1
|
Mauer J, Kuckhahn A, Ramsperger-Gleixner M, Ensminger SM, Distler JH, Weyand M, Heim C. Nintedanib reduces alloimmune-induced chronic airway changes in murine tracheal allografts. Transpl Immunol 2022; 73:101608. [DOI: 10.1016/j.trim.2022.101608] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/12/2022] [Accepted: 04/23/2022] [Indexed: 01/01/2023]
|
2
|
Hanka I, Stamminger T, Ramsperger-Gleixner M, Kuckhahn AV, Müller R, Weyand M, Heim C. Role of CMV chemokine receptor M33 in airway graft rejection in a mouse transplant model. Transpl Immunol 2021; 67:101415. [PMID: 34033867 DOI: 10.1016/j.trim.2021.101415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is a risk factor for bronchiolitis obliterans (BO), one form of chronic lung allograft dysfunction (CLAD). The viral chemokine receptor M33 is essential for successful spread of murine CMV to host salivary glands. In the present study we investigated the impact of M33 on chronic airway rejection. METHODS MHC I-mismatched tracheas of C·B10-H2b/LilMcdJ mice were transplanted into BALB/c (H2d) recipients and infected at different dates with wild type (WT) or M33-deleted (delM33) MCMV representing clinical settings of viral recipient (R)-donor (D)-serostatus: (D-/R+) or (D+/R-). Grafts were recovered for gene expression and histological / immunofluorescence analysis, respectively. RESULTS Evaluations showed significantly increased signs of chronic rejection in WT-infected mice compared to uninfected allografts seen in lower epithelium/lamina propria-ratio (ELR) (ELR 0.46 ± 0.07 [WT post] vs. ELR 0.66 ± 0.10 [non-inf.]; p < 0.05). The rejection in delM33-infected groups was significantly reduced vs. WT-infected groups (0.67 ± 0.04 [delM33 post]; vs. WT post p < 0.05). Furthermore, decreased rejection was observed in WT pre-infected compared to post-infected groups (0.56 ± 0.08 [WT pre]; vs. WT post p < 0.05). CD8+ T cell infiltration was significantly higher in WT-post compared to the delM33 infected or non-infected allografts. CONCLUSIONS These data support the role of the CMV in accelerating CLAD. The deletion of chemokine receptor M33 leads to attenuated rejection.
Collapse
Affiliation(s)
- Isabella Hanka
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Thomas Stamminger
- Institute for Virology, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Martina Ramsperger-Gleixner
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Annika V Kuckhahn
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Regina Müller
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany.
| |
Collapse
|
3
|
Heim C, Kuckhahn A, Ramsperger-Gleixner M, Nicolls MR, Weyand M, Ensminger SM. Microvasculature in murine tracheal allografts after combined therapy with clopidogrel and everolimus. Interact Cardiovasc Thorac Surg 2021; 32:960-968. [PMID: 33550369 DOI: 10.1093/icvts/ivab021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/01/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Survival after lung transplantation is mainly limited by the development of chronic lung allograft dysfunction. Previous studies have suggested T-cell mediated proliferation and microvascular changes in experimental small airways models as potential therapeutic targets. The aim of this study was to assess microvascular changes in murine orthotopic tracheal allografts after treatment with everolimus alone or in combination with clopidogrel. METHODS C57Bl/6 (H-2b) donor tracheas were orthotopically transplanted into CBA (H-2k) recipients. Mice received daily injections of everolimus (0.05 mg/kg) alone or combined with clopidogrel (1 mg/kg). Twenty-eight days after transplantation, ratio of the thickness of tracheal epithelium and lamina propria was measured as an indicator for chronic rejection. Additionally, graft oxygenation and graft perfusion were detected on postoperative days 4, 10 and 28. Quantitative reverse transcription polymerase chain reaction analysis was used for gene expression analysis. RESULTS While syngeneic grafts showed a stable tissue pO2 and undisturbed microvascular perfusion, rejecting allografts had a drastic decline in both parameters as well as a flattened epithelium and an increased thickness of the lamina propria. Treatment with everolimus reduced allogeneic fibroproliferation, but had no protective effects on the microvasculature; polymerase chain reaction analysis indicated hypoxic stress and inflammation. Combining everolimus with clopidogrel improved microvascular integrity in the tracheal grafts, but had no synergistic effect in preventing obliterative bronchiolitis development. CONCLUSIONS These data demonstrate that the ability of everolimus to reduce the development of post-transplant obliterative bronchiolitis is not caused by microvascular protection and has no synergistic effects with clopidogrel in acute airway rejection.
Collapse
Affiliation(s)
- Christian Heim
- Department of Cardiac Surgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Annika Kuckhahn
- Department of Cardiac Surgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - Mark R Nicolls
- Veterans Affairs Palo Alto Health Care System/Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Weyand
- Department of Cardiac Surgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | | |
Collapse
|
4
|
Naikawadi RP, Green G, Jones KD, Achtar-Zadeh N, Mieleszko JE, Arnould I, Kukreja J, Greenland JR, Wolters PJ. Airway Epithelial Telomere Dysfunction Drives Remodeling Similar to Chronic Lung Allograft Dysfunction. Am J Respir Cell Mol Biol 2020; 63:490-501. [PMID: 32551854 DOI: 10.1165/rcmb.2019-0374oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Telomere dysfunction is associated with multiple fibrotic lung processes, including chronic lung allograft dysfunction (CLAD)-the major limitation to long-term survival following lung transplantation. Although shorter donor telomere lengths are associated with an increased risk of CLAD, it is unknown whether short telomeres are a cause or consequence of CLAD pathology. Our objective was to test whether telomere dysfunction contributes to the pathologic changes observed in CLAD. Histopathologic and molecular analysis of human CLAD lungs demonstrated shortened telomeres in lung epithelial cells quantified by teloFISH, increased numbers of surfactant protein C immunoreactive type II alveolar epithelial cells, and increased expression of senescence markers (β-galactosidase, p16, p53, and p21) in lung epithelial cells. TRF1F/F (telomere repeat binding factor 1 flox/flox) mice were crossed with tamoxifen-inducible SCGB1a1-cre mice to generate SCGB1a1-creTRF1F/F mice. Following 9 months of tamoxifen-induced deletion of TRF1 in club cells, mice developed mixed obstructive and restrictive lung physiology, small airway obliteration on microcomputed tomography, a fourfold decrease in telomere length in airway epithelial cells, collagen deposition around bronchioles and adjacent lung parenchyma, increased type II aveolar epithelial cell numbers, expression of senescence-associated β-galactosidase in epithelial cells, and decreased SCGB1a1 expression in airway epithelial cells. These findings demonstrate that telomere dysfunction isolated to airway epithelial cells leads to airway-centric lung remodeling and fibrosis similar to that observed in patients with CLAD and suggest that lung epithelial cell telomere dysfunction may be a molecular driver of CLAD.
Collapse
Affiliation(s)
- Ram P Naikawadi
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Gary Green
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | | | - Natalia Achtar-Zadeh
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Julia E Mieleszko
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Isabel Arnould
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California; and
| | - John R Greenland
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine.,Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine
| |
Collapse
|
5
|
Hayes D, Rayner RE, Hill CL, Alsudayri A, Tadesse M, Lallier SW, Parekh H, Brock GN, Cormet-Boyaka E, Reynolds SD. Airway epithelial stem cell chimerism in cystic fibrosis lung transplant recipients. J Cyst Fibros 2020; 20:165-172. [PMID: 33187933 PMCID: PMC9078212 DOI: 10.1016/j.jcf.2020.09.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/24/2020] [Accepted: 09/29/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The conducting airway epithelium is repaired by tissue specific stem cells (TSC). In response to mild/moderate injury, each TSC repairs a discrete area of the epithelium. In contrast, severe epithelial injury stimulates TSC migration and expands the stem cell's reparative domain. Lung transplantation (LTx) can cause a moderate/severe airway injury and the remodeled airway contains a chimeric mixture of donor and recipient cells. These studies supported the hypothesis, LTx stimulates TSC migration resulting in epithelial chimerism. We tested this hypothesis in cystic fibrosis (CF) LTx patients. METHODS Airway mucosal injury was quantified using bronchoscopic imaging and a novel grading system. Bronchial brushing was used to recover TSC from 10 sites in the recipient and allograft airways. TSC chimerism was quantified by short tandem repeat analysis. TSC self-renewal and differentiation potential were assayed using the clone forming cell frequency and air-liquid-interface methods. Electrophysiology was used to determine if TSC chimerism altered epithelial ion channel activity. RESULTS LTx caused a mild to moderate airway mucosal injury. Donor and recipient TSC were identified in 91% of anastomotic sites and 93% of bronchial airways. TSC chimerism did not alter stem cell self-renewal or differentiation potential. The frequency of recipient TSC was proportional to CF Transmembrane Conductance Regulator (CFTR)-dependent ion channel activity and 33% of allograft regions were at risk for abnormal CFTR activity. CONCLUSIONS LTx in CF patients stimulates bidirectional TSC migration across the anastomoses. TSC chimerism may alter ion homeostasis and compromise the host defense capability of the allograft airway epithelium.
Collapse
Affiliation(s)
- Don Hayes
- Section of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Departments of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA; Surgery, The Ohio State University College of Medicine, Columbus, OH, USA; Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Rachael E Rayner
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Cynthia L Hill
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Alfahdah Alsudayri
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Mahelet Tadesse
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Scott W Lallier
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Hemant Parekh
- Clinical Histocompatibility/Tissue Typing Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Guy N Brock
- Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA; Center for Biostatistics and Bioinformatics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, USA
| | - Susan D Reynolds
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH, USA; Departments of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
6
|
Modulation of NLRP3 Inflammasome through Formyl Peptide Receptor 1 (Fpr-1) Pathway as a New Therapeutic Target in Bronchiolitis Obliterans Syndrome. Int J Mol Sci 2020; 21:ijms21062144. [PMID: 32244997 PMCID: PMC7139667 DOI: 10.3390/ijms21062144] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 01/12/2023] Open
Abstract
Chronic rejection is the major leading cause of morbidity and mortality after lung transplantation. Bronchiolitis obliterans syndrome (BOS), a fibroproliferative disorder of the small airways, is the main manifestation of chronic lung allograft rejection. We investigated, using transgenic mice, the mechanisms through which the deficiency of IL-1β/IL-18, Casp-1, or Fpr-1 genes could be protective in an experimental model of BOS, induced in mice by allogeneic heterotopic tracheal transplantation. Fpr-1 KO mice showed a marked reduction in histological markers of BOS and of mast cell numbers compared to other groups. Molecular analyses indicated that the absence of the Fpr-1 gene was able to decrease NF-κB nuclear translocation and modulate NLRP3 inflammasome signaling and the mitogen-activated protein kinase (MAPK) pathway in a more significant way compared to other groups. Additionally, Fpr-1 gene deletion caused a reduction in resistance to the apoptosis, assessed by the TUNEL assay. Immunohistochemical analyses indicated changes in nitrotyrosine, PARP, VEGF, and TGF-β expression associated with the pathology, which were reduced in the absence of the Fpr1 gene more so than by the deletion of IL-1β/IL-18 and Casp-1. We underline the importance of the NLRP3 inflammasome and the pathogenic role of Fpr-1 in experimental models of BOS, which is the result of the modulation of immune cell recruitment together with the modulation of local cellular activation, suggesting this gene as a new target in the control of the pathologic features of BOS.
Collapse
|
7
|
Preservation of Microvascular Integrity in Murine Orthotopic Tracheal Allografts by Clopidogrel. Transplantation 2019; 103:899-908. [PMID: 30801550 DOI: 10.1097/tp.0000000000002571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Survival after lung transplantation is mainly limited by the development of chronic lung allograft dysfunction (CLAD). The aim of this study was to investigate if platelet inhibition by clopidogrel has a functionally relevant influence on the microvascular integrity of orthotopic tracheal allografts as an anatomic basis for the development of CLAD. METHODS We orthotopically transplanted C57Bl/6 (H-2) tracheas into CBA.J (H-2) recipients who afterwards received clopidogrel (1 mg/kg). Morphometric analysis was performed by measuring epithelial height in proportion to thickness of the lamina propria (epithelium-lamina propria ratio). Tissue oxygenation was determined using a fluorescence quenching technique, and graft perfusion monitoring was performed by laser Doppler flowmetry and lectin-binding assay. Immunohistochemistry was used for detection of CD31 and inducible nitric oxide synthase while iron deposition was shown with Prussian blue reaction. Quantitative reverse transcription polymerase chain reaction analysis was used for gene expression analysis. RESULTS Isografts maintained good oxygenation and perfusion throughout the experiment, while both were drastically reduced in allografts. Treatment with clopidogrel attenuated graft hypoxia and reduced loss of perfusion. Additionally, clopidogrel led to increased epithelium-lamina propria ratio while iron deposition was impaired. Gene expression analysis revealed elevated levels of angiogenic vascular endothelial growth factor in the clopidogrel group. Improved endothelial function was shown by immunohistochemistry (CD31, inducible nitric oxide synthase). CONCLUSIONS Continuous administration of clopidogrel significantly improved tissue oxygenation, limited microvascular leakiness, and prevented airway ischemia. These data demonstrate that clopidogrel ameliorates microvascular injury during acute airway rejection, which is a known predisposing factor for the development of CLAD.
Collapse
|
8
|
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
|
9
|
Iida S, Miyairi S, Su CA, Abe T, Abe R, Tanabe K, Dvorina N, Baldwin WM, Fairchild RL. Peritransplant VLA-4 blockade inhibits endogenous memory CD8 T cell infiltration into high-risk cardiac allografts and CTLA-4Ig resistant rejection. Am J Transplant 2019; 19:998-1010. [PMID: 30372587 PMCID: PMC6433496 DOI: 10.1111/ajt.15147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 01/25/2023]
Abstract
Recipient endogenous memory CD8 T cells expressing reactivity to donor class I MHC infiltrate MHC-mismatched cardiac allografts within 24 hours after reperfusion and express effector functions mediating graft injury. The current study tested the efficacy of Very Late Antigen-4 (VLA-4) blockade to inhibit endogenous memory CD8 T cell infiltration into cardiac allografts and attenuate early posttransplant inflammation. Peritransplant anti-VLA-4 mAb given to C57BL6 (H-2b ) recipients of AJ (H-2a ) heart allografts completely inhibited endogenous memory CD4 and CD8 T cell infiltration with significant decrease in macrophage, but not neutrophil, infiltration into allografts subjected to either minimal or prolonged cold ischemic storage (CIS) prior to transplant, reduced intra-allograft IFN-γ-induced gene expression and prolonged survival of allografts subjected to prolonged CIS in CTLA-4Ig treated recipients. Anti-VLA-4 mAb also inhibited priming of donor-specific T cells producing IFN-γ until at least day 7 posttransplant. Peritransplant anti-VLA plus anti-CD154 mAb treatment similarly prolonged survival of allografts subjected to minimal or increased CIS prior to transplant. Overall, these data indicate that peritransplant anti-VLA-4 mAb inhibits early infiltration memory CD8 T cell infiltration into allografts with a marked reduction in early graft inflammation suggesting an effective strategy to attenuate negative effects of heterologous alloimmunity in recipients of higher risk grafts.
Collapse
Affiliation(s)
- Shoichi Iida
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Tokyo Women’s Medical University, Tokyo, Japan
| | - Satoshi Miyairi
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Charles A. Su
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Toyofumi Abe
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Urology, Osaka University School of Medicine, Osaka, Japan
| | - Ryo Abe
- Tokyo Women’s Medical University, Tokyo, Japan
| | | | - Nina Dvorina
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Robert L. Fairchild
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
10
|
Kawashima M, Sato M, Murakawa T, Anraku M, Konoeda C, Hosoi A, Kakimi K, Nakajima J. Role of Toll-like Receptor 4 Expressed by Fibroblasts in Allograft Fibrosis in Mouse Orthotopic Tracheal Transplantation. Transplant Proc 2018; 50:3863-3872. [PMID: 30577279 DOI: 10.1016/j.transproceed.2018.06.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/25/2018] [Accepted: 06/21/2018] [Indexed: 12/16/2022]
Abstract
Development of chronic lung allograft dysfunction involves various alloimmune-independent insults including those mediated by Toll-like receptor (TLR) signaling, which is known to activate alloimmune responses. We hypothesized that TLR signaling may also contribute to the activation of fibroblasts and promoting allograft airway fibrosis. Mouse orthotopic tracheal transplants were conducted between major histocompatibility complex (MHC)-mismatched Balb/c donor and wild-type C3H or C3H-derived TLR4 mutant recipients (nonfunctional TLR4). Immunohistochemistry on day 21 showed significantly smaller alpha-smooth muscle actin (α-SMA)-positive areas in TLR4 mutant recipients than wild-type recipients (P = .01). No difference was found for CD3+ T-cell infiltration. Proliferation of alloreactive T cells derived from the recipient spleen showed no difference between TLR4 mutant and wild-type recipients in a mixed lymphocyte reaction. The effect of TLR4 signaling was examined in primary pulmonary fibroblast cultures both with lipopolysaccharide (LPS) and transforming growth factor (TGF)-β1. Stimulation with LPS significantly increased expression of α-SMA mRNA in wild-type fibroblasts cultured with TGF-β1 compared with the control without LPS (P = .001). Taken together, these findings suggest disruption of TLR signaling leads to reduced activation of fibroblasts without affecting T-cell infiltration and proliferation in this model. TLR4-mediated activation of fibroblasts may be a potentially important mechanism of allograft remodeling.
Collapse
Affiliation(s)
- M Kawashima
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - M Sato
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - T Murakawa
- Department of Thoracic Surgery, Kansai Medical University, Osaka, Japan
| | - M Anraku
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - C Konoeda
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - A Hosoi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - K Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - J Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Bontha SV, Fernandez-Piñeros A, Maluf DG, Mas VR. Messengers of tolerance. Hum Immunol 2018; 79:362-372. [PMID: 29402484 DOI: 10.1016/j.humimm.2018.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/16/2018] [Accepted: 01/19/2018] [Indexed: 12/21/2022]
Abstract
The use of immunosuppressant drugs after organ transplantation has brought great success in the field of organ transplantation with respect to short-term outcome. However, major challenges (i.e., limited improvement of long-term survival, immunosuppressant toxicity, infections and carcinoma) demand alternate treatment approaches that minimizes the use of immunosuppressants. Interestingly, few studies have identified groups of transplant patients who developed operational tolerance and thereby keep their allograft without complications in absence of immunosuppressants. These rare groups of patients are of particular interest as study subjects for understanding mechanisms of graft tolerance that could be leveraged in future for inducing tolerance and for understanding mechanisms involved in improving long-term allograft outcomes. Also, biomarkers from these studies could benefit the larger transplant population by their application in immunosuppressant tailoring and identification of tolerant patients among patients with stably functioning allografts. This review compiles several gene expression studies performed in samples from tolerant patients in different solid organ transplantations to identify key genes and associated molecular pathways relevant to tolerance. This review is aimed at putting forth all this important work done thus far and to identify research gaps that need to be filled, in order to achieve the greater purpose of these studies.
Collapse
Affiliation(s)
- Sai Vineela Bontha
- Translational Genomics and Transplant Laboratory, Department of Surgery, University of Virginia, Charlottesville 22903, United States
| | - Angela Fernandez-Piñeros
- Translational Genomics and Transplant Laboratory, Department of Surgery, University of Virginia, Charlottesville 22903, United States
| | - Daniel G Maluf
- Translational Genomics and Transplant Laboratory, Department of Surgery, University of Virginia, Charlottesville 22903, United States; Transplant Surgery, Department of Surgery, University of Virginia, Charlottesville 22903, United States
| | - Valeria R Mas
- Translational Genomics and Transplant Laboratory, Department of Surgery, University of Virginia, Charlottesville 22903, United States.
| |
Collapse
|
12
|
Zhu P, Atkinson C, Dixit S, Cheng Q, Tran D, Patel K, Jiang YL, Esckilsen S, Miller K, Bazzle G, Allen P, Moore A, Broome AM, Nadig SN. Organ preservation with targeted rapamycin nanoparticles: a pre-treatment strategy preventing chronic rejection in vivo. RSC Adv 2018; 8:25909-25919. [PMID: 30220998 PMCID: PMC6124302 DOI: 10.1039/c8ra01555d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/24/2018] [Indexed: 12/20/2022] Open
Abstract
Hypothermic preservation is the standard of care for storing organs prior to transplantation. Endothelial and epithelial injury associated with hypothermic storage causes downstream graft injury and, as such, the choice of an ideal donor organ preservation solution remains controversial. Cold storage solutions, by design, minimize cellular necrosis and optimize cellular osmotic potential, but do little to assuage immunological cell activation or immune cell priming post transplantation. Thus, here we explore the efficacy of our previously described novel Targeted Rapamycin Micelles (TRaM) as an additive to standard-of-care University of Wisconsin preservation solution as a means to alter the immunological microenvironment post transplantation using in vivo models of tracheal and aortic allograft transplantation. In all models of transplantation, grafts pre-treated with 100 ng mL-1 of TRaM augmented preservation solution ex vivo showed a significant inhibition of chronic rejection post-transplantation, as compared to UW augmented with free rapamycin at a ten-fold higher dose. Here, for the first time, we present a novel method of organ pretreatment using a nanotherapeutic-based cellular targeted delivery system that enables donor administration of rapamycin, at a ten-fold decreased dose during cold storage. Clinically, these pretreatment strategies may positively impact post-transplant outcomes and can be readily translated to clinical scenarios.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA.,Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Carl Atkinson
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Suraj Dixit
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Qi Cheng
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA.,Institute of Organ Transplantation, Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danh Tran
- Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Kunal Patel
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Yu-Lin Jiang
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Scott Esckilsen
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Kayla Miller
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Grace Bazzle
- Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Patterson Allen
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| | - Alfred Moore
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA.
| | - Ann-Marie Broome
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, USA. .,Department of Bioengineering, Clemson University, USA
| | - Satish N Nadig
- Department of Surgery, Division of Transplant Surgery, Medical University of South Carolina, USA. .,Department of Microbiology and Immunology, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, USA
| |
Collapse
|
13
|
Lama VN, Belperio JA, Christie JD, El-Chemaly S, Fishbein MC, Gelman AE, Hancock WW, Keshavjee S, Kreisel D, Laubach VE, Looney MR, McDyer JF, Mohanakumar T, Shilling RA, Panoskaltsis-Mortari A, Wilkes DS, Eu JP, Nicolls MR. Models of Lung Transplant Research: a consensus statement from the National Heart, Lung, and Blood Institute workshop. JCI Insight 2017; 2:93121. [PMID: 28469087 DOI: 10.1172/jci.insight.93121] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lung transplantation, a cure for a number of end-stage lung diseases, continues to have the worst long-term outcomes when compared with other solid organ transplants. Preclinical modeling of the most common and serious lung transplantation complications are essential to better understand and mitigate the pathophysiological processes that lead to these complications. Various animal and in vitro models of lung transplant complications now exist and each of these models has unique strengths. However, significant issues, such as the required technical expertise as well as the robustness and clinical usefulness of these models, remain to be overcome or clarified. The National Heart, Lung, and Blood Institute (NHLBI) convened a workshop in March 2016 to review the state of preclinical science addressing the three most important complications of lung transplantation: primary graft dysfunction (PGD), acute rejection (AR), and chronic lung allograft dysfunction (CLAD). In addition, the participants of the workshop were tasked to make consensus recommendations on the best use of these complimentary models to close our knowledge gaps in PGD, AR, and CLAD. Their reviews and recommendations are summarized in this report. Furthermore, the participants outlined opportunities to collaborate and directions to accelerate research using these preclinical models.
Collapse
Affiliation(s)
- Vibha N Lama
- Department of Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - John A Belperio
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jason D Christie
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Souheil El-Chemaly
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael C Fishbein
- Department of Pathology and Laboratory Medicine, UCLA Center for the Health Sciences, Los Angeles, California, USA
| | - Andrew E Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wayne W Hancock
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shaf Keshavjee
- Division of Thoracic Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark R Looney
- Department of Medicine, UCSF School of Medicine, San Francisco, California, USA
| | - John F McDyer
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Rebecca A Shilling
- Department of Medicine, University of Illinois College of Medicine at Chicago, Illinois, USA
| | - Angela Panoskaltsis-Mortari
- Departments of Pediatrics, and Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - David S Wilkes
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jerry P Eu
- National Heart, Lung and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Mark R Nicolls
- Department of Medicine, Stanford University School of Medicine/VA Palo Alto Health Care System, Stanford, California, USA
| |
Collapse
|
14
|
Khan MA, Alanazi F, Ahmed HA, Al-Mohanna FH, Assiri AM, Broering DC. FOXP3 + regulatory T cell ameliorates microvasculature in the rejection of mouse orthotopic tracheal transplants. Clin Immunol 2016; 174:84-98. [PMID: 27939405 DOI: 10.1016/j.clim.2016.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/23/2016] [Accepted: 11/20/2016] [Indexed: 12/18/2022]
Abstract
Microvascular loss may be a root cause of chronic rejection in lung transplants, which leads to the bronchiolitis obliterans syndrome. Previous research implicates T regulatory cell (Treg) as a key component of immune modulation, however, Treg has never been examined as a reparative mediator to salvage microvasculature during transplantation. Here, we reconstituted purified Tregs in to allografts, and serially monitored allografts for tissue oxygenation, microvascular perfusion for four weeks. We demonstrated that Tregs reconstitution of allografts significantly improve tissue oxygenation, microvascular flow, epithelial repair, number of CD4+CD25highFOXP3+ Tregs, followed by an upregulation of proinflammatory, angiogenic and regulatory genes, while prevented subepithelial deposition of CD4+T cells at d10, and collagen at d28 post-transplantation. Altogether, these findings concluded that Treg-mediated immunotherapy has potential to preserve microvasculature and rescue allograft from sustained hypoxic/ischemic phase, limits airway tissue remodeling, and therefore may be a useful therapeutic tool to prevent chronic rejection after organ transplantation.
Collapse
Affiliation(s)
- Mohammad Afzal Khan
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh 11211, Saudi Arabia.
| | - Fatimah Alanazi
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh 11211, Saudi Arabia.
| | - Hala Abdalrahman Ahmed
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh 11211, Saudi Arabia.
| | - Falah Hassan Al-Mohanna
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh 11211, Saudi Arabia.
| | - Abdullah Mohammed Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh 11211, Saudi Arabia.
| | - Dieter Clemens Broering
- Organ Transplant Research Section, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, MBC 03, P.O. Box 3354, Riyadh 11211, Saudi Arabia.
| |
Collapse
|
15
|
De Vleeschauwer S, Vanaudenaerde B, Vos R, Meers C, Wauters S, Dupont L, Van Raemdonck D, Verleden G. The need for a new animal model for chronic rejection after lung transplantation. Transplant Proc 2014; 43:3476-85. [PMID: 22099823 DOI: 10.1016/j.transproceed.2011.09.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The single most important cause of late mortality after lung transplantation is obliterative bronchiolitis (OB), clinically characterized by a decrease in lung function and morphologically by characteristic changes. Recently, new insights into its pathogenesis have been acquired: risk factors have been identified and the use of azithromycin showed a dichotomy with at least 2 different phenotypes of bronchiolitis obliterans syndrome (BOS). It is clear that a good animal model is indispensable to further dissect and unravel the pathogenesis of BOS. Many animal models have been developed to study BOS but, so far, none of these models truly mimics the human situation. Looking at the definition of BOS, a good animal model implies histological OB lesions, possibility to measure lung function, and airway inflammation. This review sought to discuss, including pros and cons, all potential animal models that have been developed to study OB/BOS. It has become clear that a new animal model is needed; recent developments using an orthotopic mouse lung transplantation model may offer the answer because it mimics the human situation. The genetic variants among this species may open new perspectives for research into the pathogenesis of OB/BOS.
Collapse
Affiliation(s)
- S De Vleeschauwer
- Laboratory of Pneumology, Kathoholieke Universiteit Leuven and UZ Gasthuisberg, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Su CA, Iida S, Abe T, Fairchild RL. Endogenous memory CD8 T cells directly mediate cardiac allograft rejection. Am J Transplant 2014; 14:568-79. [PMID: 24502272 PMCID: PMC3947453 DOI: 10.1111/ajt.12605] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/21/2013] [Accepted: 11/21/2013] [Indexed: 01/25/2023]
Abstract
Differences in levels of environmentally induced memory T cells that cross-react with donor MHC molecules are postulated to account for the efficacy of allograft tolerance-inducing strategies in rodents versus their failure in nonhuman primates and human transplant patients. Strategies to study the impact of donor-reactive memory T cells on allografts in rodents have relied on the pretransplant induction of memory T cells cross-reactive with donor allogeneic MHC molecules through recipient viral infection, priming directly with donor antigen or adoptive transfer of donor antigen primed memory T cells. Each approach accelerates allograft rejection and confers resistance to tolerance induction, but also biases the T cell repertoire to strong donor reactivity. The ability of endogenous memory T cells within unprimed mice to directly reject an allograft is unknown. Here, we show a direct association between increased duration of cold ischemic allograft storage and numbers and enhanced functions of early graft infiltrating endogenous CD8 memory T cells. These T cells directly mediate rejection of allografts subjected to prolonged ischemia and this rejection is resistant to costimulatory blockade. These findings recapitulate the clinically significant impact of endogenous memory T cells with donor reactivity in a mouse transplant model in the absence of prior recipient priming.
Collapse
Affiliation(s)
- C. A. Su
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106,Glickman Urological and Kidney Institute and Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - S. Iida
- Glickman Urological and Kidney Institute and Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - T. Abe
- Glickman Urological and Kidney Institute and Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - R. L. Fairchild
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106,Glickman Urological and Kidney Institute and Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| |
Collapse
|
17
|
Abstract
Long-term allograft survival is a major challenge facing solid organ transplantation. Recent studies have shown a negative correlation between infiltration of memory T cells and allograft survival. Furthermore, blockade of leukocyte activation increases acceptance of transplanted organs, including heart, liver, and kidney. Lung allografts are associated with high rates of rejection, and therapies that increase acceptance of other transplanted organs have not translated into the lung. In this issue of the JCI, Krupnick and colleagues demonstrate in a murine model that lung allograft acceptance requires infiltration of a specific T cell population into the graft. This study highlights the unique immunobiology of the lung and the complexity of lung transplant tolerance.
Collapse
|
18
|
Epithelial to mesenchymal transition in murine tracheal allotransplantation: an immunohistochemical observation. Transplant Proc 2014; 45:1797-801. [PMID: 23769046 DOI: 10.1016/j.transproceed.2012.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 11/20/2012] [Indexed: 11/21/2022]
Abstract
BACKGROUND Aberrant epithelial repair is a crucial event in the airway remodeling that characterizes obliterative bronchiolitis (OB) in transplanted lungs. Recent data from experiments using epithelial cell lines and human airway tissues from lung transplant recipients suggest that epithelial to mesenchymal transition (EMT) plays an important role in OB. The aim of this study was to clarify whether EMT is involved in airway remodeling in an animal model. METHODS We performed orthotopic tracheal transplantation from BALB/c to C57BL/6 mice with from BALC/c to BALB/c mouse grafts as controls. Five allogeneic and 3 syngeneic recipients were humanely killed at predetermined postoperative days 2-12 as well as 14 and 21. Histology was evaluated using hematoxylin-eosin (H&E) staining. We studied the expression of specific markers, including E-cadherin, an epithelial marker; α-smooth muscle actin (SMA), and S100A4, mesenchymal markers, and zinc finger E-box-binding homeobox 1 (ZEB1), an EMT-related transcription factor. RESULTS Histologic assessment of serial H&E stains of allogeneic grafts showed remarkable pseudostratified respiratory epithelium with subepithelial inflammatory cell infiltration, as well as denuded and flattened epithelium and subepithelial fibrosis. The dynamic epithelial changes occurred earlier than the subepithelial fibrosis. Immunohistochemical evaluation indicated the emergence of α-SMA- positive epithelial cells that were most prominent on day 7. The expression of E-cadherin was attenuated in α-SMA-positive epithelial cells. S100A4 was also expressed in epithelial cells. A few days before the intraepithelial expression of α-SMA, ZEB1 emerged in the nuclei of epithelial cells. CONCLUSIONS We observed expression of an EMT-related transcription factor and mesenchymal markers along with the attenuation of epithelial marker expression in epithelial cells, several days before prominent subepithelial fibrosis formation, results that suggest epithelial cells to play an important fibrosis role in airway remodeling during epithelial to mesenchymal transition.
Collapse
|
19
|
Abstract
The myriad of co-stimulatory signals expressed, or induced, upon T-cell activation suggests that these signalling pathways shape the character and magnitude of the resulting autoreactive or alloreactive T-cell responses during autoimmunity or transplantation, respectively. Reducing pathological T-cell responses by targeting T-cell co-stimulatory pathways has met with therapeutic success in many instances, but challenges remain. In this Review, we discuss the T-cell co-stimulatory molecules that are known to have critical roles during T-cell activation, expansion, and differentiation. We also outline the functional importance of T-cell co-stimulatory molecules in transplantation, tolerance and autoimmunity, and we describe how therapeutic blockade of these pathways might be harnessed to manipulate the immune response to prevent or attenuate pathological immune responses. Ultimately, understanding the interplay between individual co-stimulatory and co-inhibitory pathways engaged during T-cell activation and differentiation will lead to rational and targeted therapeutic interventions to manipulate T-cell responses and improve clinical outcomes.
Collapse
Affiliation(s)
- Mandy L Ford
- The Emory Transplant Center, Emory University, 101 Woodruff Circle, Woodruff Memorial Research Building Suite 5105, Atlanta, GA 30322, USA
| | - Andrew B Adams
- The Emory Transplant Center, Emory University, 101 Woodruff Circle, Woodruff Memorial Research Building Suite 5105, Atlanta, GA 30322, USA
| | - Thomas C Pearson
- The Emory Transplant Center, Emory University, 101 Woodruff Circle, Woodruff Memorial Research Building Suite 5105, Atlanta, GA 30322, USA
| |
Collapse
|
20
|
Lemaître PH, Vokaer B, Charbonnier LM, Iwakura Y, Estenne M, Goldman M, Leo O, Remmelink M, Le Moine A. IL-17A mediates early post-transplant lesions after heterotopic trachea allotransplantation in Mice. PLoS One 2013; 8:e70236. [PMID: 23936171 PMCID: PMC3728020 DOI: 10.1371/journal.pone.0070236] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/18/2013] [Indexed: 12/14/2022] Open
Abstract
Primary graft dysfunction (PGD) and bronchiolitis obliterans (BO) are the leading causes of morbidity and mortality after lung transplantation. Reports from clinical and rodent models suggest the implication of IL-17A in either PGD or BO. We took advantage of the heterotopic trachea transplantation model in mice to study the direct role of IL-17A in post-transplant airway lesions. Across full MHC barrier, early lesions were controlled in IL-17A-/- or anti-IL17 treated recipients. In contrast, IL-17A deficiency did not prevent subsequent obliterative airway disease (OAD). Interestingly, this early protection occurred also in syngeneic grafts and was accompanied by a decrease in cellular stress, as attested by lower HSP70 mRNA levels, suggesting the involvement of IL-17A in ischemia-reperfusion injury (IRI). Furthermore, persistence of multipotent CK14+ epithelial stem cells underlined allograft protection afforded by IL-17A deficiency or neutralisation. Recipient-derived γδ+ and CD4+ T cells were the major source of IL-17A. However, lesions still occurred in the absence of each subset, suggesting a high redundancy between the innate and adaptive IL-17A producing cells. Notably, a double depletion significantly diminished lesions. In conclusion, this work implicated IL-17A as mediator of early post-transplant airway lesions and could be considered as a potential therapeutic target in clinical transplantation.
Collapse
Affiliation(s)
- Philippe H Lemaître
- Transplantation Medicine, Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Complement-mediated microvascular injury leads to chronic rejection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 735:233-46. [PMID: 23402031 DOI: 10.1007/978-1-4614-4118-2_16] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Microvascular loss may be an unappreciated root cause of chronic rejection for all solid organ transplants. As the only solid organ transplant that does not undergo primary systemic arterial revascularization at the time of surgery, lung transplants rely on the establishment of a microcirculation and are especially vulnerable to the effects of microvascular loss. Microangiopathy, with its attendant ischemia, can lead to tissue infarction and airway fibrosis. Maintaining healthy vasculature in lung allografts may be critical for preventing terminal airway fibrosis, also known as the bronchiolitis obliterans syndrome (BOS). BOS is the major obstacle to lung transplant success and affects up to 60% of patients surviving 5 years. The role of complement in causing acute microvascular loss and ischemia during rejection has recently been examined using the mouse orthotopic tracheal transplantation; this is an ideal model for parsing the role of airway vasculature in rejection. Prior to the development of airway fibrosis in rejecting tracheal allografts, C3 deposits on the vascular endothelium just as tissue hypoxia is first detected. With the eventual destruction of vessels, microvascular blood flow to the graft stops altogether for several days. Complement deficiency and complement inhibition lead to markedly improved tissue oxygenation in transplants, diminished airway remodeling, and accelerated vascular repair. CD4+ T cells and antibody-dependent complement activity independently mediate vascular destruction and sustained tissue ischemia during acute rejection. Consequently, interceding against complement-mediated microvascular injury with adjunctive therapy during acute rejection episodes, in addition to standard immunosuppression which targets CD4+ T cells, may help prevent the subsequent development of chronic rejection.
Collapse
|
22
|
Khan MA, Dhillon G, Jiang X, Lin YC, Nicolls MR. New methods for monitoring dynamic airway tissue oxygenation and perfusion in experimental and clinical transplantation. Am J Physiol Lung Cell Mol Physiol 2012; 303:L861-9. [PMID: 23002078 DOI: 10.1152/ajplung.00162.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A dual circulation, supplied by bronchial and pulmonary artery-derived vessels, normally perfuses the airways from the trachea to the terminal bronchioles. This vascular system has been highly conserved through mammalian evolution and is disrupted at the time of lung transplantation. In most transplant centers, this circulation is not restored. The Papworth Hospital Autopsy study has revealed that an additional attrition of periairway vessels is associated with the development of chronic rejection, otherwise known as the bronchiolitis obliterans syndrome (BOS). Experimental studies subsequently demonstrated that airway vessels are subject to alloimmune injury and that the loss of a functional microvascular system identifies allografts that cannot be rescued with immunosuppressive therapy. Therefore, surgical and medical strategies, which preserve the functionality of the existent vasculature in lung transplant patients, may conceivably limit the incidence of BOS. Given these unique anatomic and physiological considerations, there is an emerging rationale to better understand the perfusion and oxygenation status of airways in transplanted lungs. This article describes novel methodologies, some newly developed by our group, for assessing airway tissue oxygenation and perfusion in experimental and clinical transplantation.
Collapse
|
23
|
Khan MA, Jiang X, Dhillon G, Beilke J, Holers VM, Atkinson C, Tomlinson S, Nicolls MR. CD4+ T cells and complement independently mediate graft ischemia in the rejection of mouse orthotopic tracheal transplants. Circ Res 2011; 109:1290-301. [PMID: 21998328 DOI: 10.1161/circresaha.111.250167] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
RATIONALE While microvascular injury is associated with chronic rejection, the cause of tissue ischemia during alloimmune injury is not yet elucidated. OBJECTIVE We investigated the contribution of T lymphocytes and complement to microvascular injury-associated ischemia during acute rejection of mouse tracheal transplants. METHODS AND RESULTS Using novel techniques to assess microvascular integrity and function, we evaluated how lymphocyte subsets and complement specifically affect microvascular perfusion and tissue oxygenation in MHC-mismatched transplants. To characterize T cell effects on microvessel loss and recovery, we transplanted functional airway grafts in the presence and absence of CD4(+) and CD8(+) T cells. To establish the contribution of complement-mediated injury to the allograft microcirculation, we transplanted C3-deficient and C3-inhibited recipients. We demonstrated that CD4(+) T cells and complement are independently sufficient to cause graft ischemia. CD8(+) T cells were required for airway neovascularization to occur following CD4-mediated rejection. Activation of antibody-dependent complement pathways mediated tissue ischemia even in the absence of cellular rejection. Complement inhibition by CR2-Crry attenuated graft hypoxia, complement/antibody deposition on vascular endothelium and promoted vascular perfusion by enhanced angiogenesis. Finally, there was a clear relationship between the burden of tissue hypoxia (ischemia×time duration) and the development of subsequent airway remodeling. CONCLUSIONS These studies demonstrated that CD4(+) T cells and complement operate independently to cause transplant ischemia during acute rejection and that sustained ischemia is a precursor to chronic rejection.
Collapse
Affiliation(s)
- Mohammad A Khan
- Veterans Affairs Palo Alto Health Care System/Stanford University School of Medicine, CA 94304, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Jiang X, Khan MA, Tian W, Beilke J, Natarajan R, Kosek J, Yoder MC, Semenza GL, Nicolls MR. Adenovirus-mediated HIF-1α gene transfer promotes repair of mouse airway allograft microvasculature and attenuates chronic rejection. J Clin Invest 2011; 121:2336-49. [PMID: 21606594 DOI: 10.1172/jci46192] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 03/30/2011] [Indexed: 01/27/2023] Open
Abstract
Chronic rejection, manifested as small airway fibrosis (obliterative bronchiolitis [OB]), is the main obstacle to long-term survival in lung transplantation. Recent studies demonstrate that the airways involved in a lung transplant are relatively hypoxic at baseline and that OB pathogenesis may be linked to ischemia induced by a transient loss of airway microvasculature. Here, we show that HIF-1α mediates airway microvascular repair in a model of orthotopic tracheal transplantation. Grafts with a conditional knockout of Hif1a demonstrated diminished recruitment of recipient-derived Tie2⁺ angiogenic cells to the allograft, impaired repair of damaged microvasculature, accelerated loss of microvascular perfusion, and hastened denudation of epithelial cells. In contrast, graft HIF-1α overexpression induced via an adenoviral vector prolonged airway microvascular perfusion, preserved epithelial integrity, extended the time window for the graft to be rescued from chronic rejection, and attenuated airway fibrotic remodeling. HIF-1α overexpression induced the expression of proangiogenic factors such as Sdf1, Plgf, and Vegf, and promoted the recruitment of vasoreparative Tie2⁺ cells. This study demonstrates that a therapy that enhances vascular integrity during acute rejection may promote graft health and prevent chronic rejection.
Collapse
Affiliation(s)
- Xinguo Jiang
- Department of Medicine, VA Palo Alto Health Care System/Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Setoguchi K, Schenk AD, Ishii D, Hattori Y, Baldwin WM, Tanabe K, Fairchild RL. LFA-1 antagonism inhibits early infiltration of endogenous memory CD8 T cells into cardiac allografts and donor-reactive T cell priming. Am J Transplant 2011; 11:923-35. [PMID: 21466654 PMCID: PMC3215941 DOI: 10.1111/j.1600-6143.2011.03492.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alloreactive memory T cells are present in virtually all transplant recipients due to prior sensitization or heterologous immunity and mediate injury undermining graft outcome. In mouse models, endogenous memory CD8 T cells infiltrate MHC-mismatched cardiac allografts and produce IFN-γ in response to donor class I MHC within 24 h posttransplant. The current studies analyzed the efficacy of anti-LFA-1 mAb to inhibit early CD8 T cell cardiac allograft infiltration and activation. Anti-LFA-1 mAb given to C57BL/6 6 (H-2(b)) recipients of A/J (H-2(a)) heart grafts on days -1 and 0 completely inhibited CD8 T cell allograft infiltration, markedly decreased neutrophil infiltration and significantly reduced intragraft expression levels of IFN-γ-induced genes. Donor-specific T cells producing IFN-γ were at low/undetectable numbers in spleens of anti-LFA-1 mAb treated recipients until day 21. These effects combined to promote substantial prolongation (from day 8 to 27) in allograft survival. Delaying anti-LFA-1 mAb treatment until days 3 and 4 posttransplant did not inhibit early memory CD8 T cell infiltration and proliferation within the allograft. These data indicate that peritransplant anti-LFA-1 mAb inhibits early donor-reactive memory CD8 T cell allograft infiltration and inflammation suggesting an effective strategy to attenuate the negative effects of heterologous immunity in transplant recipients.
Collapse
Affiliation(s)
- Kiyoshi Setoguchi
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Austin D. Schenk
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Daisuke Ishii
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Yusuke Hattori
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - William M. Baldwin
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Robert L. Fairchild
- Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
26
|
Päiväniemi OE, Musilova P, Raivio PM, Maasilta PK, Alho HS, Rubes J, Aittomäki K, Salminen US. Ingraft chimerism in lung transplantation--a study in a porcine model of obliterative bronchiolitis. Respir Res 2011; 12:56. [PMID: 21521503 PMCID: PMC3111361 DOI: 10.1186/1465-9921-12-56] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 04/26/2011] [Indexed: 11/19/2022] Open
Abstract
Background Bronchial epithelium is a target of the alloimmune response in lung transplantation, and intact epithelium may protect allografts from rejection and obliterative bronchiolitis (OB). Herein we study the influence of chimerism on bronchial epithelium and OB development in pigs. Methods A total of 54 immunosuppressed and unimmunosuppressed bronchial allografts were serially obtained 2-90 days after transplantation. Histology (H&E) was assessed and the fluorescence in situ hybridization (FISH) method for Y chromosomes using pig-specific DNA-label was used to detect recipient derived cells in graft epithelium and bronchial wall, and donor cell migration to recipient organs. Ingraft chimerism was studied by using male recipients with female donors, whereas donor cell migration to recipient organs was studied using female recipients with male donors. Results Early appearance of recipient-derived cells in the airway epithelium appeared predictive of epithelial destruction (R = 0.610 - 0.671 and p < 0.05) and of obliteration of the bronchial lumen (R = 0.698 and p < 0.01). All allografts with preserved epithelium showed epithelial chimerism throughout the follow-up. Antirejection medication did not prevent, but delayed the appearance of Y chromosome positive cells in the epithelium (p < 0.05), or bronchial wall (p < 0.05). Conclusions In this study we demonstrate that early appearance of Y chromosomes in the airway epithelium predicts features characteristic of OB. Chimerism occurred in all allografts, including those without features of OB. Therefore we suggest that ingraft chimerism may be a mechanism involved in the repair of alloimmune-mediated tissue injury after transplantation.
Collapse
Affiliation(s)
- Outi E Päiväniemi
- Department of Cardiothoracic Surgery, Helsinki University Hospital, University of Helsinki, P.O. Box 340, 00029 HUS, Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Sustained local application of epidermal growth factor to accelerate reepithelialization of tracheal grafts. J Thorac Cardiovasc Surg 2010; 140:209-15. [DOI: 10.1016/j.jtcvs.2009.10.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2009] [Revised: 10/13/2009] [Accepted: 10/25/2009] [Indexed: 11/24/2022]
|
28
|
Babu AN, Murakawa T, Thurman JM, Miller EJ, Henson PM, Zamora MR, Voelkel NF, Nicolls MR. Microvascular destruction identifies murine allografts that cannot be rescued from airway fibrosis. J Clin Invest 2008; 117:3774-85. [PMID: 18060031 DOI: 10.1172/jci32311] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 09/12/2007] [Indexed: 11/17/2022] Open
Abstract
Small airway fibrosis (bronchiolitis obliterans syndrome) is the primary obstacle to long-term survival following lung transplantation. Here, we show the importance of functional microvasculature in the prevention of epithelial loss and fibrosis due to rejection and for the first time, relate allograft microvascular injury and loss of tissue perfusion to immunotherapy-resistant rejection. To explore the role of alloimmune rejection and airway ischemia in the development of fibroproliferation, we used a murine orthotopic tracheal transplant model. We determined that transplants were reperfused by connection of recipient vessels to donor vessels at the surgical anastomosis site. Microcirculation through the newly formed vascular anastomoses appeared partially dependent on VEGFR2 and CXCR2 pathways. In the absence of immunosuppression, the microvasculature in rejecting allografts exhibited vascular complement deposition, diminished endothelial CD31 expression, and absent perfusion prior to the onset of fibroproliferation. Rejecting grafts with extensive endothelial cell injury were refractory to immunotherapy. After early microvascular loss, neovascularization was eventually observed in the membranous trachea, indicating a reestablishment of graft perfusion in established fibrosis. One implication of this study is that bronchial artery revascularization at the time of lung transplantation may decrease the risk of subsequent airway fibrosis.
Collapse
Affiliation(s)
- Ashok N Babu
- Department of Surgery, University of Colorado at Denver and Health Sciences Center, Denver, Colorado, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Panoskaltsis-Mortari A, Tram KV, Price AP, Wendt CH, Blazar BR. A new murine model for bronchiolitis obliterans post-bone marrow transplant. Am J Respir Crit Care Med 2007; 176:713-23. [PMID: 17575098 PMCID: PMC1994233 DOI: 10.1164/rccm.200702-335oc] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Accepted: 06/14/2007] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Bronchiolitis obliterans (BO) is a major problem in lung transplantation and is also part of the spectrum of late-onset pulmonary complications that can occur after hematopoietic stem cell transplant. Better mouse models are needed to study the onset of this disease so that therapeutic interventions can be developed. OBJECTIVES Our goal was to develop a BO mouse model. METHODS Recipients were lethally conditioned and given a rescue dose of T-cell-depleted, allogeneic bone marrow (BM) supplemented with a sublethal dose of allogeneic T cells. MEASUREMENTS AND MAIN RESULTS At 2 months post-BM transplant, the lungs had extensive perivascular and peribronchiolar inflammation consisting of CD4(+) T cells, CD8(+) T cells, B cells, macrophages, neutrophils, and fibroblasts. In contrast to the acute model, histology showed airway obstruction consistent with BO. Epithelial cells of airways in the early stages of occlusion exhibited changes in expression of cytokeratins. Although the lung had severe allogeneic BM transplant-mediated disease, there was only mild to moderate graft-versus-host disease in liver, colon, skin, and spleen. High wet/dry weight ratios and elevated hydroxyproline were seen, consistent with pulmonary edema and fibrosis. Mice with BO exhibited high airway resistance and low compliance. Increases in many inflammatory mediators in the lungs of mice that develop BO were seen early post-transplant and not later at the time of BO. CONCLUSIONS This new mouse model will be useful for the study of BO associated with late post-hematopoietic stem cell transplant onset and chronic graft-versus-host disease, which also leads to poor outcome in the lung transplant setting.
Collapse
Affiliation(s)
- Angela Panoskaltsis-Mortari
- University of Minnesota, Department of Pediatrics, Division of Hematology/Oncology, Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
30
|
Lande JD, Patil J, Li N, Berryman TR, King RA, Hertz MI. Novel insights into lung transplant rejection by microarray analysis. Ann Am Thorac Soc 2007; 4:44-51. [PMID: 17202291 PMCID: PMC2647614 DOI: 10.1513/pats.200605-110jg] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Gene expression microarrays can estimate the prevalence of mRNA for thousands of genes in a small sample of cells or tissue. Organ transplant researchers are increasingly using microarrays to identify specific patterns of gene expression that predict and characterize acute and chronic rejection, and to improve our understanding of the mechanisms underlying organ allograft dysfunction. We used microarrays to assess gene expression in bronchoalveolar lavage cell samples from lung transplant recipients with and without acute rejection on simultaneous lung biopsies. These studies showed increased expression during acute rejection of genes involved in inflammation, apoptosis, and T-cell activation and proliferation. We also studied gene expression during the evolution of airway obliteration in a murine heterotopic tracheal transplant model of chronic rejection. These studies demonstrated specific patterns of gene expression at defined time points after transplantation in allografts, whereas gene expression in isografts reverted back to that of native tracheas within 2 wk after transplantation. These studies demonstrate the potential power of microarrays to identify biomarkers of acute and chronic lung rejection. The application of new genetic, genomic, and proteomic technologies is in its infancy, and the microarray-based studies described here are clearly only the beginning of their application to lung transplantation. The massive amount of data generated per tissue or cell sample has spawned an outpouring of invention in the bioinformatics field, which is developing methodologies to turn data into meaningful and reproducible clinical and mechanistic inferences.
Collapse
Affiliation(s)
- Jeffrey D Lande
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota 55405, USA
| | | | | | | | | | | |
Collapse
|
31
|
Lu BS, Yu AD, Zhu X, Garrity ER, Vigneswaran WT, Bhorade SM. Sequential gene expression profiling in lung transplant recipients with chronic rejection. Chest 2006; 130:847-54. [PMID: 16963685 DOI: 10.1378/chest.130.3.847] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
STUDY OBJECTIVES Chronic allograft rejection is the leading cause of morbidity and mortality for long-term survivors of lung transplantation. Previous studies have implicated only isolated genes in the development of chronic rejection and have not examined multiple pathways in an individual concurrently. Using microarray technology, we identified and compared gene expression profiling in lung transplant recipients with and without chronic rejection, and follow sequential expression of genes differentially expressed between the two groups. DESIGN Prospective, cohort study. SETTING Single lung transplant center. PATIENTS OR PARTICIPANTS Eleven transplant recipients with chronic rejection were matched with 9 control transplant recipients. INTERVENTIONS All recipients underwent surveillance bronchoscopies at predetermined times to rule out infection and/or acute rejection. Gene expression profiling was obtained from hybridizing BAL fluid cell RNA to a 96-gene microarray. MEASUREMENTS AND RESULTS Fifteen genes were found to be significantly differentially expressed between the two patient groups, and they are involved in inflammatory, fibrotic, and apoptotic pathways. Temporal expression of the significant genes demonstrated a change in their levels at the onset of chronic rejection, with normalization to prerejection levels as rejection continued. CONCLUSIONS We conclude that microarray technology is valuable in studying the mechanism of chronic lung rejection, and the expression of genes in multiple pathways is elevated in patients with chronic lung rejection.
Collapse
Affiliation(s)
- Brandon S Lu
- Department of Neurology (Dr. Lu), Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | | | | |
Collapse
|
32
|
Babu AN, Nicolls MR. Critical pathways leading to obliterative bronchiolitis in lung allografts. Curr Opin Organ Transplant 2006. [DOI: 10.1097/01.mot.0000244650.00717.9c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Affiliation(s)
- Marc Estenne
- Chest Service, Erasme University Hospital, 808 Route de Lennik, B-1070 Brussels, Belgium.
| | | |
Collapse
|
34
|
Abstract
Leukocyte function associated antigen-1 (LFA-1) was one of the earliest of cell-surface molecules identified by monoclonal antibodies generated against leukocyte immunogens. This integrin heterodimer is perhaps best known as a classic adhesion molecule facilitating the interaction between T cells and antigen-presenting cells. However, varied studies indicate that LFA-1 has multi-faceted roles in the immune response including adhesion, activation and trafficking of leukocyte populations. While there has been long-standing interest in LFA-1 as a therapeutic target for regulating immunity, anti-LFA-1 therapy is still not a first-line indication for any clinical condition. Antagonism of LFA-1 with monoclonal antibodies, either alone or in combination with other agents, can result in regulatory tolerance in vivo. Furthermore, new generation humanized anti-LFA-1 monoclonal antibodies (Efalizumab) show at least modest promise for continued application in clinical trials. Thus, anti-LFA-1 forms a potential, but still largely unexploited, immunotherapy which may find its greatest application as an agent which augments other therapies.
Collapse
Affiliation(s)
- M R Nicolls
- Department of Medicine, University of Colorado Health Science Center, Denver, Colorado, USA.
| | | |
Collapse
|
35
|
Douglas IS, Nicolls MR. Chemokine-mediated angiogenesis: an essential link in the evolution of airway fibrosis? J Clin Invest 2005; 115:1133-6. [PMID: 15864341 PMCID: PMC1087192 DOI: 10.1172/jci25193] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis may be an important factor in the development of fibrotic lung disease. Prior studies have strongly suggested a role for angiogenic vascular remodeling in pulmonary fibrosis, and emerging evidence indicates that new vessel formation is critical in airway fibrosis. Bronchiolitis obliterans syndrome is a fibrotic occlusion of distal airways that is largely responsible for the morbidity and mortality of patients after lung transplantation. In this issue, Belperio et al. demonstrate a role for CXC chemokine receptor 2 in the regulation of angiogenesis-mediated airway fibroproliferation. By integrating an understanding of neovascularization into the study of events that occur between inflammation and fibrosis, it becomes increasingly possible to rationally design therapies that can halt conditions of maladaptive fibrosis.
Collapse
Affiliation(s)
- Ivor S Douglas
- Department of Medicine, Pulmonary Sciences and Critical Care Medicine and Denver Health Medical Center, University of Colorado Health Sciences Center, Denver, 80262, USA.
| | | |
Collapse
|