1
|
Di Zazzo A, Lee SM, Sung J, Niutta M, Coassin M, Mashaghi A, Inomata T. Variable Responses to Corneal Grafts: Insights from Immunology and Systems Biology. J Clin Med 2020; 9:E586. [PMID: 32098130 PMCID: PMC7074162 DOI: 10.3390/jcm9020586] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
Corneal grafts interact with their hosts via complex immunobiological processes that sometimes lead to graft failure. Prediction of graft failure is often a tedious task due to the genetic and nongenetic heterogeneity of patients. As in other areas of medicine, a reliable prediction method would impact therapeutic decision-making in corneal transplantation. Valuable insights into the clinically observed heterogeneity of host responses to corneal grafts have emerged from multidisciplinary approaches, including genomics analyses, mechanical studies, immunobiology, and theoretical modeling. Here, we review the emerging concepts, tools, and new biomarkers that may allow for the prediction of graft survival.
Collapse
Affiliation(s)
- Antonio Di Zazzo
- Ophthalmology Complex Operative Unit, Campus Bio Medico University, 00128 Rome, Italy; (A.D.Z.); (M.N.); (M.C.)
| | - Sang-Mok Lee
- Department of Ophthalmology, Catholic Kwandong University College of Medicine, Gangneung-si, Gangwon-do 25601, Korea;
- Department of Cornea, External Disease & Refractive Surgery, HanGil Eye Hospital, Incheon 21388, Korea
| | - Jaemyoung Sung
- University of South Florida, Morsani College of Medicine, Tampa, FL 33612, USA;
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo 1130033, Japan
| | - Matteo Niutta
- Ophthalmology Complex Operative Unit, Campus Bio Medico University, 00128 Rome, Italy; (A.D.Z.); (M.N.); (M.C.)
| | - Marco Coassin
- Ophthalmology Complex Operative Unit, Campus Bio Medico University, 00128 Rome, Italy; (A.D.Z.); (M.N.); (M.C.)
| | - Alireza Mashaghi
- Systems Biomedicine and Pharmacology Division, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo 1130033, Japan
- Department of Strategic Operating Room Management and Improvement, Juntendo University Faculty of Medicine, Tokyo 1130033, Japan
- Department of Hospital Administration, Juntendo University Faculty of Medicine, Tokyo 1130033, Japan
| |
Collapse
|
2
|
Farkas AM, Baranyi U, Böhmig GA, Unger L, Hopf S, Wahrmann M, Regele H, Mahr B, Schwarz C, Hock K, Pilat N, Kristo I, Mraz J, Lupinek C, Thalhamer J, Bond G, Kuessel L, Wlodek E, Martin J, Clatworthy M, Pettigrew G, Valenta R, Wekerle T. Allograft rejection is associated with development of functional IgE specific for donor MHC antigens. J Allergy Clin Immunol 2019; 143:335-345.e12. [PMID: 30009843 DOI: 10.1016/j.jaci.2018.06.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 06/07/2018] [Accepted: 06/14/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Donor-specific antibodies of the IgG isotype are measured routinely for diagnostic purposes in renal transplant recipients and are associated with antibody-mediated rejection and long-term graft loss. OBJECTIVE This study aimed to investigate whether MHC-specific antibodies of the IgE isotype are induced during allograft rejection. METHODS Anti-MHC/HLA IgE levels were measured in sera of mice grafted with skin or heart transplants from various donor strains and in sera of kidney transplant patients with high levels of HLA IgG. Mediator release was triggered in vitro by stimulating basophils that were coated with murine or human IgE-positive serum, respectively, with specific recombinant MHC/HLA antigens. Kidney tissue samples obtained from organ donors were analyzed by using flow cytometry for cells expressing the high-affinity receptor for IgE (FcεRI). RESULTS Donor MHC class I- and MHC class II-specific IgE was found on acute rejection of skin and heart grafts in several murine strain combinations, as well as during chronic antibody-mediated heart graft rejection. Anti-HLA IgE, including donor HLA class I and II specificities, was identified in a group of sensitized transplant recipients. Murine and human anti-MHC/HLA IgE triggered mediator release in coated basophils on stimulation with specific MHC/HLA antigens. HLA-specific IgE was not linked to atopy, and allergen-specific IgE present in allergic patients did not cross-react with HLA antigens. FcεRI+ cells were found in the human renal cortex and medulla and provide targets for HLA-specific IgE. CONCLUSION These results demonstrate that MHC/HLA-specific IgE develops during an alloresponse and is functional in mediating effector mechanisms.
Collapse
Affiliation(s)
- Andreas M Farkas
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Ulrike Baranyi
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria; Cardiac Surgery Laboratory, Medical University of Vienna, Vienna, Austria
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lukas Unger
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefan Hopf
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Markus Wahrmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Heinz Regele
- Institute of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Benedikt Mahr
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Schwarz
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Karin Hock
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Nina Pilat
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Ivan Kristo
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Jasmin Mraz
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Christian Lupinek
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Physiology and Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Josef Thalhamer
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - Gregor Bond
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lorenz Kuessel
- Department for Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Elizabeth Wlodek
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Jack Martin
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Menna Clatworthy
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gavin Pettigrew
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Physiology and Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Thomas Wekerle
- Section of Transplantation Immunology, Department of Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Genome-Wide Analysis in Swine Associates Corneal Graft Rejection with Donor-Recipient Mismatches in Three Novel Histocompatibility Regions and One Locus Homologous to the Mouse H-3 Locus. PLoS One 2016; 11:e0152155. [PMID: 27010211 PMCID: PMC4806994 DOI: 10.1371/journal.pone.0152155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/09/2016] [Indexed: 11/19/2022] Open
Abstract
In rodents, immune responses to minor histocompatibility antigens are the most important drivers of corneal graft rejection. However, this has not been confirmed in humans or in a large animal model and the genetic loci are poorly characterised, even in mice. The gene sequence data now available for a range of relevant species permits the use of genome-wide association (GWA) techniques to identify minor antigens associated with transplant rejection. We have used this technique in a pre-clinical model of corneal transplantation in semi-inbred NIH minipigs and Babraham swine to search for novel minor histocompatibility loci and to determine whether rodent findings have wider applicability. DNA from a cohort of MHC-matched and MHC-mismatched donors and recipients was analysed for single nucleotide polymorphisms (SNPs). The level of SNP homozygosity for each line was assessed. Genome-wide analysis of the association of SNP disparities with rejection was performed using log-likelihood ratios. Four genomic blocks containing four or more SNPs significantly linked to rejection were identified (on chromosomes 1, 4, 6 and 9), none at the location of the MHC. One block of 36 SNPs spanned a region that exhibits conservation of synteny with the mouse H-3 histocompatibility locus and contains the pig homologue of the mouse Zfp106 gene, which encodes peptide epitopes known to mediate corneal graft rejection. The other three regions are novel minor histocompatibility loci. The results suggest that rejection can be predicted from SNP analysis prior to transplant in this model and that a similar GWA analysis is merited in humans.
Collapse
|
4
|
Govender L, Pascual M, Golshayan D. Potential and limitations of regulatory T-cell therapy in solid organ transplantation. Expert Rev Clin Immunol 2014; 10:1197-212. [PMID: 25073810 DOI: 10.1586/1744666x.2014.943191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Over the past few years, the therapeutic potential of Treg has been highlighted in the field of autoimmune diseases and after allogeneic transplantation. The first hurdle for the therapeutic use of Treg is their insufficient numbers in non-manipulated individuals, in particular when facing strong immune activation and expanding effector cells, such as in response to an allograft. Here we review current approaches being explored for Treg expansion in the perspective of clinical therapeutic protocols. We describe different Treg subsets that could be suitable for clinical application, as well as discuss factors such as the required dose of Treg, their antigen-specificity and in vivo stability, that have to be considered for optimal Treg-based immunotherapy in transplantation. Since Treg may not be sufficient as stand-alone therapy for solid organ transplantation in humans, we draw attention to possible hurdles and combination therapy with immunomodulatory drugs that could possibly improve the in vivo efficacy of Treg.
Collapse
Affiliation(s)
- Lerisa Govender
- Departments of Medicine and Surgery, Transplantation Centre and Transplantation Immunopathology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, 1011 Lausanne, Switzerland
| | | | | |
Collapse
|
5
|
Sabet-Baktach M, Eggenhofer E, Rovira J, Renner P, Lantow M, Farkas SA, Malaisé M, Edtinger K, Shaotang Z, Koehl GE, Dahlke MH, Schlitt HJ, Geissler EK, Kroemer A. Double deficiency for RORγt and T-bet drives Th2-mediated allograft rejection in mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:4440-6. [PMID: 24058178 DOI: 10.4049/jimmunol.1301741] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although Th1, Th2, and Th17 cells are thought to be major effector cells in adaptive alloimmune responses, their respective contribution to allograft rejection remains unclear. To precisely address this, we used mice genetically modified for the Th1 and Th17 hallmark transcription factors T-bet and RORγt, respectively, which allowed us to study the alloreactive role of each subset in an experimental transplant setting. We found that in a fully mismatched heterotopic mouse heart transplantation model, T cells deficient for T-bet (prone to Th17 differentiation) versus RORγt (prone to Th1 differentiation) rejected allografts at a more accelerated rate, indicating a predominance of Th17- over Th1-driven alloimmunity. Importantly, T cells doubly deficient for both T-bet and RORγt differentiated into alloreactive GATA-3-expressing Th2 cells, which promptly induced allograft rejection characterized by a Th2-type intragraft expression profile and eosinophilic infiltration. Mechanistically, Th2-mediated allograft rejection was contingent on IL-4, as its neutralization significantly prolonged allograft survival by reducing intragraft expression of Th2 effector molecules and eosinophilic allograft infiltration. Moreover, under IL-4 neutralizing conditions, alloreactive double-deficient T cells upregulated Eomesodermin (Eomes) and IFN-γ, but not GATA-3. Thus, in the absence of T-bet and RORγt, Eomes may salvage Th1-mediated alloimmunity that underlies IL-4 neutralization-resistant allograft rejection. We summarize that, whereas Th17 cells predictably promote allograft rejection, IL-4-producing GATA-3(+) Th2 cells, which are generally thought to protect allogeneic transplants, may actually be potent facilitators of organ transplant rejection in the absence of T-bet and RORγt. Moreover, Eomes may rescue Th1-mediated allograft rejection in the absence of IL-4, T-bet, and RORγt.
Collapse
Affiliation(s)
- Manije Sabet-Baktach
- Department of Surgery, University Hospital Regensburg, University of Regensburg, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Xiong L, Yang L. Effects of alkaloid sinomenine on levels of IFN-γ, IL-1β, TNF-α and IL-6 in a rat renal allograft model. Immunotherapy 2013; 4:785-91. [PMID: 22947007 DOI: 10.2217/imt.12.80] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS To evaluate the immunosuppressive efficacy of alkaloid sinomenine (SIN) and the synergistic effects in combination with cyclosporin A (CsA) in acute rejection after rat renal allograft. MATERIALS & METHODS Animals were treated with saline in group 1, SIN (30 mg/kg/d) in group 2, CsA (2.5 mg/kg/d) in group 3 and SIN (30 mg/kg/d) + CsA(2.5 mg/kg/d) in group 4. Another 12 syngeneic renal transplantation animals were treated with saline as control. Survival time is observed. The levels of serum creatinine (Scr) and blood urea nitrogen (Bun) were detected; the secretion of IFN-γ, IL-1β, TNF-α and IL-6 were detected by ELISA. The kidneys were fixed to perform histological staining. RESULTS The mean survival time was 8.00 ± 2.10 days in group 1, 10.67 ± 1.21 days in group 2, 11.00 ± 1.41 days in group 3 and 19.67 ± 2.80 days in group 4, while all the recipients survived more than 180 days in the control group. The 24-h urinary volume and urinary time of the other three groups were increased significantly compared with group 1. The levels of Scr and Bun, levels of IFN-γ, IL-1β, TNF-α and IL-6 were significantly higher in group 1 than that in the other three groups; there were significant differences between group 4 and group 2 or 3. CONCLUSION Our study showed that SIN had immunosupression effects in rat renal allograft models, it also had a synergistic effect in combination with CsA, which provided a new immunosuppressant for clinical application.
Collapse
Affiliation(s)
- Lie Xiong
- Department of Urology, The Second Affliated Hospital of Xiangya, 139# Middle Road of Renmin, Changsha 410000, Hunan, China
| | | |
Collapse
|
7
|
Araki R, Uda M, Hoki Y, Sunayama M, Nakamura M, Ando S, Sugiura M, Ideno H, Shimada A, Nifuji A, Abe M. Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells. Nature 2013; 494:100-4. [PMID: 23302801 DOI: 10.1038/nature11807] [Citation(s) in RCA: 359] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 11/22/2012] [Indexed: 02/07/2023]
Abstract
The advantages of using induced pluripotent stem cells (iPSCs) instead of embryonic stem (ES) cells in regenerative medicine centre around circumventing concerns about the ethics of using ES cells and the likelihood of immune rejection of ES-cell-derived tissues. However, partial reprogramming and genetic instabilities in iPSCs could elicit immune responses in transplant recipients even when iPSC-derived differentiated cells are transplanted. iPSCs are first differentiated into specific types of cells in vitro for subsequent transplantation. Although model transplantation experiments have been conducted using various iPSC-derived differentiated tissues and immune rejections have not been observed, careful investigation of the immunogenicity of iPSC-derived tissue is becoming increasingly critical, especially as this has not been the focus of most studies done so far. A recent study reported immunogenicity of iPSC- but not ES-cell-derived teratomas and implicated several causative genes. Nevertheless, some controversy has arisen regarding these findings. Here we examine the immunogenicity of differentiated skin and bone marrow tissues derived from mouse iPSCs. To ensure optimal comparison of iPSCs and ES cells, we established ten integration-free iPSC and seven ES-cell lines using an inbred mouse strain, C57BL/6. We observed no differences in the rate of success of transplantation when skin and bone marrow cells derived from iPSCs were compared with ES-cell-derived tissues. Moreover, we observed limited or no immune responses, including T-cell infiltration, for tissues derived from either iPSCs or ES cells, and no increase in the expression of the immunogenicity-causing Zg16 and Hormad1 genes in regressing skin and teratoma tissues. Our findings suggest limited immunogenicity of transplanted cells differentiated from iPSCs and ES cells.
Collapse
Affiliation(s)
- Ryoko Araki
- Transcriptome Research Group, National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Zheng P, Yang Y, Li S, Li J, Gong W, Quan Z. A given number of effector T cells can only destroy a limited number of target cells in graft rejection. Transpl Immunol 2010; 23:111-116. [PMID: 20450975 DOI: 10.1016/j.trim.2010.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 04/09/2010] [Accepted: 04/26/2010] [Indexed: 02/08/2023]
Abstract
Anti-donor T cells mediate graft rejection and the frequency of anti-donor T cells directly correlates with transplant outcome. It has long been noted that long-term tolerance was occasionally observed in minor mismatched recipients, little is known about the mechanisms underlying this phenomenon. To quantitatively analyze the relationship between anti-donor T cells and target cells, long-term tolerant C57BL/6 mice were established by infusing 3 x 10(7) F1 splenocytes during the neonatal period. The removal of anti-donor T cells in these mice was demonstrated by unresponsiveness in mixed-lymphocyte reaction. A total of 2 x 10(7) or 5 x 10(7) syngenic naive cells were transferred into long-term tolerant mice; the dose of 5 x 10(7) syngenic cells destroyed chimerism and the skin grafts, while the dose of 2 x 10(7) syngenic cells led to loss of chimerism but the survival of the skin grafts. On Day 20 after the transfusion, a portion of 5 x 10(7) syngenic cells still remained in the mice, while no syngenic cells were detected in mice that received a total of 2 x 10(7) syngenic cells suggesting that these cells were completely exhausted. Syngenic CD4+ T cells proliferated and activated in both groups, while syngenic cells in the low-dose group were more susceptible to apoptosis than those in the high-dose group. Our results suggest that a given number of effector T cells could only kill a limited number of target cells. When that limit was reached, the T cells died. This novel concept not only provides a reasonable explanation for long-term tolerance in minor mismatched transplantation but also provides new insight into tolerance induction that depleting alloreactive T cells in recipients by donor cells or agents is a prerequisite for reconstitution of thymus by donor cells, the establishment of central tolerance is the key for successful tolerance induction.
Collapse
Affiliation(s)
- Peiguo Zheng
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | | | | | | | | | | |
Collapse
|
9
|
Issa F, Schiopu A, Wood KJ. Role of T cells in graft rejection and transplantation tolerance. Expert Rev Clin Immunol 2010; 6:155-69. [PMID: 20383898 DOI: 10.1586/eci.09.64] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Transplantation is the most effective treatment for end-stage organ failure, but organ survival is limited by immune rejection and the side effects of immunosuppressive regimens. T cells are central to the process of transplant rejection through allorecognition of foreign antigens leading to their activation, and the orchestration of an effector response that results in organ damage. Long-term transplant acceptance in the absence of immunosuppressive therapy remains the ultimate goal in the field of transplantation and many studies are exploring potential therapies. One promising cellular therapy is the use of regulatory T cells to induce a state of donor-specific tolerance to the transplant. This article first discusses the role of T cells in transplant rejection, with a focus on the mechanisms of allorecognition and the alloresponse. This is followed by a detailed review of the current progress in the field of regulatory T-cell therapy in transplantation and the translation of this therapy to the clinical setting.
Collapse
Affiliation(s)
- Fadi Issa
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | | | | |
Collapse
|
10
|
Benghiat FS, Charbonnier LM, Vokaer B, De Wilde V, Le Moine A. Interleukin 17-producing T helper cells in alloimmunity. Transplant Rev (Orlando) 2009; 23:11-8. [PMID: 19027613 DOI: 10.1016/j.trre.2008.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Interleukin (IL) 17 is a proinflammatory cytokine already known to play a defense role against microbes and a pathogenic role in a number of autoimmune diseases. Although IL-17 can be produced by a variety of cells including neutrophils, CD8+, NK, and gamma-delta T cells, the concept of IL-17-secreting CD4+ T helper cells (Th17), distinct from Th1 and Th2, recently emerged. Herein, we discuss arguments in favor of a Th17-mediated alternative pathway of allograft rejection based on clinical and experimental observations drawn from the literature. We also discuss the complex interplays among regulatory T cells and Th17 cells in the allogeneic context.
Collapse
|
11
|
Jacobsen EA, Taranova AG, Lee NA, Lee JJ. Eosinophils: singularly destructive effector cells or purveyors of immunoregulation? J Allergy Clin Immunol 2007; 119:1313-20. [PMID: 17481717 DOI: 10.1016/j.jaci.2007.03.043] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2007] [Revised: 03/28/2007] [Accepted: 03/28/2007] [Indexed: 01/21/2023]
Abstract
Eosinophils are granulocytes typically associated with immune responses to a limited number of specific insults, including helminth infection and exposure to various allergens. Moreover, the overwhelming consensus from the literature is that eosinophils evolved as uniquely destructive leukocytes with cytotoxic activities as an adaptation for host defense. However, recent studies now suggest that the parochial caricature of eosinophils as effector cells with nonspecific killing abilities that evolved as a host defense mechanism against large nonphagotizable parasites is incomplete. A new paradigm has emerged describing eosinophils as initial responders to cell death/tissue damage that are a part of remodeling/repair processes and, more importantly, significant contributors to localized innate and acquired immune responses as well as systemic adaptive immunity. Significantly, this new paradigm does not preclude roles for eosinophils in host defense leading to tissue damage but instead suggests the equal importance of eosinophil-associated regulatory mechanisms modulating local tissue immune responses. The goal of this review is to summarize the data in support of this new paradigm. In turn, we believe that this expanded role provides a probable explanation for the presence of eosinophils in diverse disease settings such as asthma, allergy, cancer, transplant rejection, gastrointestinal inflammation, and viral or helminth infection.
Collapse
Affiliation(s)
- Elizabeth A Jacobsen
- Division of Pulmonary Medicine, Department of Biocheistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, USA
| | | | | | | |
Collapse
|
12
|
Loss of Transgene following ex vivo Gene Transfer is Associated with a Dominant Th2 Response: Implications for Cutaneous Gene Therapy. Mol Ther 2007. [DOI: 10.1038/sj.mt.6300086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
13
|
Lu Z, Ghazizadeh S. Loss of transgene following ex vivo gene transfer is associated with a dominant Th2 response: implications for cutaneous gene therapy. Mol Ther 2007; 15:954-61. [PMID: 17356544 PMCID: PMC2877884 DOI: 10.1038/mt.sj.6300086] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Host responses to therapeutic gene products are potentially serious complications in cutaneous gene therapy. Controlling immune responses to the therapeutic antigen may therefore be critical for an effective therapy. Both ex vivo and in vivo gene transfer to epidermal stem cells has been shown to induce transgene-specific immune responses; however, whether the mechanism of immune activation is the same is not clear. In this study, we have characterized transgene-specific immune responses in an ex vivo model of epidermal gene transfer using green fluorescent protein as a model antigen and retrovirus-mediated gene delivery. Contrary to T helper (Th)1-type responses induced following in vivo gene transfer to epidermis, rejection of ex vivo-transduced keratinocytes was associated with Th2/eosinophilc responses. These responses were characterized by interleukin (IL)-4 and IL-5 production by T cells, a predominance of anti-green fluorescent protein IgG1 in serum, the presence of numerous eosinophils within rejected skin, and a lack of class I-restricted cytotoxic T lymphocyte response. Pretreatment of mice receiving ex vivo transduced keratinocytes with neutralizing anti-IL-5 antibody prevented eosinophil infiltration and prolonged survival of transduced epidermis. These data indicate a role for the Th2/eosinophilic pathway in rejection of ex vivo-transduced keratinocytes, suggesting different requirements for achieving tolerance for ex vivo and in vivo approaches to gene therapy.
Collapse
Affiliation(s)
- Zhenmei Lu
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Soosan Ghazizadeh
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Department of Oral Biology and Pathology, School of Dental Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
14
|
Surquin M, Le Moine A, Flamand V, Rombaut K, Demoor FX, Salmon I, Goldman M, Abramowicz D. IL-4 deficiency prevents eosinophilic rejection and uncovers a role for neutrophils in the rejection of MHC class II disparate skin grafts. Transplantation 2006; 80:1485-92. [PMID: 16340795 DOI: 10.1097/01.tp.0000176486.01697.3f] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acute rejection of MHC class II-disparate bm12 skin grafts by C57BL/6 recipient mice is characterized by massive graft infiltration by eosinophils, together with increased intragraft amounts of IL-4 and IL-5 mRNA. IL-5 blockade prevents the intragraft eosinophil infiltration and prolongs the survival of skin allografts. As the differentiation of T cell precursors into Th2 cells is largely driven by IL-4, we investigated the role of IL-4 in MHC class II-disparate allograft rejection. METHODS We performed skin grafts from MHC class II incompatible bm12 mice into wild-type C57BL/6 mice (IL-4) or C57BL/6 IL-4 deficient mice (IL-4). Graft survival, in vitro T cell reactivity, and histology were compared. RESULTS We observed that 50% of IL-4 mice rapidly rejected their bm12 allograft, whereas the other 50% retained their graft 60 days after transplantation. Histological examination of bm12 allografts retained by IL-4 mice showed a normal appearance with no inflammatory infiltrate and no eosinophils. Among IL-4 mice that acutely rejected their bm12 skin graft, we observed a dense polymorphonuclear infiltrate. The depletion of neutrophils significantly prolonged bm12 graft survival. CONCLUSIONS Eosinophil infiltrates, typical of MHC class II disparate acute skin graft rejection, are critically dependent on the availability of IL-4. IL-4 mice reject MHC class II disparate skin grafts by a pathway of rejection where neutrophils play a direct causal role.
Collapse
|
15
|
Marrero I, Benvenutti LA, Kalil J, Coelho V. Autoreactivity to self H-2Kb peptides in TAP1 mice. Intravenous administration of H-2Kb class I-derived peptides induces long-term survival of grafts from C57BL/6 donors. Immunology 2005; 115:484-94. [PMID: 16011517 PMCID: PMC1782177 DOI: 10.1111/j.1365-2567.2005.02182.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We and others have previously shown that TAP1-/- mice (H-2b) reject grafts from donors without major histocompatibility complex (MHC) disparity that express wild-type levels of H-2b class I molecules (C57BL/6, TAP1+/+ mice). In this same model, we also showed that subcutaneous priming of TAP1-/- mice with synthetic peptides derived from the H-2Kb molecule accelerated graft rejection and that in vivo depletion of CD4+ T cells induced a significant prolongation of graft survival, suggesting an important role for CD4 T cells. We hypothesize that, in this model, rejection is triggered by the recognition of class I molecules or derived peptides, in an inflammatory microenvironment, by a functionally altered autoreactive T-cell repertoire that escapes the control of peripheral regulatory mechanisms. In the present study, we analysed the cellular autoreactivity induced by synthetic peptides derived from the H-2Kb sequence in naive and TAP1-/- mice transplanted with C57BL/6 grafts, and investigated whether intravenous modulation of autoreactivity to these peptides induced transplantation tolerance. We showed that TAP1-/- mice have peripheral autoreactive T cells that recognize H-2Kb peptides. A significant amplification of proliferation against these peptides was detected in TAP1-/- mice that rejected grafts, indicating that the inflammatory context of transplantation induced peripheral expansion of these autoreactive T cells. Furthermore, intravenous injection of H-2Kb-derived peptides significantly prolonged graft survival in some animals. In these mice (> 100 days graft survival), we observed intragraft inhibition of interferon-gamma and interleukin-10 expression, suggesting that these cytokines have an active role during the rejection. In conclusion, our present data indicate that inflammatory autoreactive T cells directed against H-2Kb peptides can be inhibited in the periphery to prolong graft survival in TAP1-/- mice.
Collapse
Affiliation(s)
- Idania Marrero
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Luiz Alberto Benvenutti
- Department of Pathology, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
| | - Jorge Kalil
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Verônica Coelho
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| |
Collapse
|
16
|
Trull AK, Akhlaghi F, Charman SC, Endenberg S, Majid O, Cornelissen J, Steel L, Parameshwar J, Wallwork J, Large S. Immunosuppression, eotaxin and the diagnostic changes in eosinophils that precede early acute heart allograft rejection. Transpl Immunol 2004; 12:159-66. [PMID: 14967314 DOI: 10.1016/s0966-3274(03)00077-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2003] [Revised: 07/28/2003] [Accepted: 08/05/2003] [Indexed: 10/27/2022]
Abstract
Peripheral blood eosinophil counts (EOS) are undetectable in 40% blood samples sent for routine haematology at Papworth Hospital during the first 3 months after heart transplantation (HTx). Increases in EOS usually precede the development of allograft rejection by a median of 4 days. We compared the effects of cyclosporin (dose and total blood concentration), prednisolone (dose and both total and unbound plasma concentrations) and azathioprine, as well as plasma concentrations of the CCR-3 chemokines, eotaxin and RANTES, on changes in EOS in 47 consecutive HTx recipients, with a median follow-up of 90 (IQR 85-95) days. Multivariate analysis confirmed the independent association between both prednisolone dose (P<0.0001) and eotaxin (P<0.0001) and changes in EOS. The plasma eotaxin concentration was, in turn, most closely associated with the cyclosporin dose (P<0.001) and plasma prednisolone concentration (P=0.022). The blood cyclosporin concentration (P=0.028), EOS (P=0.012) and prednisolone dose (P=0.015) were all independently associated with the risk of treated acute rejection. When prednisolone pharmacokinetic parameters were substituted for the prednisolone dose in this multivariate model, only the pharmacokinetic parameter retained a significant association with the risk of rejection. Changes in EOS preceding cardiac allograft rejection are directly associated with plasma eotaxin concentrations and indirectly with prednisolone dosage. Cyclosporin may also indirectly influence these changes by inhibiting eotaxin production. EOS, prednisolone dose and blood cyclosporin concentrations were independently associated with the risk of acute rejection. The total and unbound fractions of prednisolone in plasma appear to be even more closely related to rejection but are difficult to measure. Monitoring EOS, as a surrogate measure of prednisolone immunosuppression, may be more cost-effective for controlling rejection than conventional cyclosporin monitoring in the first 6 weeks after HTx.
Collapse
Affiliation(s)
- Andrew K Trull
- Department of Clinical Pharmacology, Papworth Hospital, Cambridge CB3 8RE, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Halloran PF, Urmson J, Ramassar V, Melk A, Zhu LF, Halloran BP, Bleackley RC. Lesions of T-cell-mediated kidney allograft rejection in mice do not require perforin or granzymes A and B. Am J Transplant 2004; 4:705-12. [PMID: 15084164 DOI: 10.1111/j.1600-6143.2004.00421.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Organ allograft rejection is strongly associated with the presence of alloreactive cytotoxic T cells but the role of cytotoxicity in the pathologic lesions is unclear. Previous studies showed that the principal lesions of kidney rejection - interstitial infiltration, tubulitis, and endothelial arteritis - are T-cell-dependent and antibody-independent. We studied the role of cytotoxic granule components perforin and granzymes A and B in the evolution of the T-cell-mediated lesions of mouse kidney transplant rejection. By real-time RT-PCR, allografts rejecting in wild-type hosts at days 5, 7, 21, and 42 showed massively elevated and persistent expression of perforin and granzymes A and B, but evolution of tubulitis and arteritis did not correlate with increasing granzyme or perforin expression. Allografts transplanted into hosts with disrupted genes for perforin or granzymes A and B showed no change in tubulitis, arteritis, or MHC induction. Thus the development of the histologic lesions diagnostic of T-cell-mediated kidney transplant rejection are associated with but not mediated by perforin or granzyme A or B. Together with previous graft survival studies, these results indicate that the granule-associated cytotoxic mechanisms of T cells are not the effectors of T-cell-mediated allograft rejection.
Collapse
Affiliation(s)
- Philip F Halloran
- Department of Medicine, Division of Nephrology & Transplantation Immunology, University of Alberta, Edmonton, Alberta, Canada.
| | | | | | | | | | | | | |
Collapse
|
18
|
He C, Schenk S, Zhang Q, Valujskikh A, Bayer J, Fairchild RL, Heeger PS. Effects of T cell frequency and graft size on transplant outcome in mice. THE JOURNAL OF IMMUNOLOGY 2004; 172:240-7. [PMID: 14688331 DOI: 10.4049/jimmunol.172.1.240] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The features that determine whether graft-reactive T lymphocytes develop into effector cells capable of mediating organ destruction are not well understood. To investigate potential factors involved in this process, we first confirmed that female recipient mice acutely rejected minor Ag-disparate male skin, but not heart transplants. Despite this difference in outcome, heart and skin transplantation induced antidonor T cell responses of similar magnitude, specificity, and cytokine profile. The heart-graft-primed T cells transiently infiltrated the graft and ultimately induced the development of chronic transplant vasculopathy. Increasing the frequency of donor-reactive T cells by presensitization or by using TCR (CD8+ antimale)-transgenic recipients did not mediate acute rejection but accelerated the pace and severity of the vasculopathy. Surprisingly, decreasing the tissue mass of the donor heart by 50% resulted in acute rejection of these smaller grafts without increasing the frequency of antidonor effector T cells in the recipients. In complementary studies, placement of one or two male skin grafts on a single recipient did not affect the frequency or cytokine profile of the induced antimale T cell repertoire. Nonetheless, the recipients of single grafts acutely rejected the transplanted skin while the recipients of two skin grafts did not. These results provide new insight into the pathogenesis of transplant vasculopathy and provide an explanation for the difference in outcome between murine skin and heart transplants by highlighting the novel concept that the efficiency of transplant-reactive T cell immunity is heavily influenced by the tissue burden it encounters at the effector stage.
Collapse
Affiliation(s)
- Chunshui He
- Department of Immunology and Glickman Urologic Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Le Moine A, Goldman M. Non-classical pathways of cell-mediated allograft rejection: new challenges for tolerance induction? Am J Transplant 2003; 3:101-6. [PMID: 12603204 DOI: 10.1034/j.1600-6143.2002.00026.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Allograft rejection results from separate pathways primarily controlled by CD4+ T cells. Refinement of transplantation models together with investigations on rejection occurring despite co-stimulation blockade revealed unexpected pathways involving CD8+ T cells, NK cells and Th2 cytokines. In this minireview, we discuss these non-classical pathways of allograft rejection and their relevance for the induction of tolerance in the clinics.
Collapse
Affiliation(s)
- Alain Le Moine
- Laboratory of Experimental Immunology, Faculty of Medicine, Université Libre de Bruxelles, 808 route de Lennik, B-1070 Brussels, Belgium
| | | |
Collapse
|