell Biology, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Robert Burns
- Bioinformatics Core, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin
| | - Michael H Reimer
- Laboratory of Stem Cell Biology, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin.,Department of Cell Biology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sridhar Rao
- Laboratory of Stem Cell Biology, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin.,Department of Cell Biology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Angela Lemke
- Genome Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Shirng-Wern Tsaih
- Genome Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael J Flister
- Genome Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Shunhua Lao
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Richard Dahl
- Indiana University School of Medicine, South Bend, Indiana
| | - Monica S Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin. .,Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Genome Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
7
|
Atif SM, Gibbings SL, Redente EF, Camp FA, Torres RM, Kedl RM, Henson PM, Jakubzick CV. Immune Surveillance by Natural IgM Is Required for Early Neoantigen Recognition and Initiation of Adaptive Immunity. Am J Respir Cell Mol Biol 2018; 59:580-591. [PMID: 29953261 PMCID: PMC6236687 DOI: 10.1165/rcmb.2018-0159oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022] Open
Abstract
Early recognition of neoantigen-expressing cells is complex, involving multiple immune cell types. In this study, in vivo, we examined how antigen-presenting cell subtypes coordinate and induce an immunological response against neoantigen-expressing cells, particularly in the absence of a pathogen-associated molecular pattern, which is normally required to license antigen-presenting cells to present foreign or self-antigens as immunogens. Using two reductionist models of neoantigen-expressing cells and two cancer models, we demonstrated that natural IgM is essential for the recognition and initiation of adaptive immunity against neoantigen-expressing cells. Natural IgM antibodies form a cellular immune complex with the neoantigen-expressing cells. This immune complex licenses surveying monocytes to present neoantigens as immunogens to CD4+ T cells. CD4+ T helper cells, in turn, use CD40L to license cross-presenting CD40+ Batf3+ dendritic cells to elicit a cytotoxic T cell response against neoantigen-expressing cells. Any break along this immunological chain reaction results in the escape of neoantigen-expressing cells. This study demonstrates the surprising, essential role of natural IgM as the initiator of a sequential signaling cascade involving multiple immune cell subtypes. This sequence is required to coordinate an adaptive immune response against neoantigen-expressing cells.
Collapse
Affiliation(s)
- Shaikh M. Atif
- Department of Pediatrics, National Jewish Health, Denver, Colorado; and
| | | | | | - Faye A. Camp
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| | - Ross M. Kedl
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| | - Peter M. Henson
- Department of Pediatrics, National Jewish Health, Denver, Colorado; and
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| | - Claudia V. Jakubzick
- Department of Pediatrics, National Jewish Health, Denver, Colorado; and
- Department of Immunology and Microbiology, University of Colorado, Aurora, Colorado
| |
Collapse
|
8
|
Abel AM, Yang C, Thakar MS, Malarkannan S. Natural Killer Cells: Development, Maturation, and Clinical Utilization. Front Immunol 2018; 9:1869. [PMID: 30150991 PMCID: PMC6099181 DOI: 10.3389/fimmu.2018.01869] [Citation(s) in RCA: 757] [Impact Index Per Article: 108.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/30/2018] [Indexed: 12/25/2022] Open
Abstract
Natural killer (NK) cells are the predominant innate lymphocyte subsets that mediate anti-tumor and anti-viral responses, and therefore possess promising clinical utilization. NK cells do not express polymorphic clonotypic receptors and utilize inhibitory receptors (killer immunoglobulin-like receptor and Ly49) to develop, mature, and recognize “self” from “non-self.” The essential roles of common gamma cytokines such as interleukin (IL)-2, IL-7, and IL-15 in the commitment and development of NK cells are well established. However, the critical functions of pro-inflammatory cytokines IL-12, IL-18, IL-27, and IL-35 in the transcriptional-priming of NK cells are only starting to emerge. Recent studies have highlighted multiple shared characteristics between NK cells the adaptive immune lymphocytes. NK cells utilize unique signaling pathways that offer exclusive ways to genetically manipulate to improve their effector functions. Here, we summarize the recent advances made in the understanding of how NK cells develop, mature, and their potential translational use in the clinic.
Collapse
Affiliation(s)
- Alex M Abel
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chao Yang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Monica S Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, United States.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States.,Center of Excellence in Prostate Cancer, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
9
|
Hirayama M, Azuma E. Major and minor histocompatibility antigens to NIMA: Prediction of a tolerogenic NIMA effect. CHIMERISM 2017; 2:23-4. [PMID: 21547034 DOI: 10.4161/chim.2.1.15127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 02/10/2011] [Indexed: 12/24/2022]
Abstract
The immunologic effects of developmental exposure to non-inherited maternal antigens (NIMA) are heterogeneous, either tolerogenic or immunogenic. The role of minor histocompatibility antigens (MiHA) in NIMA effects is unknown. We have recently reported that the NIMA effect can be classified into two distinct reactivities, low and high responder, to NIMA in utero and during nursing depending on the degree of maternal microchimerism (MMc) and Foxp3 expression of peripheral blood CD4(+)CD25(+) cells after graft-versus-host disease (GVHD) induction. These reactivities were predictable before transplantation, using an MLR-ELISPOT (mixed lymphocyte reaction; enzyme-linked immunospot) assay by comparing the number of IFNγ-producing cells stimulated with NIMA. Moreover, this assay was also applicable in both major and minor NIMA-mismatched setting. These observations are clinically relevant and suggest that it is possible to predict the immunological tolerance to NIMA.
Collapse
Affiliation(s)
- Masahiro Hirayama
- Department of Pediatrics and Mie University Graduate School of Medicine; Tsu, Mie Japan
| | | |
Collapse
|
10
|
Ju JM, Kim MB, Ryu SJ, Kim JY, Chang J, Choi EY. Selection of Thymocytes Expressing Transgenic TCR Specific for a Minor Histocompatibility Antigen, H60. Immune Netw 2015; 15:222-31. [PMID: 26557806 PMCID: PMC4637343 DOI: 10.4110/in.2015.15.5.222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/03/2015] [Accepted: 09/18/2015] [Indexed: 01/11/2023] Open
Abstract
Minor histocompatibility antigens are MHC-bound peptides and contribute to the generation of allo-responses after allogeneic transplantation. H60 is a dominant minor H antigen that induces a strong CD8 T-cell response in MHC-matched allogeneic transplantation settings. Here, we report establishment of a TCR transgenic mouse line named J15, wherein T cells express TCRs specific for H60 in complex with H-2K(b), and different fates of the thymocytes expressing J15 TCRs in various thymic antigenic environments. Thymocytes expressing the J15 TCRs were positively selected and differentiated into CD8(+) single positive (SP) cells in the thymus of C57BL/6 mice, wherein the cognate antigen H60 is not expressed. However, thymocytes were negatively selected in thymus tissue where H60 was transgenically expressed under the control of the actin promoter, with double-positive stages of cells being deleted. Despite the ability of the H60H peptide (LTFHYRNL) variant to induce cytotoxic activity from H60-specific CTL lines at ~50% of the activity induced by normal H60 peptides (LTFNYRNL), J15-expressing thymocytes were positively selected in the thymus where the variant H60H was transgenically expressed. These results demonstrate that a single amino-acid change in the H60 epitope peptide influences the fate of thymocytes expressing the cognate TCR.
Collapse
Affiliation(s)
- Ji-Min Ju
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Min Bum Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Su Jeong Ryu
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Joo Young Kim
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Jun Chang
- Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
11
|
Binsfeld M, Beguin Y, Belle L, Otjacques E, Hannon M, Briquet A, Heusschen R, Drion P, Zilberberg J, Bogen B, Baron F, Caers J. Establishment of a murine graft-versus-myeloma model using allogeneic stem cell transplantation. PLoS One 2014; 9:e113764. [PMID: 25415267 PMCID: PMC4240591 DOI: 10.1371/journal.pone.0113764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 10/29/2014] [Indexed: 11/18/2022] Open
Abstract
Background Multiple myeloma (MM) is a malignant plasma cell disorder with poor long-term survival and high recurrence rates. Despite evidence of graft-versus-myeloma (GvM) effects, the use of allogeneic hematopoietic stem cell transplantation (allo-SCT) remains controversial in MM. In the current study, we investigated the anti-myeloma effects of allo-SCT from B10.D2 mice into MHC-matched myeloma-bearing Balb/cJ mice, with concomitant development of chronic graft-versus-host disease (GvHD). Methods and results Balb/cJ mice were injected intravenously with luciferase-transfected MOPC315.BM cells, and received an allogeneic (B10.D2 donor) or autologous (Balb/cJ donor) transplant 30 days later. We observed a GvM effect in 94% of the allogeneic transplanted mice, as the luciferase signal completely disappeared after transplantation, whereas all the autologous transplanted mice showed myeloma progression. Lower serum paraprotein levels and lower myeloma infiltration in bone marrow and spleen in the allogeneic setting confirmed the observed GvM effect. In addition, the treated mice also displayed chronic GvHD symptoms. In vivo and in vitro data suggested the involvement of effector memory CD4 and CD8 T cells associated with the GvM response. The essential role of CD8 T cells was demonstrated in vivo where CD8 T-cell depletion of the graft resulted in reduced GvM effects. Finally, TCR Vβ spectratyping analysis identified Vβ families within CD4 and CD8 T cells, which were associated with both GvM effects and GvHD, whereas other Vβ families within CD4 T cells were associated exclusively with either GvM or GvHD responses. Conclusions We successfully established an immunocompetent murine model of graft-versus-myeloma. This is the first murine GvM model using immunocompetent mice that develop MM which closely resembles human MM disease and that are treated after disease establishment with an allo-SCT. Importantly, using TCR Vβ spectratyping, we also demonstrated the presence of GvM unique responses potentially associated with the curative capacity of this immunotherapeutic approach.
Collapse
Affiliation(s)
- Marilène Binsfeld
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Ludovic Belle
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Eléonore Otjacques
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Muriel Hannon
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Alexandra Briquet
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | | | - Jenny Zilberberg
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey, United States of America
| | - Bjarne Bogen
- Centre for Immune Regulation, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
- KG Jebsen centre for research on influenza vaccines, Institute of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Frédéric Baron
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
12
|
Hirayama M, Azuma E. Major and minor histocompatibility antigens to NIMA. CHIMERISM 2014. [DOI: 10.4161/chim.15127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
13
|
Impact of genomic polymorphisms on the repertoire of human MHC class I-associated peptides. Nat Commun 2014; 5:3600. [PMID: 24714562 PMCID: PMC3996541 DOI: 10.1038/ncomms4600] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 03/10/2014] [Indexed: 12/23/2022] Open
Abstract
For decades, the global impact of genomic polymorphisms on the repertoire of peptides presented by major histocompatibility complex (MHC) has remained a matter of speculation. Here we present a novel approach that enables high-throughput discovery of polymorphic MHC class I-associated peptides (MIPs), which play a major role in allorecognition. On the basis of comprehensive analyses of the genomic landscape of MIPs eluted from B lymphoblasts of two MHC-identical siblings, we show that 0.5% of non-synonymous single nucleotide variations are represented in the MIP repertoire. The 34 polymorphic MIPs found in our subjects are encoded by bi-allelic loci with dominant and recessive alleles. Our analyses show that, at the population level, 12% of the MIP-coding exome is polymorphic. Our method provides fundamental insights into the relationship between the genomic self and the immune self and accelerates the discovery of polymorphic MIPs (also known as minor histocompatibility antigens). Mass spectrometry (MS) has furthered our understanding of MHC class I-associated peptides (MIPs), but the technique is inadequate for studying MIP-associated polymorphisms. Here, the authors combine high-throughput MS with exome and transcriptome sequencing to identify polymorphic MIPs from two female siblings.
Collapse
|
14
|
Signaling by Fyn-ADAP via the Carma1-Bcl-10-MAP3K7 signalosome exclusively regulates inflammatory cytokine production in NK cells. Nat Immunol 2013; 14:1127-36. [PMID: 24036998 PMCID: PMC3855032 DOI: 10.1038/ni.2708] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/09/2013] [Indexed: 11/29/2022]
Abstract
Inflammation is a critical component of the immune response. However, acute or chronic inflammation can be highly destructive. Uncontrolled inflammation forms the basis for allergy, asthma, and multiple autoimmune disorders. Here, we identify a signaling pathway that is exclusively responsible for inflammatory cytokine production but not for cytotoxicity. Recognition of H60+ or CD137L+ tumor cells by murine NK cells led to efficient cytotoxicity and inflammatory cytokine production. Both of these effector functions required Lck, Fyn, PI(3)K-p85α, PI(3)K-p110δ, and PLC-γ2. However, the complex of Fyn and the adapter ADAP exclusively regulated inflammatory cytokine production but not cytotoxicity in NK cells. This unique function of ADAP required a Carma1-Bcl10-MAP3K7 signaling axis. Our results identify molecules that can be targeted to regulate inflammation without compromising NK cell cytotoxicity.
Collapse
|
15
|
Hirayama M, Azuma E, Komada Y. Tolerogenic effect of non-inherited maternal antigens in hematopoietic stem cell transplantation. Front Immunol 2012; 3:135. [PMID: 22654885 PMCID: PMC3360462 DOI: 10.3389/fimmu.2012.00135] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 05/10/2012] [Indexed: 01/03/2023] Open
Abstract
Major histocompatibility complex antigens that provoke severe transplant reactions are referred to as the human leukocyte antigen (HLA) in human and as the H-2 in mice. Even if the donor and recipient are HLA-identical siblings, graft-versus-host reactions have been linked to differences in the minor histocompatibility antigen. As the chance of finding an HLA-identical sibling donor is only 25%, attention has been focused on using alternative donors. An HLA-mismatched donor with non-inherited maternal antigens (NIMA) is less immunogenic than that with non-inherited paternal antigens, because the contact between the immune systems of the mother and child during pregnancy affects the immune response of the child against NIMA. However, the immunologic effects of developmental exposure to NIMA are heterogeneous, and can be either tolerogenic or immunogenic. We recently have devised a novel method for predicting the tolerogenic effect of NIMA. In this review, we overview the evidence for the existence of the NIMA tolerogenic effect, the possible cellular and molecular basis of the phenomenon, and its utilization in hematopoietic stem cell transplantation. We suggest a future direction for the safe clinical use of this phenomenon, fetomaternal tolerance, in the transplantation field.
Collapse
Affiliation(s)
- Masahiro Hirayama
- Department of Pediatrics and Cell Transplantation, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | | |
Collapse
|
16
|
Kato G, Kondo H, Aoki T, Hirono I. A novel immune-related gene, microtubule aggregate protein homologue, is up-regulated during IFN-γ-related immune responses in Japanese flounder, Paralichthys olivaceus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 36:349-358. [PMID: 21824491 DOI: 10.1016/j.dci.2011.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/02/2011] [Accepted: 06/02/2011] [Indexed: 05/31/2023]
Abstract
Delayed-type hypersensitivity (DTH) response mediated by antigen-specific Th1 cells is used as a test to detect exposure to tuberculosis in humans. Japanese flounder (Paralichthys olivaceus) microtubule aggregate protein homologue (PoMTAP) was identified as a gene strongly induced during fish DTH response. In this study, PoMTAP gene was cloned and its expression profile was analyzed. The PoMTAP gene has a transcriptional regulatory region that includes two interferon-stimulated response elements and two IFN-γ activated sites. Expressions of PoMTAP and IFN-γ genes were up-regulated at the same time points during the DTH response, Edwardsiella tarda infection and VHSV infection. Furthermore, PoMTAP gene expressing cells also expressed CD3ε, confirming that PoMTAP is expressed by T lymphocytes. These results suggest that PoMTAP is a novel immune-related gene expressed by T lymphocytes that is preferentially induced by IFN-γ and has a role in Th1-mediated immune responses in Japanese flounder.
Collapse
Affiliation(s)
- Goshi Kato
- Laboratory of Genome Science, Graduate School of Tokyo University of Marine Science and Technology, Tokyo, Japan
| | | | | | | |
Collapse
|
17
|
Memory T cells from minor histocompatibility antigen-vaccinated and virus-immune donors improve GVL and immune reconstitution. Blood 2011; 118:5965-76. [PMID: 21917752 DOI: 10.1182/blood-2011-07-367011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Donor T cells contribute to the success of allogeneic hematopoietic stem cell transplantation (alloSCT). Alloreactive donor T cells attack leukemia cells, mediating the GVL effect. Donor T cells, including the memory T cells (T(M)) that are generated after infection, also promote immune reconstitution. Nonetheless, leukemia relapse and infection are major sources of treatment failure. Efforts to augment GVL and immune reconstitution have been limited by GVHD, the attack by donor T cells on host tissues. One approach to augmenting GVL has been to infuse ex vivo-generated T cells with defined specificities; however, this requires expertise that is not widely available. In the present study, we tested an alternative approach, adoptive immunotherapy with CD8+ T(M) from donors vaccinated against a single minor histocompatibility antigen (miHA) expressed by leukemia cells. Vaccination against the miHA H60 greatly augmented T(M)-mediated GVL against mouse chronic-phase (CP-CML) and blast crisis chronic myeloid leukemia (BC-CML). T(M)-mediated GVL was antigen specific and was optimal when H60 expression was hematopoietically restricted. Even when H60 was ubiquitous, donor H60 vaccination had a minimal impact on GVHD. T(M) from lymphocytic choriomeningitis virus (LCMV)-immune and H60-vaccinated donors augmented GVL and protected recipients from LCMV. These data establish a strategy for augmenting GVL and immune reconstitution without elaborate T-cell manipulation.
Collapse
|
18
|
Rajasekaran K, Chu H, Kumar P, Xiao Y, Tinguely M, Samarakoon A, Kim TW, Li X, Thakar MS, Zhang J, Malarkannan S. Transforming growth factor-beta-activated kinase 1 regulates natural killer cell-mediated cytotoxicity and cytokine production. J Biol Chem 2011; 286:31213-24. [PMID: 21771792 DOI: 10.1074/jbc.m111.261917] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Carma1, a caspase recruitment domain-containing membrane-associated guanylate kinase, initiates a unique signaling cascade via Bcl10 and Malt1 in NK cells. Carma1 deficiency results in reduced phosphorylation of JNK1/2 and activation of NF-κB that lead to impaired NK cell-mediated cytotoxicity and cytokine production. However, the precise identities of the downstream signaling molecules that link Carma1 to these effector functions were not defined. Here we show that transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) is abundantly present in NK cells, and activation via NKG2D results in its phosphorylation. Lack of Carma1 considerably reduced TAK1 phosphorylation, demonstrating the dependence of TAK1 on Carma1 in NKG2D-mediated NK cell activations. Pharmacological inhibitor to TAK1 significantly reduced NK-mediated cytotoxicity and its potential to generate IFN-γ, GM-CSF, MIP-1α, MIP-1β, and RANTES. Conditional in vivo knockdown of TAK1 in NK cells from Mx1Cre(+)TAK1(fx/fx) mice resulted in impaired NKG2D-mediated cytotoxicity and cytokine/chemokine production. Inhibition or conditional knockdown of TAK1 severely impaired the NKG2D-mediated phosphorylation of ERK1/2 and JNK1/2 and activation of NF-κB and AP1. Our results show that TAK1 links Carma1 to NK cell-mediated effector functions.
Collapse
Affiliation(s)
- Kamalakannan Rajasekaran
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Araki M, Hirayama M, Azuma E, Kumamoto T, Iwamoto S, Toyoda H, Ito M, Amano K, Komada Y. Prediction of reactivity to noninherited maternal antigen in MHC-mismatched, minor histocompatibility antigen-matched stem cell transplantation in a mouse model. THE JOURNAL OF IMMUNOLOGY 2010; 185:7739-45. [PMID: 21078914 DOI: 10.4049/jimmunol.1001226] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The immunologic effects of developmental exposure to noninherited maternal Ags (NIMAs) are quite variable. Both tolerizing influence and inducing alloreaction have been observed on clinical transplantation. The role of minor histocompatibility Ags (MiHAs) in NIMA effects is unknown. MiHA is either matched or mismatched in NIMA-mismatched transplantation because a donor of the transplantation is usually limited to a family member. To exclude the participation of MiHA in a NIMA effect for MHC (H-2) is clinically relevant because mismatched MiHA may induce severe alloreaction. The aim of this study is to understand the mechanism of NIMA effects in MHC-mismatched, MiHA-matched hematopoietic stem cell transplantation. Although all offsprings are exposed to the maternal Ags, the NIMA effect for the H-2 Ag was not evident. However, they exhibit two distinct reactivities, low and high responder, to NIMA in utero and during nursing depending on the degree of maternal microchimerism. Low responders survived longer with less graft-versus-host disease. These reactivities were correlated with Foxp3 expression of peripheral blood CD4(+)CD25(+) cells after graft-versus-host disease induction and the number of IFN-γ-producing cells stimulated with NIMA pretransplantation. These observations are clinically relevant and suggest that it is possible to predict the immunological tolerance to NIMA.
Collapse
Affiliation(s)
- Mariko Araki
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Minor histocompatibility antigens: presentation principles, recognition logic and the potential for a healing hand. Curr Opin Organ Transplant 2010; 15:512-25. [PMID: 20616723 DOI: 10.1097/mot.0b013e32833c1552] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW There is ample evidence indicating a pathologic role for minor histocompatibility antigens in inciting graft-versus-host disease in major histocompatibility complex (MHC)-matched bone marrow transplantation and rejection of solid organ allografts. Here we review the current knowledge of the genetic and biochemical bases for the cause of minor histoincompatibility and the structural basis for the recognition of the resulting alloantigens by the T-cell receptor. RECENT FINDINGS Recent evidence indicates that we as independently conceived individuals are genetically unique, thus, offering a mechanism for minor histoincompatibility between MHC-identical donor-recipient pairs. Furthermore, advances in delineating the mechanisms underlying antigen cross-presentation by MHC class I molecules and a critical role for autophagy in presenting cytoplasmic antigens by MHC class II molecules have been made. These new insights coupled with the X-ray crystallographic solution of several peptide/MHC-T-cell receptor structures have revealed mechanisms of histoincompatibility. SUMMARY On the basis of these new insights, ways to test for allograft compatibility and concoction of immunotherapies are discussed.
Collapse
|
21
|
Xu H, Huang Y, Hussain LR, Zhu Z, Bozulic LD, Ding C, Yan J, Ildstad ST. Sensitization to minor antigens is a significant barrier in bone marrow transplantation and is prevented by CD154:CD40 blockade. Am J Transplant 2010; 10:1569-79. [PMID: 20642683 PMCID: PMC3195648 DOI: 10.1111/j.1600-6143.2010.03148.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Sensitization to major histocompatibility complex (MHC) alloantigens is critical in transplantation rejection. The mechanism of sensitization to minor histocompatibility antigens (Mi-HAg) has not been thoroughly explored. We used a mouse model of allosensitization to Mi-HAg to study the Mi-HAg sensitization barrier in bone marrow transplantation (BMT). AKR mice were sensitized with MHC congenic Mi-HAg disparate B10.BR skin grafts. Adaptive humoral (B-cells) and cellular (T cells) responses to Mi-HAg are elicited. In subsequent BMT, only 20% of sensitized mice engrafted, while 100% of unsensitized mice did. In vivo cytotoxicity assays showed that Mi-HAg sensitized AKR mice eliminated CFSE labeled donor splenocytes significantly more rapidly than naïve AKR mice but less rapidly than MHC-sensitized recipients. Sera from Mi-HAg sensitized mice also reacted with cells from other mouse strains, suggesting that Mi-HAg peptides were broadly shared between mouse strains. The production of anti-donor-Mi-HAg antibodies was totally prevented in mice treated with anti-CD154 during skin grafting, suggesting a critical role for the CD154:CD40 pathway in B-cell reactivity to Mi-HAg. Moreover, anti-CD154 treatment promoted BM engraftment to 100% in recipients previously sensitized to donor Mi-HAg. Taken together, Mi-HAg sensitization poses a significant barrier in BMT and can be overcome with CD154:CD40 costimulatory blockade.
Collapse
Affiliation(s)
- Hong Xu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202
| | - Yiming Huang
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202
| | - Lala R. Hussain
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202
| | - Ziqiang Zhu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202
| | - Larry D. Bozulic
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202
| | - Chuanlin Ding
- James Brown Cancer Center, University of Louisville, Louisville, KY 40202
| | - Jun Yan
- James Brown Cancer Center, University of Louisville, Louisville, KY 40202
| | - Suzanne T. Ildstad
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202,Correspondence should be addressed to (STI): Suzanne T. Ildstad, M.D., Director Institute for Cellular Therapeutics Jewish Hospital Distinguished Professor of Transplantation Distinguished University Scholar Professor of Surgery University of Louisville 570 South Preston Street, Suite 404 Louisville, Kentucky 40202-1760, USA Telephone: 502-852-2080 Fax: 502-852-2079
| |
Collapse
|
22
|
Escande-Beillard N, Washburn L, Zekzer D, Wu ZP, Eitan S, Ivkovic S, Lu Y, Dang H, Middleton B, Bilousova TV, Yoshimura Y, Evans CJ, Joyce S, Tian J, Kaufman DL. Neurons preferentially respond to self-MHC class I allele products regardless of peptide presented. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:816-23. [PMID: 20018625 PMCID: PMC2997386 DOI: 10.4049/jimmunol.0902159] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Studies of mice lacking MHC class I (MHC I)-associated proteins have demonstrated a role for MHC I in neurodevelopment. A central question arising from these observations is whether neuronal recognition of MHC I has specificity for the MHC I allele product and the peptide presented. Using a well-established embryonic retina explant system, we observed that picomolar levels of a recombinant self-MHC I molecule inhibited neurite outgrowth. We then assessed the neurobiological activity of a panel of recombinant soluble MHC Is, consisting of different MHC I heavy chains with a defined self- or nonself-peptide presented, on cultured embryonic retinas from mice with different MHC I haplotypes. We observed that self-MHC I allele products had greater inhibitory neuroactivity than nonself-MHC I molecules, regardless of the nature of the peptide presented, a pattern akin to MHC I recognition by some innate immune system receptors. However, self-MHC I molecules had no effect on retinas from MHC I-deficient mice. These observations suggest that neuronal recognition of MHC I may be coordinated with the inherited MHC I alleles, as occurs in the innate immune system. Consistent with this notion, we show that MHC I and MHC I receptors are coexpressed by precursor cells at the earliest stages of retina development, which could enable such coordination.
Collapse
Affiliation(s)
- Nathalie Escande-Beillard
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fanning SL, Appel MY, Berger SA, Korngold R, Friedman TM. The immunological impact of genetic drift in the B10.BR congenic inbred mouse strain. THE JOURNAL OF IMMUNOLOGY 2009; 183:4261-72. [PMID: 19752227 DOI: 10.4049/jimmunol.0900971] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The MHC-matched, minor histocompatibility Ag (miHA)-mismatched B10.BR-->CBA strain combination has been used to elucidate the immunobiology of graft-vs-host disease (GVHD) following allogeneic bone marrow transplantation. Studies conducted in the 1980s had established that B10.BR CD8+ T cells were capable of mediating GVHD in the absence of CD4+ T cells, and that CD4+ T cells were unable to induce lethal disease. In more recent studies with this GVHD model, we detected etiological discrepancies with the previously published results, which suggested that genetic drift might have occurred within the B10.BR strain. In particular, there was increased allorecognition of CBA miHA by B10.BR CD4+ T cells, as determined by both TCR Vbeta spectratype analysis and the induction of lethal GVHD in CBA recipients. Additionally, alloreactivity was observed between the genetically drifted mice (B10.BR/Jdrif) and mice rederived from frozen embryos of the original strain (B10.BR/Jrep) using Vbeta spectratype analysis and IFN-gamma ELISPOT assays, suggesting that new miHA differences had arisen between the mice. Furthermore, T cell-depleted B10.BR/Jdrif bone marrow cells were unable to provide long-term survival following either allogeneic or syngeneic bone marrow transplantation. Gene expression analysis revealed several genes involved in hematopoiesis that were overexpressed in the lineage-negative fraction of B10.BR/Jdrif bone marrow, as compared with B10.BR/Jrep mice. Taken together, these results suggest that genetic drift in the B10.BR strain has significantly impacted the immune alloreactive response in the GVHD model by causing altered expression of miHA and diminished capacity for survival following transplantation into lethally irradiated recipients.
Collapse
Affiliation(s)
- Stacey L Fanning
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | | | | | | | | |
Collapse
|
24
|
Starck SR, Ow Y, Jiang V, Tokuyama M, Rivera M, Qi X, Roberts RW, Shastri N. A distinct translation initiation mechanism generates cryptic peptides for immune surveillance. PLoS One 2008; 3:e3460. [PMID: 18941630 PMCID: PMC2565129 DOI: 10.1371/journal.pone.0003460] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 09/29/2008] [Indexed: 12/04/2022] Open
Abstract
MHC class I molecules present a comprehensive mixture of peptides on the cell surface for immune surveillance. The peptides represent the intracellular protein milieu produced by translation of endogenous mRNAs. Unexpectedly, the peptides are encoded not only in conventional AUG initiated translational reading frames but also in alternative cryptic reading frames. Here, we analyzed how ribosomes recognize and use cryptic initiation codons in the mRNA. We find that translation initiation complexes assemble at non-AUG codons but differ from canonical AUG initiation in response to specific inhibitors acting within the peptidyl transferase and decoding centers of the ribosome. Thus, cryptic translation at non-AUG start codons can utilize a distinct initiation mechanism which could be differentially regulated to provide peptides for immune surveillance.
Collapse
Affiliation(s)
- Shelley R. Starck
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Yongkai Ow
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Vivian Jiang
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Maria Tokuyama
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Mark Rivera
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Xin Qi
- Howard Hughes Medical Institute and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Richard W. Roberts
- Department of Chemistry, Chemical Engineering, and Biology, University of Southern California, Los Angeles, California, United States of America
| | - Nilabh Shastri
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Rezvani K, Barrett AJ. Characterizing and optimizing immune responses to leukaemia antigens after allogeneic stem cell transplantation. Best Pract Res Clin Haematol 2008; 21:437-53. [PMID: 18790448 PMCID: PMC3757471 DOI: 10.1016/j.beha.2008.07.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Allogeneic stem cell transplantation remains a curative treatment for haematological malignancies resistant to other treatment approaches through the unique graft-versus-leukaemia effect (GvL). However, the lack of specificity of this response results in the targeting of normal tissue, and the morbidity and mortality associated with graft-versus-host disease (GvHD). Further improvements in exploiting the GvL effect to prevent relapse in high-risk leukaemias while minimizing toxicity have focused on the use of targeted anti-leukaemic immunotherapy. These strategies include the use of vaccines against minor histocompatibility antigens (HA-1, HA-2 and H-Y) and leukaemia-specific antigens (proteinase 3, Wilms' tumour 1 and BCR-ABL), and the adoptive transfer of leukaemia-specific T cells. The unique post-transplant milieu, which is characterized by lymphopenia, regulatory T-cell depletion and the release of growth factors, offers the opportunity to promote the expansion of engrafted T cells and enhance the specific GvL response. Techniques to reduce regulatory T-cell control over T-cell responses to leukaemia antigens could further enhance GvL reactivity. Finally, these approaches to increase GvL effects would be facilitated by transplant approaches to deplete GvHD alloresponses selectively while preserving GvL reactivity.
Collapse
Affiliation(s)
- Katayoun Rezvani
- Department of Haematology, 4th Floor Commonwealth Building, Hammersmith Hospital, DuCane Road, London W12 0NN, UK.
| | | |
Collapse
|
26
|
Takada A, Yoshida S, Kajikawa M, Miyatake Y, Tomaru U, Sakai M, Chiba H, Maenaka K, Kohda D, Fugo K, Kasahara M. Two novel NKG2D ligands of the mouse H60 family with differential expression patterns and binding affinities to NKG2D. THE JOURNAL OF IMMUNOLOGY 2008; 180:1678-85. [PMID: 18209064 DOI: 10.4049/jimmunol.180.3.1678] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
H60, originally described as a dominant minor histocompatibility Ag, is an MHC class I-like molecule that serves as a ligand for the NKG2D receptor. In the present study, we identified two novel mouse chromosome 10-encoded NKG2D ligands structurally resembling H60. These ligands, which we named H60b and H60c, encode MHC class I-like molecules with two extracellular domains. Whereas H60b has a transmembrane region, H60c is a GPI-anchored protein. Recombinant soluble H60b and H60c proteins bound to NKG2D with affinities typical of cell-cell recognition receptors (K(d) = 310 nM for H60b and K(d) = 8.7 muM for H60c). Furthermore, expression of H60b or H60c rendered Ba/F3 cells susceptible to lysis by NK cells, thereby establishing H60b and H60c as functional ligands for NKG2D. H60b and H60c transcripts were detected only at low levels in tissues of healthy adult mice. Whereas H60b transcripts were detectable in various tissues, H60c transcripts were detected mainly in the skin. Infection of mouse embryonic fibroblasts with murine cytomegalovirus induced expression of H60b, but not H60c or the previously known H60 gene, indicating that transcriptional activation of the three types of H60 genes is differentially regulated. The present study adds two new members to the current list of NKG2D ligands.
Collapse
Affiliation(s)
- Akio Takada
- Department of Pathology, Hokkaido University Graduate School of Medicine, North-15 West-7, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hyka-Nouspikel N, Lucian L, Murphy E, McClanahan T, Phillips JH. DAP10 deficiency breaks the immune tolerance against transplantable syngeneic melanoma. THE JOURNAL OF IMMUNOLOGY 2007; 179:3763-71. [PMID: 17785813 DOI: 10.4049/jimmunol.179.6.3763] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
DAP10, an activating adaptor protein, associates with the NKG2D protein to form a multisubunit receptor complex that is expressed in lymphoid and myeloid cells. The ligands for NKG2D-DAP10 receptor are expressed in both normal and tumor cells, suggesting distinct roles for this receptor in autoimmunity and cancer. In this study, we report that constitutive DAP10 activating signaling is part of regulatory mechanisms that control immunity against tumors. Mice lacking DAP10 (DAP10KO), showed enhanced immunity against melanoma malignancies due to hyperactive functioning of NK1.1+CD3+ NKT cells. DAP10 deficiency resulted in substantially increased NKT cell functions, including cytokine production and cytotoxicity, leading to efficient killing of melanoma tumors. Moreover, the antitumor phenotype of DAP10KO mice correlated with impaired activation status of CD4+CD25+ T regulatory cells (Tregs). Upon activation, DAP10KO Tregs maintained higher levels of IL-2 and produced significantly lower amounts of IL-10 and IFN-gamma cytokines when compared with wild-type Tregs. Our data suggest that DAP10 signaling is involved in adjusting the activation threshold and generation of NKT cells and Tregs to avoid autoreactivity, but also modulates antitumor mechanisms.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Immune Tolerance/genetics
- Immunophenotyping
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Lymphocyte Activation/genetics
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Transplantation
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/physiology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Nevila Hyka-Nouspikel
- Cellular Immunology Laboratory, Department of Discovery Research, Schering-Plough Biopharma, Palo Alto, CA 94304, USA.
| | | | | | | | | |
Collapse
|
28
|
Malarkannan S, Regunathan J, Chu H, Kutlesa S, Chen Y, Zeng H, Wen R, Wang D. Bcl10 plays a divergent role in NK cell-mediated cytotoxicity and cytokine generation. THE JOURNAL OF IMMUNOLOGY 2007; 179:3752-62. [PMID: 17785812 DOI: 10.4049/jimmunol.179.6.3752] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activating receptors such as NKG2D and Ly49D mediate a multitude of effector functions including cytotoxicity and cytokine generation in NK cells. However, specific signaling events that are responsible for the divergence of distinct effector functions have yet to be determined. In this study, we show that lack of caspase recruitment domain-containing protein Bcl10 significantly affected receptor-mediated cytokine and chemokine generation, but not cytotoxicity against tumor cells representing "missing-self" or "induced-self." Lack of Bcl10 completely abrogated the generation of GM-CSF and chemokines and it significantly reduced the generation of IFN-gamma (>75%) in NK cells. Commitment, development, and terminal maturation of NK cells were largely unaffected in the absence of Bcl10. Although IL-2-activated NK cells could mediate cytotoxicity to the full extent, the ability of the freshly isolated NK cells to mediate cytotoxicity was somewhat reduced. Therefore, we conclude that the Carma1-Bcl10-Malt1 signaling axis is critical for cytokine and chemokine generation, although it is dispensable for cytotoxic granule release depending on the activation state of NK cells. These results indicate that Bcl10 represents an exclusive "molecular switch" that links the upstream receptor-mediated signaling to cytokine and chemokine generations.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/biosynthesis
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/physiology
- Animals
- Antigens, Ly/physiology
- Antigens, Surface/physiology
- B-Cell CLL-Lymphoma 10 Protein
- CHO Cells
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line, Tumor
- Chemokines/antagonists & inhibitors
- Chemokines/biosynthesis
- Cricetinae
- Cricetulus
- Cytokines/antagonists & inhibitors
- Cytokines/biosynthesis
- Cytotoxicity, Immunologic/genetics
- Immunity, Innate/genetics
- Interleukin-2/physiology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lectins, C-Type/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NK Cell Lectin-Like Receptor Subfamily A
- NK Cell Lectin-Like Receptor Subfamily B
- NK Cell Lectin-Like Receptor Subfamily K
- Receptors, Immunologic/physiology
- Receptors, NK Cell Lectin-Like
- Receptors, Natural Killer Cell
- Self Tolerance/genetics
- Self Tolerance/immunology
Collapse
|
29
|
Archbold JK, Ely LK, Kjer-Nielsen L, Burrows SR, Rossjohn J, McCluskey J, Macdonald WA. T cell allorecognition and MHC restriction--A case of Jekyll and Hyde? Mol Immunol 2007; 45:583-98. [PMID: 17869342 DOI: 10.1016/j.molimm.2006.05.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Accepted: 05/19/2006] [Indexed: 01/14/2023]
Abstract
A great paradox in cellular immunology is how T cell allorecognition exists at high frequencies (up to 10%) despite the stringent requirements of discriminating 'self' from 'non-self' imposed by MHC restriction. Thus, in tissue transplantation, a substantial proportion of the recipient's T cells will have the ability to recognize the graft and instigate an immune response against the transplanted tissue, ultimately resulting in graft rejection--a manifestation of T cell alloreactivity. Transplantation of human organs and lymphoid cells as treatment for otherwise life-threatening diseases has become a more routine medical procedure making this problem of great importance. Immunologists have gained important insights into the mechanisms of T cell alloreactivity from cytotoxic T cell assays, affinity-avidity studies, and crystal structures of peptide-MHC (pMHC) molecules and T cell receptors (TCRs) both alone and in complex. Despite the clinical significance of alloreactivity, the crystal structure of an alloreactive human TCR in complex with both cognate pMHC and an allogeneic pMHC complex has yet to be determined. This review highlights some of the important findings from studies characterizing the way in which alloreactive T cell receptors and pMHC molecules interact in an attempt to resolve this great irony of the cellular immune response.
Collapse
Affiliation(s)
- Julia K Archbold
- The Protein Crystallography Unit, Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Allogeneic haematopoietic stem-cell transplantation (SCT) is a curative therapy for haematological malignancies and inherited disorders of blood cells, such as sickle-cell anaemia. Mature alphabeta T cells that are contained in the allografts reconstitute T-cell immunity and can eradicate malignant cells in the recipient. Unfortunately, these T cells recognize the recipient as 'non-self' and employ a wide range of immune mechanisms to attack recipient tissues in a process known as graft-versus-host disease (GVHD). The full therapeutic potential of allogeneic haematopoietic SCT will not be realized until approaches to minimize GVHD, while maintaining the positive contributions of donor T cells, are developed. This Review focuses on research in mouse models pursued to achieve this goal.
Collapse
Affiliation(s)
- Warren D Shlomchik
- Yale University School of Medicine, sections of Medical Oncology and Immunobiology, PO BOX 208032, New Haven, Connecticut 06520, USA.
| |
Collapse
|
31
|
Wettstein PJ, Borson ND. Distributions of single nucleotide polymorphisms in differential chromosome segments of congenic resistant strains that define minor histocompatibility antigens. Immunogenetics 2007; 59:631-9. [PMID: 17541577 DOI: 10.1007/s00251-007-0231-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 05/08/2007] [Indexed: 01/25/2023]
Abstract
Minor histocompatibility antigens (MiHAs) stimulate the rejection of allografts when donors and recipients are matched at the major histocompatibility complex (MHC). The majority of identified autosomal MiHAs were generated by non-synonymous (NS) substitutions that alter MHC class I-binding peptides. The mosaic distribution of single nucleotide polymorphisms (SNPs) that distinguish inbred mouse strains led us to hypothesize that MiHA genes defined by congenic strains on C57BL/6 and C57BL/10 backgrounds map to chromosomal regions with relatively high numbers of NS SNPs that distinguish C57 strains from other common inbred strains. To test this hypothesis, we mapped the ends of differential chromosome segments of congenic strains, which define 12 MiHAs, relative to microsatellites and SNPs. The lengths of differential segments ranged from 9.7 to 105.9 Mbp in congenic strains where no attempts were made to select recombinants within these segments. There was no apparent correlation between differential segment length and number of backcrosses, suggesting that factors other than the number of opportunities for recombination affected the differential segment lengths in these congenics. These differential segments included higher numbers of NS SNPs that distinguish C57BL/6J from A/J, DBA/2J, and 129S1/J than would be predicted if these SNPs were uniformly distributed along the chromosomes. The most extreme case was the H8 congenic that included 74% of the SNPs on chromosome 14 within its 9.7-11.1 Mbp differential segment. These results point toward a direct relationship between the level of genomic divergence, as indicated by numbers of NS SNPs, and numbers of MiHAs that collectively determine the magnitude of allograft rejection.
Collapse
Affiliation(s)
- Peter J Wettstein
- Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | | |
Collapse
|
32
|
Brickner AG. Mechanisms of minor histocompatibility antigen immunogenicity: the role of infinitesimal versus structurally profound polymorphisms. Immunol Res 2007; 36:33-41. [PMID: 17337764 DOI: 10.1385/ir:36:1:33] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
Minor histocompatibility antigens (mHAgs) are a diverse collection of major histocompatibility complex (MHC)-bound peptides that play a critical role in the induction of detrimental graft-versus-host disease (GVHD) or the development of beneficial graft-versustumor (GVT) effects after allogeneic hematopoietic stem cell transplantation. mHAgs are a consequence of allelic polymorphism that translates to disparity in MHC-presented peptide epitopes between transplant donor and recipient. This donor/recipient allelic disparity can range from infinitesimal amino side chain differences between MHC-presented peptides, to profound structural polymorphisms in genes and proteins that can nullify transcription or translation of one allelic variant and result in the complete abrogation of its presentation by MHC.
Collapse
Affiliation(s)
- Anthony G Brickner
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-1863, USA.
| |
Collapse
|
33
|
Hyka-Nouspikel N, Phillips JH. Physiological roles of murine DAP10 adapter protein in tumor immunity and autoimmunity. Immunol Rev 2007; 214:106-17. [PMID: 17100879 DOI: 10.1111/j.1600-065x.2006.00456.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The immune system has evolved to tolerate what is self and reject what is foreign. The recognition of self from non-self is performed by activating and inhibitory receptors, which signal immune cells via adapter molecules, determining the outcome of the immune response. DAP10, a transmembrane adapter protein expressed broadly in hematopoietic cells, associates with NKG2D activating receptor forming a multisubunit complex, which recognizes self-proteins upregulated during tumorigenesis, infection, and autoimmune response. Analysis of immune reactions against syngeneic tumors, as well as autoimmune responses in the DAP10-deficient mice, revealed an important physiological role of DAP10 signaling in maintaining tolerance to self, probably by controlling the development and activation threshold of autoreactive T cells.
Collapse
Affiliation(s)
- Nevila Hyka-Nouspikel
- Cellular Immunology Laboratory, Department of Discovery Research, Schering-Plough Biopharma (formerly DNAX Research, Inc.), Palo Alto, CA 94304, USA
| | | |
Collapse
|
34
|
de Witte MA, Toebes M, Song JY, Wolkers MC, Schumacher TNM. Effective graft depletion of MiHAg T-cell specificities and consequences for graft-versus-host disease. Blood 2007; 109:3830-8. [PMID: 17202318 DOI: 10.1182/blood-2006-07-037713] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Minor histocompatibility antigen (MiHAg) differences between donor and recipient in MHC-matched allogeneic hematopoietic stem cell transplantation (allo-HSCT) often result in graft-versus-host disease (GVHD). While MiHAg-specific T-cell responses can in theory be directed against a large number of polymorphic differences between donor and recipient, in practice, T-cell responses against only a small set of MiHAgs appear to dominate the immune response, and it has been suggested that immunodominance may predict an important contribution to the development of GVHD. Here, we addressed the feasibility of graft engineering by ex vivo removal of T cells with 1 or more defined antigen specificities in a well-characterized experimental HSCT model (B6 → BALB.B). We demonstrate that immunodominant H60- and H4-specific CD8+ T-cell responses can be effectively suppressed through MHC class I tetramer–mediated purging of the naive CD8+ T cell repertoire. Importantly, the development of GVHD occurs unimpeded upon suppression of the immunodominant MiHAg-specific T-cell response. These data indicate that antigen-specific graft engineering is feasible, but that parameters other than immunodominance may be required to select T-cell specificities that are targeted for removal.
Collapse
Affiliation(s)
- Moniek A de Witte
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
35
|
Dror N, Alter-Koltunoff M, Azriel A, Amariglio N, Jacob-Hirsch J, Zeligson S, Morgenstern A, Tamura T, Hauser H, Rechavi G, Ozato K, Levi BZ. Identification of IRF-8 and IRF-1 target genes in activated macrophages. Mol Immunol 2006; 44:338-46. [PMID: 16597464 DOI: 10.1016/j.molimm.2006.02.026] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 02/26/2006] [Accepted: 02/26/2006] [Indexed: 12/12/2022]
Abstract
Interferon regulatory factor 1 (IRF-1) and IRF-8, also known as interferon consensus sequence binding protein (ICSBP), are important regulators of macrophage differentiation and function. These factors exert their activities through the formation of heterocomplexes. As such, they are coactivators of various interferon-inducible genes in macrophages. To gain better insights into the involvement of these two transcription factors in the onset of the innate immune response and to identify their regulatory network in activated macrophages, DNA microarray was employed. Changes in the expression profile were analyzed in peritoneal macrophages from wild type mice and compared to IRF-1 and IRF-8 null mice, before and following 4 h exposure to IFN-gamma and LPS. The expression pattern of 265 genes was significantly changed (up/down) in peritoneal macrophages extracted from wild type mice following treatment with IFN-gamma and LPS, while no changes in the expression levels of these genes were observed in samples of the same cell-type from both IRF-1 and IRF-8 null mice. Among these putative target genes, numerous genes are involved in macrophage activity during inflammation. The expression profile of 10 of them was further examined by quantitative RT-PCR. In addition, the promoter regions of three of the identified genes were analyzed by reporter gene assay for the ability to respond to IRF-1 and IRF-8. Together, our results suggest that both IRF-1 and IRF-8 are involved in the transcriptional regulation of these genes. We therefore suggest a broader role for IRF-1 and IRF-8 in macrophages differentiation and maturation, being important inflammatory mediators.
Collapse
Affiliation(s)
- Natalie Dror
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kappel BJ, Pinilla-Ibarz J, Kochman AA, Eng JM, Hubbard VM, Leiner I, Pamer EG, Heller G, van den Brink MRM, Scheinberg DA. Remodeling specific immunity by use of MHC tetramers: demonstration in a graft-versus-host disease model. Blood 2006; 107:2045-51. [PMID: 16269613 PMCID: PMC1895712 DOI: 10.1182/blood-2005-07-2828] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Accepted: 10/19/2005] [Indexed: 02/01/2023] Open
Abstract
Major histocompatibility complex (MHC) molecules carrying selected peptides will bind specifically to their cognate T-cell receptor on individual clones of reactive T cells. Fluorescently labeled, tetrameric MHC-peptide complexes have been widely used to detect and quantitate antigen-specific T-cell populations via flow cytometry. We hypothesized that such MHC-peptide tetramers could also be used to selectively deplete unique reactive T-cell populations, while leaving the remaining T-cell repertoire and immune response intact. In this report, we successfully demonstrate that a tetramer-based depletion of T cells can be achieved in a murine model of allogeneic bone marrow transplantation. Depletion of a specific alloreactive population of donor splenocytes (< 0.5% of CD8+ T cells) prior to transplantation significantly decreased morbidity and mortality from graft-versus-host disease. There was no early regrowth of the antigen-specific T cells in the recipient and in vivo T-cell proliferation was greatly reduced as well. Survival was increased more than 3-fold over controls, yet the inherent antitumor activity of the transplant was retained. This method also provides the proof-of-concept for similar strategies to selectively remove other unwanted T-cell clones, which could result in novel therapies for certain autoimmune disorders, T-cell malignancies, and solid organ graft rejection.
Collapse
Affiliation(s)
- Barry J Kappel
- Department of Molecular Pharmacology, Memorial Sloan-Kettering Cancer Center, Howard 719, Mailbox 531, 1275 York Ave, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Millrain M, Scott D, Addey C, Dewchand H, Ellis P, Ehrmann I, Mitchell M, Burgoyne P, Simpson E, Dyson J. Identification of the immunodominant HY H2-D(k) epitope and evaluation of the role of direct and indirect antigen presentation in HY responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2005; 175:7209-17. [PMID: 16301625 DOI: 10.4049/jimmunol.175.11.7209] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Minor histocompatibility Ags derive from self-proteins and provoke allograft rejection and graft-vs-host disease in MHC-matched donor-recipient combinations. In this study, we define the HYD(k) epitope of the HY minor histocompatibility Ag as the 8mer peptide RRLRKTLL derived from the Smcy gene. Using HY tetramers, the response to this peptide was found to be immunodominant among the four characterized MHC class I-restricted HY epitopes (HYD(k)Smcy (defined here), HYK(k)Smcy, HYD(b)Uty, and HYD(b)Smcy). Indirect presentation stimulated a robust primary HYD(k)Smcy response. Indirect presentation and priming of HY-specific CD8+ T cells is also operative in the presence of a full MHC mismatch. To determine whether the indirect route of Ag presentation is required for HY priming, female parent into F1 (H2bxk) female recipient bone marrow chimeras were immunized with male cells of the other parental haplotype, limiting presentation to the direct pathway. The dominant H2b HY response (HYD(b)Uty) was dependent on indirect presentation. However, the dominant H2k HY response (HYD(k)Smcy) could be stimulated efficiently by the direct pathway. In contrast, secondary expansion of both HYD(k)Smcy and HYD(b)Uty-specific CD8+ T cells was effective only when Ag was presented by the direct route. Transgenic overproduction of Smcy mRNA within the immunizing cells resulted in a corresponding increase in the HYD(k)Smcy, HYD(b)Smcy, and HYK(k)Smcy-specific CD8+ T cell responses when presented via the direct pathway but did not enhance indirect presentation demonstrating the independent regulation of MHC class I-peptide occupancy in the two Ag-processing pathways.
Collapse
Affiliation(s)
- Maggie Millrain
- Transplantation Biology Group, Department of Immunology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Dalheimer SL, Richards DM, Mueller DL. Sharing of class I MHC molecules between donor and host promotes the infiltration of allografts by mHAg-reactive CD8 T cells. Am J Transplant 2005; 5:832-8. [PMID: 15760409 DOI: 10.1111/j.1600-6143.2005.00752.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Indirect recognition of minor histocompatibility antigens (mHAg) and/or MHC-encoded allopeptides is an important barrier to long-term allograft acceptance following solid organ transplantation. Efficient priming of CD8+ T cells can occur after allotransplantation as a consequence of cross-presentation of donor-derived proteins by the graft recipient's APC. Consistent with this, draining lymph node clonal expansion of OVA-reactive OT-I CD8+ T cells following placement of OVA-transgenic skin grafts did not depend on graft expression of K(b). However, OT-I T cells did accumulate in OVA-transgenic skin grafts most efficiently only when both the donor and host expressed K(b). OT-I infiltration of (B6-OVA x BALB/c)F1 grafts in B6 recipients was not suppressed by graft expression of H-2d. Furthermore, B6 animals transplanted with both B6-OVA and BALB/c-OVA skin had more OT-I T cells infiltrating their B6-OVA MHC-matched graft. Therefore, class I MHC matching between donor and host may not always favor an avoidance of alloreactivity within the graft tissue.
Collapse
Affiliation(s)
- Stacy L Dalheimer
- Department of Medicine and Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | | |
Collapse
|
39
|
Roy-Proulx G, Baron C, Perreault C. CD8 T-cell ability to exert immunodomination correlates with T-cell receptor: Epitope association rate. Biol Blood Marrow Transplant 2005; 11:260-71. [PMID: 15812391 DOI: 10.1016/j.bbmt.2004.12.334] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
When presented alone, H7 a and HY antigens elicit CD8 T-cell responses of similar amplitude, but H7 a totally abrogates the response to HY when both antigens are presented on the same antigen-presenting cell. We found that H7a- and HY-specific T-cell precursors had similar frequencies in nonimmune mice and expressed similar levels of CD5. The H7a -specific CD8 T-cell repertoire harvested at the time of primary response showed highly restricted T-cell receptor (TCR) diversity. Furthermore, T cells specific for H7a and HY expressed equivalent levels of CD8 and TCR and displayed similar tetramer decay rates. The key difference was that anti-H7a T cells exhibited a much more rapid TCR:epitope on-rate than anti-HY T cells. Coupled with evidence that primed CD8 T cells limit the duration of antigen presentation by killing or inactivating antigen-presenting cells, our data support a novel and simple model for immunodomination: the main feature of T cells that exert immunodomination is that, compared with other T cells, they are functionally primed after a shorter duration of antigen presentation.
Collapse
Affiliation(s)
- Guillaume Roy-Proulx
- Institute of Research in Immunology and Cancer, University of Montreal, Montreal, Quebec, Canada
| | | | | |
Collapse
|
40
|
Higuchi T, Maruyama T, Jaramillo A, Mohanakumar T. Induction of obliterative airway disease in murine tracheal allografts by CD8+ CTLs recognizing a single minor histocompatibility antigen. THE JOURNAL OF IMMUNOLOGY 2005; 174:1871-8. [PMID: 15699113 DOI: 10.4049/jimmunol.174.4.1871] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of minor histocompatibility Ag (mHAg)-specific CD8+ CTLs in the pathogenesis of chronic lung allograft rejection (bronchiolitis obliterans syndrome) remains to be elucidated. Thus, the goal of this study was to define the role of a single mHAg mismatch at the polymorphic H13 allele in the development of obliterative airway disease (OAD) after murine heterotopic tracheal transplantation. The H13a and H13b alleles encode for the SSVVGVWYL (SVL9) and SSVIGVWYL (SIL9) mHAgs, respectively, presented in the context of the H2Db MHC class I molecule. Toward this, C56BL/10SnJ (H13a) tracheal allografts were transplanted into congenic B10.CE-H13b Aw(30NX)/Sn (H13b) recipients. The allografts were harvested at 30, 60, and 90 days after transplantation, and OAD lesions (epithelial damage, cellular infiltration, and luminal fibrosis) were confirmed histologically. Selected groups of mice were immunized (s.c.) or tolerized (i.v.) with the SVL9 peptide before transplantation. This single mHAg mismatch induced the development of OAD within 90 days. SVL9 immunization significantly accelerated the kinetics of the OAD lesions. In contrast, SVL9 tolerization completely abrogated the development of OAD. This was correlated with a complete abrogation of H13a-specific CD8+ CTL responses with a significant reduction in the frequency of IFN-gamma-producing CTLs and the activation of TGF-beta-producing CD8+ T cells. In conclusion, a single mHAg mismatch can induce the development of OAD. These data also suggest that mHAg-reactive CD8+ CTLs may play an important role in the pathogenesis of chronic lung allograft rejection in humans.
Collapse
MESH Headings
- Animals
- Antigen Presentation/immunology
- Bronchiolitis Obliterans/immunology
- Bronchiolitis Obliterans/pathology
- Cytotoxicity, Immunologic/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Graft Enhancement, Immunologic/methods
- Graft Survival/immunology
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred A
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Minor Histocompatibility Antigens/administration & dosage
- Minor Histocompatibility Antigens/immunology
- Minor Histocompatibility Antigens/metabolism
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Skin Transplantation/immunology
- Skin Transplantation/pathology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Trachea/transplantation
- Transforming Growth Factor beta/biosynthesis
- Transplantation Tolerance/immunology
- Transplantation, Heterotopic/immunology
- Transplantation, Heterotopic/pathology
Collapse
Affiliation(s)
- Toru Higuchi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
41
|
Minor Histocompatibility Antigens: Molecular targets for immunomodulation in tissue transplantation and tumor therapy. ACTA ACUST UNITED AC 2005. [DOI: 10.1016/j.cair.2004.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
|
43
|
Regunathan J, Chen Y, Wang D, Malarkannan S. NKG2D receptor-mediated NK cell function is regulated by inhibitory Ly49 receptors. Blood 2005; 105:233-40. [PMID: 15328154 PMCID: PMC3889208 DOI: 10.1182/blood-2004-03-1075] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interaction of the activating ligand H60 with NKG2D receptor constitutes a major stimulatory pathway for natural killer (NK) cells. The influence of inhibitory Ly49 receptors on NKG2D-mediated activation is not clearly understood. Here we show that the magnitude of NKG2D-mediated cytotoxicity is directly proportional to both the levels of H60 and the nature of major histocompatibility complex (MHC) class I molecules expressed on the target cells. The expression levels of H60 on the target cells determined the extent to which the inhibition by Ly49C/I receptors can be overridden. In contrast, even a higher expression of H60 molecule on the target cells failed to overcome the inhibition mediated by Ly49A/G receptors. Also, the level of interferon-gamma (IFN-gamma) and granulocyte-macrophage colony-stimulating factor (GM-CSF) generated by NK cells through anti-NKG2D monoclonal antibody (mAb)-mediated activation is significantly reduced by the presence of immobilized anti-Ly49A/G mAbs. Thus, NKG2D-mediated cytotoxicity and cytokine secretion results from the fine balance between activating and inhibitory receptors, thereby defining the NK cell-mediated immune responses.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Antibodies, Monoclonal/immunology
- Antigens, Ly/immunology
- Antigens, Ly/metabolism
- Cell Nucleus/metabolism
- Cells, Cultured
- Cytotoxicity, Immunologic/immunology
- Down-Regulation
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Histocompatibility Antigen H-2D
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lectins, C-Type
- Ligands
- Mice
- Minor Histocompatibility Antigens/metabolism
- NF-kappa B/metabolism
- NK Cell Lectin-Like Receptor Subfamily A
- NK Cell Lectin-Like Receptor Subfamily K
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, NK Cell Lectin-Like
- Receptors, Natural Killer Cell
- Signal Transduction
Collapse
Affiliation(s)
- Jeyarani Regunathan
- Blood Research Institute, Blood Center of Southeastern Wisconsin, Milwaukee 53226, USA
| | | | | | | |
Collapse
|
44
|
Araki J, Ohashi J, Muramatsu M. Application of discordant sib-pair linkage analysis for mapping minor histocompatibility antigen loci in a novel graft-vs-host-disease model. TISSUE ANTIGENS 2004; 64:243-50. [PMID: 15304004 DOI: 10.1111/j.1399-0039.2004.00268.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Graft-vs-host disease (GVHD) is an adverse effect of allogenic bone marrow transplantation. Although a major cause of GVHD following bone marrow transplantation is incompatibility of major histocompatibility antigen (human leukocyte antigen, HLA) in donor-recipient pairs, the incompatibility of minor histocompatibility antigen (mHa) is known as another cause, especially in HLA-matched donor-recipient pairs. In 1998, Lunetta and Rogus proposed the use of discordant sib-pair (DSP) linkage analysis for detecting mHa and calculated the statistical power using the GVHD model, assuming single mHa locus with multiple alleles. Recently, we proposed a different GVHD model, assuming multiple mHa loci with two alleles (biallelic), considering the single-nucleotide polymorphisms. When the effect of each mHa locus on the occurrence of GVHD is independent, the possible triangle for DSP proposed by Lunetta and Rogus is not optimum, but a new possible triangle, named here as GVHD region, is needed. We evaluated, based on Monte Carlo simulation, the test criteria [log of odds (lod) score cutoffs] and power of DSP using the GVHD region for various parameter sets. The GVHD region showed a higher power than the DSP and entire regions in plausible situations. Our results suggest that the application of GVHD region to DSP is effective for the screening of mHa loci.
Collapse
Affiliation(s)
- J Araki
- Department of Molecular Epidemiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
| | | | | |
Collapse
|
45
|
Hamerman JA, Ogasawara K, Lanier LL. Cutting edge: Toll-like receptor signaling in macrophages induces ligands for the NKG2D receptor. THE JOURNAL OF IMMUNOLOGY 2004; 172:2001-5. [PMID: 14764662 DOI: 10.4049/jimmunol.172.4.2001] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Macrophages recognize the presence of infection by using the Toll-like receptor (TLR) family of proteins that detect ligands on bacterial, viral, and fungal pathogens. We show that murine macrophages stimulated with pathogen products known to signal through TLRs express ligands for the NKG2D receptor, found on NK cells, activated CD8(+) T cells and activated macrophages. TLR signaling, through the MyD88 adaptor, up-regulates transcription of the retinoic acid early inducible-1 (RAE-1) family of NKG2D ligands, but not H-60 or murine UL16-binding protein-like transcript-1. RAE-1 proteins are found on the surface of activated, but not resting, macrophages and can be detected by NKG2D on NK cells resulting in down-regulation of this receptor both in vitro and in vivo. RAE-1-NKG2D interactions provide a mechanism by which NK cells and infected macrophages communicate directly during an innate immune response to infection.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antigens, Differentiation/genetics
- Antigens, Differentiation/physiology
- Cells, Cultured
- Escherichia coli/immunology
- Ligands
- Lipopolysaccharides/pharmacology
- Listeria monocytogenes/immunology
- Macrophage Activation
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/microbiology
- Membrane Glycoproteins/physiology
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Myeloid Differentiation Factor 88
- NK Cell Lectin-Like Receptor Subfamily K
- Poly I-C/pharmacology
- RNA, Messenger/biosynthesis
- Receptors, Cell Surface/physiology
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Receptors, Natural Killer Cell
- Signal Transduction/immunology
- Toll-Like Receptors
- Tretinoin/metabolism
- Zymosan/pharmacology
Collapse
Affiliation(s)
- Jessica A Hamerman
- Department of Microbiology and Immunology, and Cancer Research Institute, University of California, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
46
|
Yoshimura Y, Yadav R, Christianson GJ, Ajayi WU, Roopenian DC, Joyce S. Duration of Alloantigen Presentation and Avidity of T Cell Antigen Recognition Correlate with Immunodominance of CTL Response to Minor Histocompatibility Antigens. THE JOURNAL OF IMMUNOLOGY 2004; 172:6666-74. [PMID: 15153482 DOI: 10.4049/jimmunol.172.11.6666] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CD8 T lymphocytes (CTL) responsive to immunodominant minor histocompatibility (minor H) Ags are thought to play a disproportionate role in allograft rejection in MHC-identical solid and bone marrow transplant settings. Although many studies have addressed the mechanisms underlying immunodominance in models of infectious diseases, cancer immunotherapy, and allograft immunity, key issues regarding the molecular basis of immunodominance remain poorly understood. In this study, we exploit the minor H Ag system to understand the relationship of the various biochemical parameters of Ag presentation and recognition to immunodominance. We show that the duration of individual minor H Ag presentation and the avidity of T cell Ag recognition influence the magnitude and, hence, the immunodominance of the CTL response to minor H Ags. These properties of CTL Ag presentation and recognition that contribute to immunodominance have implications not only for tissue transplantation, but also for autoimmunity and tumor vaccine design.
Collapse
Affiliation(s)
- Yoshitaka Yoshimura
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
NK cells are crucial components of the innate immune system, capable of directly eliminating infected or tumorigenic cells and regulating down-stream adaptive immune responses. Unlike T cells, where the key recognition event driving activation is mediated by the unique T cell receptor (TCR) expressed on a given cell, NK cells express multiple activating and inhibitory cell-surface receptors (NKRs), often with overlapping ligand specificities. NKRs display two ectodomain structural homologies, either immunoglobulin- or C-type lectin-like (CTLD). The CTLD immunoreceptor NKG2D is found on NK cells but is also widely expressed on T cells and other immune system cells, providing stimulatory or co-stimulatory signals. NKG2D drives target cell killing following engagement of diverse, conditionally expressed MHC class I-like protein ligands whose expression can signal cellular distress due to infection or transformation. The symmetric, homodimeric receptor interacts with its asymmetric, monomeric ligands in similar 2:1 complexes, with an equivalent surface on each NKG2D monomer binding extensively and intimately to distinct, structurally divergent surfaces on the ligands. Thus, NKG2D ligand-binding site recognition is highly degenerate, further demonstrated by NKG2D's ability to simultaneously accommodate multiple non-conservative allelic or isoform substitutions in the ligands. In TCRs, "induced-fit" recognition explains cross-reactivity, but structural, computational, thermodynamic and kinetic analyses of multiple NKG2D-ligand pairs show that rather than classical "induced-fit" binding, NKG2D degeneracy is achieved using distinct interaction mechanisms at each rigid interface: recognition degeneracy by "rigid adaptation." While likely forming similar complexes with their ligand (HLA-E), other NKG2x NKR family members do not require such recognition degeneracy.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Histocompatibility Antigens Class I/immunology
- Humans
- Killer Cells, Natural/immunology
- Ligands
- Models, Molecular
- Molecular Sequence Data
- NK Cell Lectin-Like Receptor Subfamily K
- Phylogeny
- Protein Binding
- Protein Structure, Tertiary
- Receptors, Antigen, T-Cell/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Natural Killer Cell
- Sequence Homology, Amino Acid
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Roland K Strong
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | |
Collapse
|
48
|
Bezouška K. Carbohydrate and Non-Carbohydrate Ligands for the C-Type Lectin-Like Receptors of Natural Killer Cells. A Review. ACTA ACUST UNITED AC 2004. [DOI: 10.1135/cccc20040535] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The superfamily of C-type animal lectins is defined by a sequence motif of the carbohydrate- recognition domains (CRDs) and comprises seven groups of molecules. The soluble proteins are group I proteoglycans, group III collectins, and group VII containing the isolated CRDs. Type I membrane proteins include group IV selectins and group VI macrophage receptors and related molecules. Type II membrane proteins are group II hepatic lectins and group V natural killer cell receptors. The latter group has recently attracted considerable attention of the biomedical community. These receptors are arranged at the surface of lymphocytes as homo- or heterodimers composed of two polypeptides consisting of N-terminal peptide tails responsible for signaling, transmembrane domain, neck regions of varying length, and C-terminal lectin-like domains (CTLDs). Since this group is evolutionarily most distant from the rest of C-type animal lectins, the sequence of the C-terminal ligand-binding domain has diversified to accommodate other ligands than calcium or carbohydrates. These domains are referred to as natural killer domains (NKDs) forming a large percentage of CTLDs in vertebrates. Here are summarized the data indicating that calcium, carbohydrates, peptides, and large proteins such as major histocompatibility complex (MHC) class I can all be ligands for NKDs. The wide range of ligands that can be recognized by NKDs includes some new, unexpected compounds such as signal peptide-derived fragments, heat shock proteins, or oxidized lipids. The biological importance of this extended range of recognition abilities is also discussed. A review with 134 references.
Collapse
|
49
|
Schwab SR, Li KC, Kang C, Shastri N. Constitutive display of cryptic translation products by MHC class I molecules. Science 2003; 301:1367-71. [PMID: 12958358 DOI: 10.1126/science.1085650] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Major histocompatibility complex (MHC) class I molecules display tens of thousands of peptides on the cell surface, derived from virtually all endogenous proteins, for inspection by cytotoxic T cells (CTLs). We show that, in normal mouse cells, MHC I molecules present a peptide encoded in the 3' "untranslated" region. Despite its rarity, the peptide elicits CTL responses and induces self-tolerance, establishing that immune surveillance extends well beyond conventional polypeptides. Furthermore, translation of this cryptic peptide occurs by a previously unknown mechanism that decodes the CUG initiation codon as leucine rather than the canonical methionine.
Collapse
Affiliation(s)
- Susan R Schwab
- Division of Immunology, Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720-3200, USA
| | | | | | | |
Collapse
|
50
|
Yang J, Jaramillo A, Liu W, Olack B, Yoshimura Y, Joyce S, Kaleem Z, Mohanakumar T. Chronic rejection of murine cardiac allografts discordant at the H13 minor histocompatibility antigen correlates with the generation of the H13-specific CD8+ cytotoxic T cells. Transplantation 2003; 76:84-91. [PMID: 12865791 DOI: 10.1097/01.tp.0000072013.21336.64] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Minor histocompatibility antigen (mHag) discordances have been shown to play a critical role in graft-versus-host disease after bone marrow transplantation. However, the role of mHag in rejection of solid-organ allografts remains unknown. Therefore, the goal of this study was to define the role of a single mHag discordance derived from the polymorphic H13 allele in the development of cardiac allograft rejection in mice. The H13a and H13b alleles encode for the SSVVGVWYL (SVL9) and SSVIGVWYL (SIL9) mHag bound to the H2Db molecule, respectively. METHODS C56BL/10SnJ (H13a) cardiac allografts were transplanted into congenic B10.CE-H13b Aw(30NX)/Sn (H13b) mice. Allograft function was monitored daily and rejection was defined by cessation of heart beat. Rejection was confirmed histologically. The phenotypic and functional characteristics of the graft-infiltrating cells were analyzed by in situ and in vitro staining with H13a-specific tetramers and by chromium-51 (51Cr)-release assay, respectively. RESULTS Sixty-five percent of H13-incompatible allografts were rejected in 37.0+/-14.5 days. Sixty-eight percent of the H13a allografts transplanted into H13a-sensitized mice were rejected earlier, in 27.6+/-15.9 days. Rejected allografts showed histopathologic signs of chronic rejection with diffuse mononuclear cell infiltration, concentric intimal hyperplasia, and fibrosis. Both CD8+ (87%) and CD4+ (13%) T cells were observed in rejected allografts. In addition, 60% of the graft-infiltrating CD8+ T cells recognized a H2Db/SVL9 tetramer. Graft-infiltrating CD8+ T cells showed a significant H2Db-restricted, SVL9-specific cytotoxic activity. CONCLUSIONS A single mHag discordance, as demonstrated with H13 disparity, results in the pathogenesis of chronic rejection of major histocompatibility complex-matched vascularized solid-organ allograft.
Collapse
Affiliation(s)
- Junbao Yang
- Department of Surgery, Washington University School of Medicine, St Louis, MO 63110-1093, USA
| | | | | | | | | | | | | | | |
Collapse
|