1
|
House EL, Kim SY, Chalupa D, Hernady E, Groves AM, Johnston CJ, McGraw MD. IL-17A neutralization fails to attenuate airway remodeling and potentiates a proinflammatory lung microenvironment in diacetyl-exposed rats. Am J Physiol Lung Cell Mol Physiol 2023; 325:L434-L446. [PMID: 37642674 PMCID: PMC10639012 DOI: 10.1152/ajplung.00082.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Bronchiolitis obliterans (BO) is a devastating lung disease that can develop following inhalation exposure to certain chemicals. Diacetyl (DA) is one chemical commonly associated with BO development when inhaled at occupational levels. Previous studies in rats have shown that repetitive DA vapor exposures increased lung CD4+CD25+ T cells and bronchoalveolar (BAL) interleukin-17A (IL-17A) concentrations concurrent with the development of airway remodeling. We hypothesized that IL-17A neutralization would attenuate the severity of airway remodeling after repetitive DA vapor exposures. Sprague-Dawley rats were exposed to 200 parts-per-million DA vapor or filtered air (RA) for 6 h/day × 5 days and monitored for 2 wk postexposure. Treatment with IL-17A neutralization (αIL-17A) or IgG (control) began immediately following exposures and continued twice weekly until study's end. Lungs were harvested for histology, flow cytometry, and BAL analyses. Survival, oxygen saturations, and percent weight change decreased significantly in DA-exposed versus RA-exposed rats, but did not differ significantly between DA + αIL-17A versus DA + IgG. Similarly, the number nor severity of airway lesions did not differ significantly between DA + αIL-17A versus DA + IgG rats despite the percentage of lung regulatory T cells increasing with decreased BAL IL-17A concentrations. Ashcroft scoring of the distal lung parenchyma suggested worse parenchymal remodeling in DA + αIL-17A versus DA + IgG rats with increased expression of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and nuclear factor-kappa B (NF-κB). Collectively, IL-17A neutralization in DA-exposed rats failed to attenuate airway remodeling with increased expression of pro-inflammatory cytokines TNF-α, IL-1β, and NF-κB.NEW & NOTEWORTHY Interleukin-17A (IL-17A) neutralization has shown benefit previously in preclinical models of transplant-associated bronchiolitis obliterans (BO), yet it remains unknown whether IL-17A neutralization has similar benefit for other forms of BO. Here, IL-17A neutralization fails to prevent severe airway remodeling in rats exposed repetitively to the flavoring chemical diacetyl, and instead, promotes a proinflammatory microenvironment with increased expression of TNF-α, IL-1β, and NF-κB within the lung.
Collapse
Affiliation(s)
- Emma L House
- Department of Pathology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York, United States
| | - So-Young Kim
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - David Chalupa
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Eric Hernady
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, United States
| | - Angela M Groves
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, New York, United States
| | - Carl J Johnston
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pediatrics, Division of Neonatology, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew D McGraw
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
2
|
Inhibiting S100A8/A9 attenuates airway obstruction in a mouse model of heterotopic tracheal transplantation. Biochem Biophys Res Commun 2022; 629:86-94. [DOI: 10.1016/j.bbrc.2022.08.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022]
|
3
|
Higuchi H, Kamimura D, Jiang JJ, Atsumi T, Iwami D, Hotta K, Harada H, Takada Y, Kanno-Okada H, Hatanaka KC, Tanaka Y, Shinohara N, Murakami M. Orosomucoid 1 is involved in the development of chronic allograft rejection after kidney transplantation. Int Immunol 2020; 32:335-346. [PMID: 31930291 DOI: 10.1093/intimm/dxaa003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/09/2020] [Indexed: 12/22/2022] Open
Abstract
Chronic allograft rejection is the most common cause of long-term allograft failure. One reason is that current diagnostics and therapeutics for chronic allograft rejection are very limited. We here show that enhanced NFκB signaling in kidney grafts contributes to chronic active antibody-mediated rejection (CAAMR), which is a major pathology of chronic kidney allograft rejections. Moreover, we found that urinary orosomucoid 1 (ORM1) is a candidate marker molecule and therapeutic target for CAAMR. Indeed, urinary ORM1 concentration was significantly higher in kidney transplant recipients pathologically diagnosed with CAAMR than in kidney transplant recipients with normal histology, calcineurin inhibitor toxicity, or interstitial fibrosis and tubular atrophy. Additionally, we found that kidney biopsy samples with CAAMR expressed more ORM1 and had higher NFκB and STAT3 activation in tubular cells than samples from non-CAAMR samples. Consistently, ORM1 production was induced after cytokine-mediated NFκB and STAT3 activation in primary kidney tubular cells. The loss- and gain-of-function of ORM1 suppressed and promoted NFκB activation, respectively. Finally, ORM1-enhanced NFκB-mediated inflammation development in vivo. These results suggest that an enhanced NFκB-dependent pathway following NFκB and STAT3 activation in the grafts is involved in the development of chronic allograft rejection after kidney transplantation and that ORM1 is a non-invasive candidate biomarker and possible therapeutic target for chronic kidney allograft rejection.
Collapse
Affiliation(s)
- Haruka Higuchi
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daisuke Kamimura
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Jing-Jing Jiang
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Institute of Preventive Genomic Medicine, School of Life Sciences, Northwest University, Xian, China
| | - Toru Atsumi
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daiki Iwami
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kiyohiko Hotta
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Harada
- Department of Kidney Transplant Surgery, Sapporo City General Hospital, Sapporo, Japan
| | - Yusuke Takada
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiromi Kanno-Okada
- Institute of Preventive Genomic Medicine, School of Life Sciences, Northwest University, Xian, China
| | - Kanako C Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Yuki Tanaka
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
4
|
IL-17A Contributes to Lung Fibrosis in a Model of Chronic Pulmonary Graft-versus-host Disease. Transplantation 2020; 103:2264-2274. [PMID: 31658231 DOI: 10.1097/tp.0000000000002837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic pulmonary graft-versus-host disease (cpGVHD) after hematopoietic cell transplant (HCT) manifests as progressive airway and parenchymal lung fibrosis. On the basis of our prior data, mice that undergo allogeneic HCT with Tbet-knockout donors (AlloTbet) have increased lung Th17 cells and IL-17A and develop fibrosis resembling human cpGVHD. The role of IL-17A in posttransplant pulmonary fibrosis remains incompletely understood. We hypothesized that IL-17A is necessary for development of murine cpGVHD in this model. METHODS AlloTbet mice received weekly intraperitoneal anti-IL-17A or IgG (200 μg/mouse) starting 2 weeks post-HCT and were sacrificed after week 5. Histologic airway and parenchymal fibrosis were semiquantitatively graded in a blinded fashion. Lung cells and proteins were measured by flow cytometry, ELISA, and multicytokine assays. RESULTS Anti-IL-17A modestly decreased airway and parenchymal lung fibrosis, along with a striking reduction in pulmonary neutrophilia, IL-6, MIP-1α, MIP-1β, CXCL1, and CXCL5 in AlloTbet mice. Additionally, anti-IL-17A decreased CCL2, inflammatory monocytes and macrophages, and Th17 cells. CONCLUSIONS In the setting of murine AlloHCT with Tbet donors, IL-17A blockade decreases fibrotic features of cpGVHD. This may be mediated by the observed reduction in neutrophils or specific lung monocyte and macrophage populations or alternatively via a direct effect on fibroblasts. Collectively, our results further suggest that anti-IL-17A strategies could prove useful in preventing alloimmune-driven fibrotic lung diseases.
Collapse
|
5
|
IL-17A Is Critical for CD8+ T Effector Response in Airway Epithelial Injury After Transplantation. Transplantation 2019; 102:e483-e493. [PMID: 30211827 DOI: 10.1097/tp.0000000000002452] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Airway epithelium is the primary target of trachea and lung transplant rejection, the degree of epithelial injury is closely correlated with obliterative bronchiolitis development. In this study, we investigated the cellular and molecular mechanisms of IL-17A-mediated airway epithelial injury after transplantation. METHODS Murine orthotopic allogeneic trachea or lung transplants were implemented in wild type or RORγt mice. Recipients received anti-IL-17A or anti-IFNγ for cytokine neutralization, anti-CD8 for CD8 T-cell depletion, or STAT3 inhibitor to suppress type 17 CD4+/CD8+ T cell development. Airway injury and graft inflammatory cell infiltration were examined by histopathology and immunohistochemistry. Gene expression of IL-17A, IFNγ, perforin, granzyme B, and chemokines in grafts was quantitated by real-time RT-PCR. RESULTS IL-17A and IFNγ were rapidly expressed and associated with epithelial injury and CD8 T-cell accumulation after allotransplantation. Depletion of CD8 T cells prevented airway epithelial injury. Neutralization of IL-17A or devoid of IL-17A production by RORγt deficiency improved airway epithelial integrity of the trachea allografts. Anti-IL-17A reduced the expression of CXCL9, CXCL10, CXCL11, and CCL20, and abolished CD8 T-cell accumulation in the trachea allografts. Inhibition of STAT3 activation significantly reduced IL-17A expression in both trachea and lung allografts; however, it increased IFNγ expression and cytotoxic activities, which resulted in the failure of airway protection. CONCLUSIONS Our data reveal the critical role of IL-17A in mediating CD8 T effector response that causes airway epithelial injury and lung allograft rejection, and indicate that inhibition of STAT3 signals could drive CD8 T cells from Tc17 toward Tc1 development.
Collapse
|
6
|
Haynes LD, Julliard WA, Mezrich JD, Leverson G, Meyer KC, Burlingham WJ. Specific Donor HLA-DR Types Correlate With Altered Susceptibility to Development of Chronic Lung Allograft Dysfunction. Transplantation 2019; 102:1132-1138. [PMID: 29360666 DOI: 10.1097/tp.0000000000002107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The greatest challenge to long-term graft survival is the development of chronic lung allograft dysfunction. Th17 responses to collagen type V (colV) predispose lung transplant patients to the severe obstructive form of chronic lung allograft dysfunction, known as bronchiolitis obliterans syndrome (BOS). In a previous study cohort (n = 54), pretransplant colV responses were increased in recipients expressing HLA-DR15, consistent with the high binding avidity of colV (α1) peptides for HLA-DR15, whereas BOS incidence, which was known to be strongly associated with posttransplant autoimmunity to colV, was higher in patients who themselves lacked HLA-DR15, but whose lung donor expressed it. METHODS To determine if this DR-restricted effect on BOS incidence could be validated in a larger cohort, we performed a retrospective analysis of outcomes for 351 lung transplant recipients transplanted between 1988 and 2008 at the University of Wisconsin. All subjects were followed until graft loss, death, loss to follow-up, or through 2014, with an average follow-up of 7 years. Comparisons were made between recipients who did or did not develop BOS. Grading of BOS followed the recommendations of the international society for heart and lung transplantation. RESULTS Donor HLA-DR15 was indeed associated with increased susceptibility to severe BOS in this population. We also discovered that HLA-DR7 expression by the donor or HLA-DR17 expression by the recipient decreased susceptibility. CONCLUSIONS We show in this retrospective study that specific donor HLA class II types are important in lung transplantation, because they are associated with either protection from or susceptibility to development of severe BOS.
Collapse
Affiliation(s)
- Lynn D Haynes
- Transplant Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| | - Walker A Julliard
- Transplant Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| | - Joshua D Mezrich
- Transplant Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| | - Glen Leverson
- Transplant Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| | - Keith C Meyer
- Department of Pulmonology, University of Wisconsin-Madison, Madison, WI
| | - William J Burlingham
- Transplant Division, Department of Surgery, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
7
|
SOCS3 overexpression in T cells ameliorates chronic airway obstruction in a murine heterotopic tracheal transplantation model. Surg Today 2019; 49:443-450. [PMID: 30617600 DOI: 10.1007/s00595-018-1753-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/30/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE Suppressor of cytokine signaling-3 (SOCS3) is a negative feedback inhibitor of cytokine signaling with T-cell-mediated immunosuppressive effects on obliterative bronchiolitis (OB). In this study, we aimed to investigate the impact of T-cell-specific overexpression of SOCS3 using a murine heterotopic tracheal transplantation (HTT) model. METHODS Tracheal allografts from BALB/c mice were subcutaneously transplanted into wild-type C57BL/6J (B6; WT) mice and SOCS3 transgenic B6 (SOCS3TG) mice. Tracheal allografts were analyzed by immunohistochemistry and quantitative polymerase chain reaction assays at days 7 and 21. RESULTS At day 21, allografts in SOCS3TG mice showed significant amelioration of airway obstruction and epithelial loss compared with allografts in WT mice. The intragraft expression of IFN-γ and CXCL10 was suppressed, while that of IL-4 was enhanced in SOCS3TG mice at day 7. The T-bet levels were lower in SOCS3TG allografts than in WT allografts at day 7. CONCLUSION We revealed that the overexpression of SOCS3 in T cells effectively ameliorates OB development in a murine HTT model by inhibiting the Th1 phenotype in the early phase. Our results suggest that the regulation of the T-cell response, through the modulation of SOCS expression, has potential as a new therapeutic strategy for chronic lung allograft dysfunction.
Collapse
|
8
|
McCallion O, Hester J, Issa F. Deciphering the Contribution of γδ T Cells to Outcomes in Transplantation. Transplantation 2018; 102:1983-1993. [PMID: 29994977 PMCID: PMC6215479 DOI: 10.1097/tp.0000000000002335] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
γδ T cells are a subpopulation of lymphocytes expressing heterodimeric T-cell receptors composed of γ and δ chains. They are morphologically and functionally heterogeneous, innate yet also adaptive in behavior, and exhibit diverse activities spanning immunosurveillance, immunomodulation, and direct cytotoxicity. The specific responses of γδ T cells to allografts are yet to be fully elucidated with evidence of both detrimental and tolerogenic roles in different settings. Here we present an overview of γδ T-cell literature, consider ways in which their functional heterogeneity contributes to the outcomes after transplantation, and reflect on methods to harness their beneficial properties.
Collapse
Affiliation(s)
- Oliver McCallion
- Transplantation Research Immunology Group, University of Oxford, Oxford, United Kingdom
| | - Joanna Hester
- Transplantation Research Immunology Group, University of Oxford, Oxford, United Kingdom
| | - Fadi Issa
- Transplantation Research Immunology Group, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Chen R, Liang F, Chen Q, Xu J, Ding Y. A novel model for dissecting roles of IL-17 in lung transplantation. J Thorac Dis 2018; 10:3298-3307. [PMID: 30069326 DOI: 10.21037/jtd.2018.05.176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background The long-term success of lung transplantation is limited by the development of chronic lung allograft dysfunction (CLAD) in which IL-17 plays an important role. Direct evidence of IL-17-mediated allograft rejection has been observed when T-bet is absent. However, lack of T-bet also leads to failure in production of IFN-γ which is required for tolerance induction and allograft acceptance, as T-bet deficiency results in IL-17-expressing CD8+ T cells mediated costimulation blockade-resistant allograft rejection. Our previous research demonstrated that additional STAT6 deficiency to T-bet deficiency resulted in Th17-dominant immune responses, and importantly, restored IFN-γ production. Here we investigated whether T-bet/STAT6 double knout-out (DKO) mice as allograft recipients could provide a useful model to study IL-17 and Th17 in lung transplantation. Methods Murine orthotopic allogeneic lung transplants were performed in C57BL/6 wild type (WT) or T-bet/STAT6 DKO (C57BL/6 background) mice using MHC fully mismatched BALB/c donors. Syngeneic transplants were also performed in WT C57BL/6 mice using C57BL/6 donors. At day 10, histopathologic characteristics and rejection status of transplanted grafts were assessed; graft-infiltrating cells were isolated and real-time RT-PCR was performed for IL-17, IFN-γ and IL-4 expressions. Results Isografts showed no apparent rejection as anticipated. Allografts of both WT and DKO recipients displayed vigorous acute rejection and expressed comparable levels of IFN-γ; while T-bet/STAT6 double deficiency resulted in much more IL-17 and less IL-4 production. Histopathologic examination demonstrated that allografts of both WT and DKO recipients have marked inflammatory cell infiltration and pulmonary parenchyma lesion. In contrast to lymphocyte-predominant inflammation observed in WT recipients, allografts of DKO recipients displayed obvious polymorphonuclear cell infiltration and severer obliterative airway inflammation. Compared to WT recipients, the ratio of graft-infiltrating CD8+ versus CD4+ T cells increased significantly with much higher numbers of neutrophils in allografts of DKO recipients. Conclusions T-bet/STAT6 DKO recipients of lung allografts result in IL-17-dominant transplant immunity, retain IFN-γ responses, and develop neutrophilia, obliterative airway inflammation and acute transplant rejection. Our results indicate that T-bet/STAT6 DKO mice serving as allograft recipient could be utilized as a new viable model to study the roles of IL-17 in lung transplantation.
Collapse
Affiliation(s)
- Rongjuan Chen
- Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Fan Liang
- Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Qirui Chen
- Department of Thoracic Surgery, Chaoyang Hospital, Beijing 100020, China
| | - Jiangnan Xu
- Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Yaozhong Ding
- Department of Immunology, Capital Medical University, Beijing 100069, China
| |
Collapse
|
10
|
IL-17 contributes to the pathogenesis of obliterative bronchiolitis via regulation of M1 macrophages polarization in murine heterotopic trachea transplantation models. Int Immunopharmacol 2017; 52:51-60. [DOI: 10.1016/j.intimp.2017.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/02/2017] [Accepted: 08/25/2017] [Indexed: 12/21/2022]
|
11
|
Yamada Y, Vandermeulen E, Heigl T, Somers J, Vaneylen A, Verleden SE, Bellon H, De Vleeschauwer S, Verbeken EK, Van Raemdonck DE, Vos R, Verleden GM, Jungraithmayr W, Vanaudenaerde BM. The role of recipient derived interleukin-17A in a murine orthotopic lung transplant model of restrictive chronic lung allograft dysfunction. Transpl Immunol 2016; 39:10-17. [PMID: 27737799 DOI: 10.1016/j.trim.2016.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/10/2016] [Accepted: 10/10/2016] [Indexed: 02/06/2023]
Abstract
The single most important cause of late mortality after lung transplantation is chronic lung allograft dysfunction (CLAD). However, the pathological development of CLAD was not as simple as previously presumed and subclassification phenotypes, bronchiolitis obliterans syndrome (BOS) and restrictive CLAD (rCLAD), have been introduced. We want to re-investigate how CLAD manifests in the murine orthotopic lung transplant model and investigate the role of interleukin 17A (IL-17A) within this model. Orthotopic LTx was performed in CB57BL/6, IL-17 WT and IL-17 KO mice. In a first experiment, CB57BL/6 mice receiving an isograft (CB57BL/6) or allograft (BALB/C) were compared. In a second experiment IL-17 WT and IL-17 KO mice (both CB57BL/6 background) received an allograft (BALB/C). Mice received daily immunosuppression with steroids and cyclosporine and were sacrificed 10weeks after transplantation for histopathological analysis by an experienced lung pathologist. After murine orthotopic lung transplantation, the allograft histopathologically presented features of human rCLAD (i.e. overt inflammation, pleural/parenchymal fibrosis and obliterative bronchiolitis). In the IL-17A KO group, less inflammation in the bronchovascular axis (p=0.03) was observed and a non-significant trend towards less bronchovascular fibrosis, pleural/septal inflammation and fibrosis, and parenchymal inflammation and fibrosis when compared to WT mice. The major mismatch orthotopic lung transplant model resembles features of human rCLAD. IL-17A mediated immunity is involved in the inflammatory component, but had little influence on the degree of fibrosis. Further mechanistic and therapeutic studies in this mouse model are needed to fully understand the mechanisms in rCLAD.
Collapse
Affiliation(s)
- Y Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - E Vandermeulen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - T Heigl
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - J Somers
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - A Vaneylen
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - S E Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - H Bellon
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - S De Vleeschauwer
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - E K Verbeken
- Department of Pathology, UZ Leuven, Leuven, Belgium
| | - D E Van Raemdonck
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - R Vos
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - G M Verleden
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium
| | - W Jungraithmayr
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - B M Vanaudenaerde
- Laboratory of Respiratory Disease and Laboratory for Experimental Thoracic Surgery, Department of Clinical and Experimental Medicine, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Chen QR, Wang LF, Xia SS, Zhang YM, Xu JN, Li H, Ding YZ. Role of interleukin-17A in early graft rejection after orthotopic lung transplantation in mice. J Thorac Dis 2016; 8:1069-79. [PMID: 27293822 DOI: 10.21037/jtd.2015.12.08] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND The cellular and molecular mechanisms underlying lung allograft rejection remain poorly understood. We investigated the potential role of interleukin (IL)-17A in lung transplant rejection in a mouse model, because previous studies in clinical and rodent models have implicated IL-17A in both acute and chronic rejection. METHODS To generate an orthotopic lung transplantation model, lungs from C57BL/6 or BALB/c mice were transplanted into C57BL/6 mice (isograft and allograft models, respectively). The effects of anti-IL-17A treatment in allograft recipients were investigated. The histological features and rejection status of isografts and allografts were assessed at 3, 7, and 28 days after transplantation, and differences in graft infiltrating cells and mRNA expression of relevant cytokines were quantified at 3 and 7 days after transplantation. RESULTS As expected, isografts showed no obvious signs of rejection, whereas allografts exhibited minimal-to-mild rejection (grade A1-A2) by day 3 and moderate-to-severe rejection (grade A3-A4) by day 7, without evidence of obliterative bronchiolitis (OB). However, by 28 days, evidence of OB was observed in 67% (2/3) of allografts and severe rejection (grade A4) was observed in all. IL-17 mRNA expression in allografts was increased with rejection, and interferon (IFN)-γ and IL-6 mRNA expression levels followed a similar pattern. In contrast, IL-22 expression in allografts was only slightly increased. Antibody (Ab) neutralization of IL-17A diminished the signs of acute rejection at 7 days after transplantation in allografts, and this early protection was accompanied by a decrease in cellular stress according to histological evaluation, suggesting the involvement of IL-17A in the development of early post-transplantation lesions. CONCLUSIONS Our data indicate that IL-17A is important in the pathophysiology of allograft rejection, and neutralization of IL-17A is a potential therapeutic strategy to preventing lung transplant rejection.
Collapse
Affiliation(s)
- Qi-Rui Chen
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Li-Feng Wang
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Si-Si Xia
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Ya-Mei Zhang
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Jiang-Nan Xu
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Hui Li
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| | - Yao-Zhong Ding
- 1 Department of Thoracic Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China ; 2 Department of Immunology, Capital Medical University, Beijing 100069, China
| |
Collapse
|
13
|
Yamada Y, Jang JH, De Meester I, Baerts L, Vliegen G, Inci I, Yoshino I, Weder W, Jungraithmayr W. CD26 costimulatory blockade improves lung allograft rejection and is associated with enhanced interleukin-10 expression. J Heart Lung Transplant 2015; 35:508-17. [PMID: 26755203 DOI: 10.1016/j.healun.2015.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 10/15/2015] [Accepted: 11/19/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The ectoenzyme CD26/dipeptidyl peptidase 4 (DPP4) has costimulatory activity that contributes to T cell activation and proliferation. Here, we aimed to target this costimulatory activity for the attenuation of the alloreactive Th17-cell response during acute rejection after mouse lung transplantation. METHODS To test the CD26-costimulatory blockade in vitro, mixed lymphocyte reaction was performed between major histocompatibility complex class I and II fully mismatched cells (CD4(+) splenocytes, C57BL/6, responders, and antigen-presenting cells, BALB/c, stimulators) by adding the CD26 inhibitor vildagliptin (0-15 μg). Lung transplantation between BALB/c (donor) and C57BL/6 (recipient) mice was performed, including controls, CD26-inhibited (CD26-I, daily administration of vildagliptin [GLSynthesis, Worcester, MA], 10 mg/kg subcutaneous), and CD26 knockout (CD26KO) mice was performed. Analysis on Day 1 and 5 after transplant included immunohistochemistry, fluorescence-activated cell sorting, and enzyme-linked immunosorbent assay (ELISA) for immune cell detection and their key cytokines. RESULTS In vitro, there was a significant reduction of the Th17 cytokines interleukin (IL)-17 and IL-21. In vivo, CD26-I-treated and CD26KO mice showed significantly preserved macroscopic and histologic characteristics on Day 5 (p < 0.01), a higher partial pressure of arterial oxygen/fraction of inspired oxygen ratio (p ≤ 0.05), fewer infiltrating CD3(+) T cells (p < 0.01), but more interstitial macrophages on Day 1 (p < 0.01) compared with control. Fewer IL-17(+) cells were found in CD26-I allografts on Day 1 (p = 0.05). Higher levels of IL-10 in CD26-I and CD26KO allografts on day 5 were seen (p < 0.05). IL-10/CD206 double-staining (alternative macrophages) revealed more positive cells in CD26-I and CD26KO on Day 1 and 5 (p < 0.01). CONCLUSIONS CD26 costimulatory blockade promotes lung allograft acceptance via reduced T cell infiltration, less expression of IL-17, and increased expression of IL-10, likely to be derived from alternatively activated macrophages.
Collapse
Affiliation(s)
- Yoshito Yamada
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Jae-Hwi Jang
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ingrid De Meester
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Lesley Baerts
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Gwendolyn Vliegen
- Department of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Ilhan Inci
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Walter Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
14
|
Wu Q, Gupta PK, Suzuki H, Wagner SR, Zhang C, W.Cummings O, Fan L, Kaplan MH, Wilkes DS, Shilling RA. CD4 T Cells but Not Th17 Cells Are Required for Mouse Lung Transplant Obliterative Bronchiolitis. Am J Transplant 2015; 15:1793-1804. [PMID: 25773063 PMCID: PMC4679154 DOI: 10.1111/ajt.13215] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 12/16/2014] [Accepted: 01/05/2015] [Indexed: 01/25/2023]
Abstract
Lung transplant survival is limited by obliterative bronchiolitis (OB), but the mechanisms of OB development are unknown. Previous studies in a mouse model of orthotopic lung transplantation suggested a requirement for IL-17. We have used this orthotopic mouse model to investigate the source of IL-17A and the requirement for T cells producing IL-17A. The major sources of IL-17A were CD4(+) T cells and γδ T cells. Depletion of CD4(+) T cells led to a significantly decreased frequency and number of IL-17A(+) lymphocytes and was sufficient to prevent acute rejection and OB. However, mice with STAT3-deficient T cells, which are unable to differentiate into Th17 cells, rejected lung allografts and developed OB similar to control mice. The frequency of IL-17A(+) cells was not decreased in mice with STAT3-deficient T cells due mainly to the presence of IL-17A(+) γδ T cells. Deficiency of γδ T cells also did not affect the development of airway fibrosis. Our data suggest that CD4(+) T cells are required for OB development and expansion of IL-17A responses in the lung, while Th17 and γδ T cells are not absolutely required and may compensate for each other.
Collapse
Affiliation(s)
- Qiang Wu
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Pawan Kumar Gupta
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Hidemi Suzuki
- Pulmonary and Critical Care Medicine and Center for Immunobiology, Departments of Medicine and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sarah R. Wagner
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Chen Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Oscar W.Cummings
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lin Fan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| | - Mark H. Kaplan
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - David S. Wilkes
- Pulmonary and Critical Care Medicine and Center for Immunobiology, Departments of Medicine and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rebecca A. Shilling
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine and Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
15
|
Hu H, Zhu X, Joshi S, Lu L, Xia CQ, Patel JM. Thioredoxin priming prolongs lung allograft survival by promoting immune tolerance. PLoS One 2015; 10:e0124705. [PMID: 25933390 PMCID: PMC4416780 DOI: 10.1371/journal.pone.0124705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/07/2015] [Indexed: 01/05/2023] Open
Abstract
Tolerance to allograft antigen is the major challenge and final goal of transplant medicine. Our previous study demonstrated that thioredoxin-1 (Trx) priming of donor lung significantly protected allogeneic lung graft. To determine whether Trx priming of donor lung inhibits allograft rejection, extends allograft survival and induces immune tolerance, orthotopic left lung transplantation was performed from Lewis to Sprague-Dawley rats without immunosuppression. Donor lungs were primed with Trx at 4°C for 4 hr prior to transplantation. After up to 37 days post-transplantation, allograft lung morphology, recipient T cell and humoral alloantigen-specific immune responses were examined. We found that Trx-primed lungs exhibited much reduced acute rejection and associated lung injuries resulting in loss of graft functional area at 5-37 days post-transplant in contrast to the control groups. CD4+ T cells from the recipients with Trx-primed grafts responded to the stimulation of dendritic cells (DCs) of donor origin, in contrast to DCs from the third party, with significantly reduced proliferation. Consistent with above findings, we observed that CD4+Foxp3+ regulatory T cells in spleen cells from the recipients with Trx-primed grafts were significantly increased compared to controls, and CD4+ T cells from the recipients with Trx-primed grafts produced much higher levels of immunosuppressive cytokine, IL-10 when stimulated with allogeneic donor DCs. In addition, humoral immune tolerance was also induced as there was no significant increase levels of serum antibodies against donor antigens in Trx-lung recipients when re-challenged with allogeneic donor antigens. Our results demonstrate that one-time Trx-priming of donor lung grafts prior to transplantation significantly prolongs the survival of the grafts through inducing or promoting cellular and humoral alloantigen-specific immune tolerance, which might be associated with the induction of immunosuppressive regulatory T cells.
Collapse
Affiliation(s)
- Hanbo Hu
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida 32608, United States of America
| | - Xiaoyan Zhu
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida 32608, United States of America
| | - Sunil Joshi
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida 32608, United States of America
| | - Li Lu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida 32608, United States of America
- Research Service, North Florida/South Georgia Veterans Health System, Gainesville, Florida 32608, United States of America
| | - Chang-Qing Xia
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida 32608, United States of America
- * E-mail: (JMP); (CQX)
| | - Jawaharlal M. Patel
- Department of Medicine, University of Florida College of Medicine, Gainesville, Florida 32608, United States of America
- Research Service, North Florida/South Georgia Veterans Health System, Gainesville, Florida 32608, United States of America
- * E-mail: (JMP); (CQX)
| |
Collapse
|
16
|
Xu J, Wu Y, Wang G, Qin Y, Zhu L, Tang G, Shen Q. Inducible costimulatory molecule deficiency induced imbalance of Treg and Th17/Th2 delays rejection reaction in mice undergoing allogeneic tracheal transplantation. Am J Transl Res 2014; 6:777-785. [PMID: 25628788 PMCID: PMC4297345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/15/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVE This study aimed to investigate the role of inducible costimulatory molecule (ICOS) pathway in the rejection reaction of mice undergoing allogeneic tracheal transplantation. METHODS The bronchus was separated from wide-type (WT) BalB/c mice and transplanted into WT BalB/c mice, C57 mice and icos(-/-) mice to prepare the obliterative bronchiolitis (OB) animal model. The transplanted bronchus was pathologically examined; flow cytometry was done to detect the T cell subsets and activity of the bronchus and spleen of recipient mice. RESULTS 21 d after transplantation, evident rejection reaction was observed and the proportion of Th2 and Th17 cells increased significantly in the bronchus and spleen in C57 mice receiving allogeneic tracheal transplantation when compared with mice with autologous transplantation, but the proportion of Treg cells was comparable between them. When compared with WT BalB/c mice, the proportion of Th2, Th17 and Treg cells reduced markedly and rejection reaction was attenuated in icos(-/-) mice receiving tracheal transplantation, although rejection reaction was still noted. CONCLUSION icos knockout may delay the rejection reaction after tracheal transplantation, which might be ascribed to the imbalance among Th2, Th17 and Treg cells.
Collapse
Affiliation(s)
- Jingsong Xu
- Department of Pulmonary Medicine, 94th Hospital of The Chinese People’s Liberation ArmyNanchang, China
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical UniversityShanghai, China
| | - Yu Wu
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical UniversityShanghai, China
- Department of Laboratory Medicine, 94th Hospital of The Chinese People’s Liberation ArmyNanchang, China
| | - Guifang Wang
- Department of Pulmonary Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Yanghua Qin
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical UniversityShanghai, China
| | - Li Zhu
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical UniversityShanghai, China
| | - Gusheng Tang
- Institute of Hematology, Changhai Hospital, Second Military Medical UniversityShanghai, China
| | - Qian Shen
- Department of Laboratory Medicine, Changhai Hospital, Second Military Medical UniversityShanghai, China
| |
Collapse
|