1
|
Vorlat A, van Eijk J, Wiersma S, Smid L, Depooter S, Paelinck B, Guerti K, Peeters B, Sturkenboom N, Van Craenenbroeck E, Heidbuchel H, Van De Heyning C. Clinical determinants and biomarkers associated with cardiac fibrosis after heart transplantation as assessed by magnetic resonance: Size matters. IJC HEART & VASCULATURE 2024; 54:101479. [PMID: 39221115 PMCID: PMC11365389 DOI: 10.1016/j.ijcha.2024.101479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Background Cardiac fibrosis is increasingly recognized as a marker of worse outcomes in long-term follow-up after heart transplantation (HTX). We investigated the clinical determinants and biomarkers of focal and interstitial cardiac fibrosis as assessed with cardiac magnetic resonance (CMR). Methods Consecutive HTX recipients underwent CMR with late gadolinium enhancement for focal myocardial fibrosis and T1 mapping for interstitial fibrosis. We calculated the correlations of these findings with clinical parameters, history, biomarkers of fibrosis (B-type natriuretic peptide (BNP), growth differentiation factor-15, galectin-3 and soluble ligand ST2) and echocardiography. Results Forty-eight HTX patients were included: median age 63 ± 13 years, 11 ± 6 years after heart transplantation. Only donor weight (p 0.044) and the rate of a > 30 % mismatch between donor and recipient weight (p 0.02) were significantly different in patients with vs. without late LGE. Extracellular volume (ECV) was correlated with the weight mismatch between donor and recipient (r = 0.32, p 0.04), resulting in a higher ECV for oversized donors. BNP was the only biomarker of the four studied that was correlated with interstitial fibrosis as assessed by ECV (r = 0.35, p 0.04). T1 relaxation time was correlated with treated acute cellular rejection grade ≥ 2 (ISHLT grading) (r = 0.34, p 0.02). Conclusion Both focal and interstitial fibrosis, as determined by CMR, after heart transplantation are correlated with donor and recipient weight mismatch. BNP was the only biomarker clinically relevant to interstitial cardiac fibrosis.
Collapse
Affiliation(s)
- Anne Vorlat
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Belgium
| | | | | | - Leroy Smid
- Medicine, University of Antwerp, Belgium
| | | | - Bernard Paelinck
- Department Cardiac Surgery, Antwerp University Hospital, University of Antwerp, Belgium
| | - Khadija Guerti
- Department of Clinical Chemistry, Antwerp University Hospital, University of Antwerp, Belgium
| | - Bart Peeters
- Department of Clinical Chemistry, Antwerp University Hospital, University of Antwerp, Belgium
| | - Nicole Sturkenboom
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Belgium
| | | | - Hein Heidbuchel
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Belgium
| | | |
Collapse
|
2
|
Zhou X, Xu Q, Li W, Dong N, Stomberski C, Narla G, Lin Z. Protein Phosphatase 2A Activation Promotes Heart Transplant Acceptance in Mice. Transplantation 2024; 108:e36-e48. [PMID: 38126420 PMCID: PMC10922415 DOI: 10.1097/tp.0000000000004832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
BACKGROUND Although heart transplantation is the definitive treatment for heart failure in eligible patients, both acute and chronic transplant rejection frequently occur. Protein phosphatase 2A (PP2A) activity is critical in maintaining tissue and organ homeostasis. In this study, we evaluated the effect of a novel class of small molecule activators of PP2A (SMAPs) on allograft rejection in a mouse heterotopic heart transplantation model. METHODS Recipient mice were administered with DT-061 (a pharmaceutically optimized SMAP) or vehicle by oral gavage beginning 1 d after transplantation. Histological and immunofluorescence analyses were performed to examine allograft rejection. Regulatory T cells (Treg) from recipient spleens were subjected to flow cytometry and RNA sequencing analysis. Finally, the effect of DT-061 on smooth muscle cells (SMCs) migration and proliferation was assessed. RESULTS DT-061 treatment prolonged cardiac allograft survival. SMAPs effectively suppressed the inflammatory immune response while increasing Treg population in the allografts, findings corroborated by functional analysis of RNA sequencing data derived from Treg of treated splenic tissues. Importantly, SMAPs extended immunosuppressive agent cytotoxic T lymphocyte-associated antigen-4-Ig-induced cardiac transplantation tolerance and allograft survival. SMAPs also strongly mitigated cardiac allograft vasculopathy as evidenced by a marked reduction of neointimal hyperplasia and SMC proliferation. Finally, our in vitro studies implicate suppression of MEK/ERK pathways as a unifying mechanism for the effect of PP2A modulation in Treg and SMCs. CONCLUSIONS PP2A activation prevents cardiac rejection and prolongs allograft survival in a murine model. Our findings highlight the potential of PP2A activation in improving alloengraftment in heart transplantation.
Collapse
Affiliation(s)
- Xianming Zhou
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xu
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Cardiovascular Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Wangzi Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Colin Stomberski
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Goutham Narla
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhiyong Lin
- Cardiology Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
3
|
Patrichi G, Patrichi A, Satala CB, Sin AI. Matrix Metalloproteinases and Heart Transplantation-A Pathophysiological and Clinical View. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1295. [PMID: 37512106 PMCID: PMC10383867 DOI: 10.3390/medicina59071295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
Heart transplantation is undergoing a continuous development, with rates of success increasing substantially due to advances in immunosuppressive therapy and surgical techniques. The most worrying complication occurring after cardiac transplantation is graft rejection, a phenomenon that is much affected by matrix metalloproteinases (MMPs), with the role of these proteases in the cardiac remodeling process being well established in the literature. A detailed investigation of the association between MMPs and cardiac rejection is necessary for the future development of more targeted therapies in transplanted patients, and to discover prognostic serum and immunohistochemical markers that will lead to more organized therapeutic management in these patients. The aim of this review is therefore to highlight the main MMPs relevant to cardiovascular pathology, with particular emphasis on those involved in complications related to heart transplantation, including cardiac graft rejection.
Collapse
Affiliation(s)
- Gabriela Patrichi
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- Department of Pathology, Clinical County Emergency Hospital, 540136 Targu Mures, Romania
| | - Andrei Patrichi
- Department of Pathology, Clinical County Emergency Hospital, 540136 Targu Mures, Romania
| | - Catalin-Bogdan Satala
- Department of Pathology, Clinical County Emergency Hospital, 540136 Targu Mures, Romania
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Anca Ileana Sin
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
- Department of Pathology, Clinical County Emergency Hospital, 540136 Targu Mures, Romania
| |
Collapse
|
4
|
Hao CL, Lin HL, Cheng PW, Tu YC, Yeh BC, Wu BN, Shen KP. Efficiency comparison of an isoeugenol-derivated compound, eugenosedin-A, with glibenclamide and pioglitazone in protecting cardiovascular dysfunction of diabetic SHR. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2023; 25:471-483. [PMID: 35852140 DOI: 10.1080/10286020.2022.2096597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/28/2022] [Indexed: 06/15/2023]
Abstract
This study was to investigate three agents possible protective effect against DM-induced cardiovascular dysfunction in spontaneously hypertensive rats (SHR). Control group was fed normal diet, DM group was injected with STZ/NA and fed high fat diet (HFD), and treatment groups were given STZ/NA, fed HFD, and then oral gavaged with eugenosedin-A (Eu-A), glibenclamide (Gli), or pioglitazone (Pio) 5 mg/kg/per day for 4-week, respectively. Eu-A, Gli, and Pio clearly ameliorated the changes of body weight, cardiac weight, and the biochemical parameters, cardiovascular disorders and inflammation. Like Gli and Pio, Eu-A may be effectively to control DM and the cardiovascular dysfunction.
Collapse
Affiliation(s)
- Chi-Long Hao
- Division of Cardiology, Department of Internal Medicine, Pingtung Christian Hospital, Pingtung 90059, Taiwan
| | - Hui-Li Lin
- Graduate Institute of Food Culture and Innovation, National Kaohsiung University of Hospitality and Tourism, Kaohsiung 81271, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Yi-Chen Tu
- Graduate Institute of Food Culture and Innovation, National Kaohsiung University of Hospitality and Tourism, Kaohsiung 81271, Taiwan
| | - Bor-Chun Yeh
- Graduate Institute of Food Culture and Innovation, National Kaohsiung University of Hospitality and Tourism, Kaohsiung 81271, Taiwan
| | - Bin-Nan Wu
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Kuo-Ping Shen
- Department of Nursing, Meiho University, Pingtung 91202, Taiwan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| |
Collapse
|
5
|
Tang PC, Haft JW, Lei I, Wang Z, Chen YE, Abou El Ela A, Wu X, Pitt B, Aaronson KD, Pagani FD. Impact of donor blood type on outcomes after prolonged allograft ischemic times. J Thorac Cardiovasc Surg 2022; 164:981-993.e8. [PMID: 33558115 PMCID: PMC11170339 DOI: 10.1016/j.jtcvs.2020.12.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/29/2020] [Accepted: 12/23/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE The study objective was to determine the influence of allograft ischemic time on heart transplant outcomes among ABO donor organ types given limited prior reports of its survival impact. METHODS We identified 32,454 heart transplants (2000-2016) from the United Network for Organ Sharing database. Continuous and categoric variables were analyzed by parametric and nonparametric testing. Survival was determined using log-rank or Cox regression tests. Propensity matching adjusted for preoperative variables. RESULTS By comparing allograft ischemic time less than 4 hours (n = 6579) with 4 hours or more (n = 25,875), the hazard ratios for death at 15 years after prolonged ischemic time (≥4 hours) for blood types O, A, B, and AB were 1.106 (P < .001), 1.062 (P < .001), 1.059 (P = .062), and 1.114 (P = .221), respectively. Unadjusted data demonstrated higher mortality for transplantation of O versus non-O donor hearts for ischemic time 4 hours or more (hazard ratio, 1.164; P < .001). After propensity matching, O donor hearts continued to have worse survival if preserved for 4 hours or more (hazard ratio, 1.137, P = .008), but not if ischemic time was less than 4 hours (hazard ratio, 1.042, P = .113). In a matched group with 4 hours or more of ischemic time, patients receiving O donor organs were more likely to experience death from primary graft dysfunction (2.5% vs 1.7%, P = .052) and chronic allograft rejection (1.9% vs 1.1%, P = .021). No difference in death from primary graft dysfunction or chronic allograft rejection was seen with less than 4 hours of ischemic time (P > .150). CONCLUSIONS Compared with non-O donor hearts, transplantation with O donor hearts with ischemic time 4 hours or more leads to worse survival, with higher rates of primary graft dysfunction and chronic rejection. Caution should be practiced when considering donor hearts with the O blood type when anticipating extended cold ischemic times.
Collapse
Affiliation(s)
- Paul C Tang
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich.
| | - Jonathan W Haft
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich
| | - Ienglam Lei
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich
| | - Zhong Wang
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich
| | - Y Eugene Chen
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich; Division of Cardiovascular Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Ashraf Abou El Ela
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich
| | - Xiaoting Wu
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich
| | - Bertram Pitt
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Keith D Aaronson
- Division of Cardiovascular Medicine, Department of Internal Medicine, Ann Arbor, Mich
| | - Francis D Pagani
- Department of Cardiac Surgery, University of Michigan Frankel Cardiovascular Center, Ann Arbor, Mich
| |
Collapse
|
6
|
Raghu VK, Sico R, Rudolph JA, Mazariegos GV, Squires J, Squires JE. Sarcopenia prevalence in pediatric intestinal transplant recipients: Implications on post-transplant outcomes. Pediatr Transplant 2022; 26:e14256. [PMID: 35187765 DOI: 10.1111/petr.14256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/14/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Sarcopenia has been associated with poor surgical outcomes but has not been studied in pediatric intestinal transplantation. We aimed to determine sarcopenia prevalence in intestinal transplant recipients and the association of sarcopenia with outcomes. METHODS We performed a cross-sectional retrospective chart review of intestinal transplant recipients from 2000-present. We estimated total psoas muscle area (tPMA) at L3-L4 and L4-L5 from computed tomography scans prior to or in the immediate peri-operative period. Sarcopenia was defined by tPMA below the 5th percentile for age and sex. We built a Cox-proportional hazards model to determine the association between sarcopenia and patient and graft survival. RESULTS Of the 56 intestinal transplant recipients included, 36 (64%) were sarcopenic. Graft survival was 79% at one year and 59% at five years. Overall patient survival was 86% at one year and 76% at five years. Peri-transplant sarcopenia was associated with improved graft survival (Hazard ratio 0.42, 95% confidence interval: 0.20-0.88) but not overall survival (Hazard ratio 0.47, 95% confidence interval: 0.19-1.20). CONCLUSIONS In this first report of sarcopenia in pediatric intestinal transplant, we found a high sarcopenia prevalence without an association with worse outcomes. The potential improved graft survival in sarcopenic patients along with underlying mechanisms warrant further exploration.
Collapse
Affiliation(s)
- Vikram K Raghu
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, UPMC Children's Hospital Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rita Sico
- Department of Pediatric Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey A Rudolph
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, UPMC Children's Hospital Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - George V Mazariegos
- Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Judy Squires
- Department of Pediatric Radiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James E Squires
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, UPMC Children's Hospital Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Peyster EG, Janowczyk A, Swamidoss A, Kethireddy S, Feldman MD, Margulies KB. Computational Analysis of Routine Biopsies Improves Diagnosis and Prediction of Cardiac Allograft Vasculopathy. Circulation 2022; 145:1563-1577. [PMID: 35405081 DOI: 10.1161/circulationaha.121.058459] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Cardiac allograft vasculopathy (CAV) is a leading cause of morbidity and mortality for heart transplant recipients. While clinical risk factors for CAV have been established, no personalized prognostic test exists to confidently identify patients at high vs. low risk of developing aggressive CAV. The aim of this investigation was to leverage computational methods for analyzing digital pathology images from routine endomyocardial biopsies (EMB) to develop a precision medicine tool for predicting CAV years before overt clinical presentation. Methods: Clinical data from 1-year post-transplant was collected on 302 transplant recipients from the University of Pennsylvania, including 53 'early CAV' patients and 249 'no-CAV' controls. This data was used to generate a 'clinical model' (ClinCAV-Pr) for predicting future CAV development. From this cohort, n=183 archived EMBs were collected for CD31 and modified trichrome staining and then digitally scanned. These included 1-year post-transplant EMBs from 50 'early CAV' patients and 82 no-CAV patients, as well as 51 EMBs from 'disease control' patients obtained at the time of definitive coronary angiography confirming CAV. Using biologically-inspired, hand-crafted features extracted from digitized EMBs, quantitative histologic models for differentiating no-CAV from disease controls (HistoCAV-Dx), and for predicting future CAV from 1-year post-transplant EMBs were developed (HistoCAV-Pr). The performance of histologic and clinical models for predicting future CAV (i.e. HistoCAV-Pr and ClinCAV-Pr, respectively) were compared in a held-out validation set, before being combined to assess the added predictive value of an integrated predictive model (iCAV-Pr). Results: ClinCAV-Pr achieved modest performance on the independent test set, with area under the receiver operating curve (AUROC) of 0.70. The HistoCAV-Dx model for diagnosing CAV achieved excellent discrimination, with an AUROC of 0.91, while HistoCAV-Pr model for predicting CAV achieved good performance with an AUROC of 0.80. The integrated iCAV-Pr model achieved excellent predictive performance, with an AUROC of 0.93 on the held-out test set. Conclusions: Prediction of future CAV development is greatly improved by incorporation of computationally extracted histologic features. These results suggest morphologic details contained within regularly obtained biopsy tissue have the potential to enhance precision and personalization of treatment plans for post-heart transplant patients.
Collapse
Affiliation(s)
- Eliot G Peyster
- Cardiovascular Research Institute (E.G.P., K.B.M.), University of Pennsylvania, Philadelphia
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH (A.J., A.S., S.K.)
- Department of Oncology, Lausanne University Hospital and Lausanne University, Switzerland (A.J.)
| | - Abigail Swamidoss
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH (A.J., A.S., S.K.)
| | - Samhith Kethireddy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH (A.J., A.S., S.K.)
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine (M.D.F.), University of Pennsylvania, Philadelphia
| | - Kenneth B Margulies
- Cardiovascular Research Institute (E.G.P., K.B.M.), University of Pennsylvania, Philadelphia
| |
Collapse
|
8
|
Abstract
Purpose of Review IL-6 is a pleiotropic, pro-inflammatory cytokine that plays an integral role in the development of acute and chronic rejection after solid organ transplantation. This article reviews the experimental evidence and current clinical application of IL-6/IL-6 receptor (IL-6R) signaling inhibition for the prevention and treatment of allograft injury. Recent Findings There exists a robust body of evidence linking IL-6 to allograft injury mediated by acute inflammation, adaptive cellular/humoral responses, innate immunity, and fibrosis. IL-6 promotes the acute phase reaction, induces B cell maturation/antibody formation, directs cytotoxic T-cell differentiation, and inhibits regulatory T-cell development. Importantly, blockade of the IL-6/IL-6R signaling pathway has been shown to mitigate its harmful effects in experimental studies, particularly in models of kidney and heart transplant rejection. Currently, available agents for IL-6 signaling inhibition include monoclonal antibodies against IL-6 or IL-6R and janus kinase inhibitors. Recent clinical trials have investigated the use of tocilizumab, an anti-IL-6R mAb, for desensitization and treatment of antibody-mediated rejection (AMR) in kidney transplant recipients, with promising initial results. Further studies are underway investigating the use of alternative agents including clazakizumab, an anti-IL-6 mAb, and application of IL-6 signaling blockade to clinical cardiac transplantation. Summary IL-6/IL-6R signaling inhibition provides a novel therapeutic option for the prevention and treatment of allograft injury. To date, evidence from clinical trials supports the use of IL-6 blockade for desensitization and treatment of AMR in kidney transplant recipients. Ongoing and future clinical trials will further elucidate the role of IL-6 signaling inhibition in other types of solid organ transplantation.
Collapse
|
9
|
Haese NN, Burg JM, Andoh TF, Jones IK, Kreklywich CN, Smith PP, Orloff SL, Streblow DN. Macrophage depletion of CMV latently infected donor hearts ameliorates recipient accelerated chronic rejection. Transpl Infect Dis 2021; 23:e13514. [PMID: 33205500 PMCID: PMC8068575 DOI: 10.1111/tid.13514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/01/2020] [Accepted: 11/08/2020] [Indexed: 12/21/2022]
Abstract
Cytomegalovirus (CMV) infection is linked to acceleration of solid organ transplant vascular sclerosis (TVS) and chronic rejection (CR). Donor latent CMV infection increases cardiac-resident macrophages and T cells leading to increased inflammation, promoting allograft rejection. To investigate the role of cardiac-resident passenger macrophages in CMV-mediated TVS/CR, macrophages were depleted from latently ratCMV (RCMV)-infected donor allografts prior to transplantation. Latently RCMV-infected donor F344 rats were treated with clodronate, PBS, or control liposomes 3 days prior to cardiac transplant into RCMV-naïve Lewis recipients. Clodronate treatment significantly increased graft survival from post-operative day (POD)61 to POD84 and decreased TVS at rejection. To determine the kinetics of the effect of clodronate treatment's effect, a time study revealed that clodronate treatment significantly decreased macrophage infiltration into allograft tissues as early as POD14; altered allograft cytokine/chemokine protein levels, fibrosis development, and inflammatory gene expression profiles. These findings support our hypothesis that increased graft survival as a result of allograft passenger macrophage depletion was in part a result of altered immune response kinetics. Depletion of donor macrophages prior to transplant is a strategy to modulate allograft rejection and reduce TVS in the setting of CMV + donors transplanted into CMV naïve recipients.
Collapse
Affiliation(s)
- Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health Sciences University, Beaverton, OR 97006
| | - Jennifer M. Burg
- Department of Surgery, Oregon Health Sciences University, Portland, OR 97239
| | - Takeshi F. Andoh
- Vaccine and Gene Therapy Institute, Oregon Health Sciences University, Beaverton, OR 97006
| | - Iris K.A. Jones
- Vaccine and Gene Therapy Institute, Oregon Health Sciences University, Beaverton, OR 97006
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health Sciences University, Beaverton, OR 97006
| | - Patricia P. Smith
- Vaccine and Gene Therapy Institute, Oregon Health Sciences University, Beaverton, OR 97006
| | - Susan L. Orloff
- Department of Surgery, Oregon Health Sciences University, Portland, OR 97239
- Department of Molecular Microbiology & Immunology, Oregon Health Sciences University, Portland, OR, USA
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health Sciences University, Beaverton, OR 97006
| |
Collapse
|
10
|
Anti-IL-6 eluting immunomodulatory biomaterials prolong skin allograft survival. Sci Rep 2019; 9:6535. [PMID: 31024011 PMCID: PMC6484015 DOI: 10.1038/s41598-019-42349-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/26/2019] [Indexed: 12/26/2022] Open
Abstract
A primary goal in the management of burn wounds is early wound closure. The use of skin allografts represents a lifesaving strategy for severe burn patients, but their ultimate rejection limits their potential efficacy and utility. IL-6 is a major pleiotropic cytokine which critically links innate and adaptive immune responses. Here, we devised anti-IL-6 receptor eluting gelatin methacryloyl (GelMA) biomaterials (GelMA/anti-IL-6), which were implanted at the interface between the wound beds and skin allografts. Our visible light crosslinked GelMA/anti-IL-6 immunomodulatory biomaterial (IMB) demonstrated a stable kinetic release profile of anti-IL-6. In addition, the incorporation of anti-IL-6 within the GelMA hydrogel had no effect on the mechanical properties of the hydrogels. Using a highly stringent skin transplant model, the GelMA/anti-IL-6 IMB almost doubled the survival of skin allografts. The use of GelMA/anti-IL-6 IMB was far superior to systemic anti-IL-6 receptor treatment in prolonging skin allograft survival. As compared to the untreated control group, skin from the GelMA/anti-IL-6 IMB group contained significantly fewer alloreactive T cells and macrophages. Interestingly, the environmental milieu of the draining lymph nodes (DLNs) of the mice implanted with the GelMA/anti-IL-6 IMB was also considerably less pro-inflammatory. The percentage of CD4+ IFNγ+ cells was much lower in the DLNs of the GelMA/anti-IL-6 IMB group in comparison to the GelMA group. These data highlight the importance of localized immune delivery in prolonging skin allograft survival and its potential utility in treating patients with severe burns.
Collapse
|
11
|
Qiu H, Ji C, Liu W, Wu Y, Lu Z, Lin Q, Xue Z, Liu X, Wu H, Jiang W, Zou C. Chronic Kidney Disease Increases Atrial Fibrillation Inducibility: Involvement of Inflammation, Atrial Fibrosis, and Connexins. Front Physiol 2018; 9:1726. [PMID: 30564139 PMCID: PMC6288485 DOI: 10.3389/fphys.2018.01726] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) causes atrial structural remodeling and subsequently increases the incidence of atrial fibrillation (AF). Atrial connexins and inflammatory responses may be involved in this remodeling process. In this study, nephrectomy was used to produce the CKD rat model. Three months post-nephrectomy, cardiac structure, function and AF vulnerability were quantified using echocardiography and electrophysiology methods. The left atrial tissue was tested for quantification of fibrosis and inflammation, and for the distribution and expression of connexin (Cx) 40 and Cx43. An echocardiography showed that CKD resulted in the left atrial enlargement and left ventricular hypertrophy, but had no functional changes. CKD caused a significant increase in the AF inducible rate (91.11% in CKD group vs. 6.67% in sham group, P < 0.001) and the AF duration [107 (0–770) s in CKD vs. 0 (0–70) s in sham, P < 0.001] with prolonged P-wave duration. CKD induced severe interstitial fibrosis, activated the transforming growth factor-β1/Smad2/3 pathway with a massive extracellular matrix deposition of collagen type I and α-smooth muscle actin, and matured the NLR (nucleotide-binding domain leucine-rich repeat-containing receptor) pyrin domain-containing protein 3 (NLRP3) inflammasome with an inflammatory cascade response. CKD resulted in an increase in non-phosphorylated-Cx43, a decrease in Cx40 and phosphorylated-Cx43, and lateralized the distribution of Cx40 and Cx43 proteins with upregulations of Rac-1, connective tissue growth factor and N-cadherin. These findings implicate the transforming growth factor-β1/Smad2/3, the NLRP3 inflammasome and the connexins as potential mediators of increased AF vulnerability in CKD.
Collapse
Affiliation(s)
- Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chunlan Ji
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yuchi Wu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zhaoyu Lu
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qizhan Lin
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zheng Xue
- Department of Cardiology, Guangzhou Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Huanlin Wu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Department of internal medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Jiang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chuan Zou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
12
|
Li S, Wang S, Murugan R, Al-Khafaji A, Lebovitz DJ, Souter M, Stuart SRN, Kellum JA. Donor biomarkers as predictors of organ use and recipient survival after neurologically deceased donor organ transplantation. J Crit Care 2018; 48:42-47. [PMID: 30172032 DOI: 10.1016/j.jcrc.2018.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/05/2018] [Accepted: 08/14/2018] [Indexed: 02/01/2023]
Abstract
PURPOSE We sought to build prediction models for organ transplantation and recipient survival using both biomarkers and clinical information. MATERIALS AND METHODS We abstracted clinical variables from a previous randomized trial (n = 556) of donor management. In a subset of donors (n = 97), we measured two candidate biomarkers in plasma at enrollment and just prior to explantation. RESULTS Secretory leukocyte protease inhibitor (SLPI) was significant for predicting liver transplantation (C-statistic 0.65 (0.53, 0.78)). SLPI also significantly improved the predictive performance of a clinical model for liver transplantation (integrated discrimination improvement (IDI): 0.090 (0.009, 0.210)). For other organs, clinical variables alone had strong predictive ability (C-statistic >0.80). Recipient 3-years survival was 80.0% (71.9%, 87.0%). Donor IL-6 was significantly associated with recipient 3-years survival (adjusted Hazard Ratio (95%CI): 1.26(1.08, 1.48), P = .004). Neither clinical variables nor biomarkers showed strong predictive ability for 3-year recipient survival. CONCLUSIONS Plasma biomarkers in neurologically deceased donors were associated with organ use. SLPI enhanced prediction within a liver transplantation model, whereas IL-6 before transplantation was significantly associated with recipient 3-year survival. Clinicaltrials.gov: NCT00987714.
Collapse
Affiliation(s)
- Shengnan Li
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Shu Wang
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, United States
| | - Raghavan Murugan
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ali Al-Khafaji
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daniel J Lebovitz
- Department of Critical Care, Akron Children's Hospital, Akron, OH, United States
| | - Michael Souter
- Department of Anesthesiology & Pain Medicine, University of Washington, Harborview Medical Center, Seattle, WA, United States
| | - Susan R N Stuart
- Center for Organ Recovery and Education, Pittsburgh, PA, United States
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; The CRISMA (Clinical Research, Investigation and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| | | |
Collapse
|
13
|
Luo S, Yang M, Jin H, Xu ZQ, Li YF, Xia P, Yang YR, Chen BC, Zhang Y. The role of sildenafil in the development of transplant arteriosclerosis in rat aortic grafts. Am J Transl Res 2017; 9:4914-4924. [PMID: 29218089 PMCID: PMC5714775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
Chronic rejection (CR), which is characterized histologically by progressive graft arteriosclerosis, remains a significant barrier to the long-term survival of a graft. Sildenafil has been shown to protect vascular endothelial cells. In this study, we found that sildenafil significantly reduces the thickness of transplant vascular intima in a rat aortic transplant model. Moreover, sildenafil dramatically decreased the expression of transforming growth factor-β1 (TGF-β1), vascular endothelial growth factor (VEGF), and α-smooth muscle actin (α-SMA) in the grafted aortas and increased the concentrations of cyclic guanosine monophosphate (cGMP) and endothelial nitric oxide synthase (eNOS) in serum. Furthermore, the ratio of regulatory T (Treg) cells and the expression of FoxP3 were increased, and the ratio of Th17 cells was decreased in the sildenafil-treated group. These results demonstrate that sildenafil enhances nitric oxide (NO) signaling by increasing the availability of cGMP, leading to an increase in the ratio of Treg/Th17 cells to attenuate transplant arteriosclerosis in a rat aortic transplant model.
Collapse
Affiliation(s)
- Shuai Luo
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
- Department of Urology, Huangshi Central HospitalHuangshi 435000, Hubei Province, China
| | - Mei Yang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Hao Jin
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Zi-Qiang Xu
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Yi-Fu Li
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Peng Xia
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Yi-Rrong Yang
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Bi-Cheng Chen
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Yan Zhang
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| |
Collapse
|
14
|
Solhjou Z, Uehara M, Bahmani B, Maarouf OH, Ichimura T, Brooks CR, Xu W, Yilmaz M, Elkhal A, Tullius SG, Guleria I, McGrath M, Abdi R. Novel Application of Localized Nanodelivery of Anti-Interleukin-6 Protects Organ Transplant From Ischemia-Reperfusion Injuries. Am J Transplant 2017; 17:2326-2337. [PMID: 28296000 PMCID: PMC5573642 DOI: 10.1111/ajt.14266] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/03/2017] [Accepted: 02/25/2017] [Indexed: 01/25/2023]
Abstract
Ischemia-reperfusion injury (IRI) evokes intragraft inflammatory responses, which markedly augment alloimmune responses against the graft. Understanding the mechanisms underlying these responses is fundamental to develop therapeutic regimens to prevent/ameliorate organ IRI. Here, we demonstrate that IRI results in a marked increase in mitochondrial damage and autophagy in dendritic cells (DCs). While autophagy is a survival mechanism for ischemic DCs, it also augments their production of interleukin (IL)-6. Allograft-derived dendritic cells (ADDCs) lacking autophagy-related gene 5 (Atg5) showed higher death rates posttransplantation. Transplanted ischemic hearts from CD11cCre/Atg5 conditional knockout mice showed marked reduction in intragraft expression of IL-6 compared with controls. To antagonize the effect of IL-6 locally in the heart, we synthesized novel anti-IL-6 nanoparticles with capacity for controlled release of anti-IL-6 over time. Compared with systemic delivery of anti-IL-6, localized delivery of anti-IL-6 significantly reduced chronic rejection with a markedly lower amount administered. Despite improved allograft histology, there were no changes to splenic T cell populations, illustrating the importance of local IL-6 in driving chronic rejection after IRI. These data carry potential clinical significance by identifying an innovative, targeted strategy to manipulate organs before transplantation to diminish inflammation, leading to improved long-term outcomes.
Collapse
Affiliation(s)
- Zhabiz Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Baharak Bahmani
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Omar H. Maarouf
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig R. Brooks
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wanlong Xu
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mine Yilmaz
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abdala Elkhal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Indira Guleria
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Martina McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA,Address correspondence to: Reza Abdi, MD, Transplant Research Center, Brigham and Women's Hospital, 221 Longwood Ave, Boston MA 02115, USA, Tel: 617-732-5259, Fax: 617-732-5254,
| |
Collapse
|
15
|
AS2553627, a novel JAK inhibitor, prevents chronic rejection in rat cardiac allografts. Eur J Pharmacol 2017; 796:69-75. [DOI: 10.1016/j.ejphar.2016.12.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022]
|
16
|
Schiechl G, Hermann FJ, Rodriguez Gomez M, Kutzi S, Schmidbauer K, Talke Y, Neumayer S, Goebel N, Renner K, Brühl H, Karasuyama H, Obata-Ninomiya K, Utpatel K, Evert M, Hirt SW, Geissler EK, Fichtner-Feigl S, Mack M. Basophils Trigger Fibroblast Activation in Cardiac Allograft Fibrosis Development. Am J Transplant 2016; 16:2574-88. [PMID: 26932231 DOI: 10.1111/ajt.13764] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/29/2016] [Accepted: 02/22/2016] [Indexed: 01/25/2023]
Abstract
Fibrosis is a major component of chronic cardiac allograft rejection. Although several cell types are able to produce collagen, resident (donor-derived) fibroblasts are mainly responsible for excessive production of extracellular matrix proteins. It is currently unclear which cells regulate production of connective tissue elements in allograft fibrosis and how basophils, as potential producers of profibrotic cytokines, are involved this process. We studied this question in a fully MHC-mismatched model of heart transplantation with transient depletion of CD4(+) T cells to largely prevent acute rejection. The model is characterized by myocardial infiltration of leukocytes and development of interstitial fibrosis and allograft vasculopathy. Using depletion of basophils, IL-4-deficient recipients and IL-4 receptor-deficient grafts, we showed that basophils and IL-4 play crucial roles in activation of fibroblasts and development of fibrotic organ remodeling. In the absence of CD4(+) T cells, basophils are the predominant source of IL-4 in the graft and contribute to expansion of myofibroblasts, interstitial deposition of collagen and development of allograft vasculopathy. Our results indicated that basophils trigger the production of various connective tissue elements by myofibroblasts. Basophil-derived IL-4 may be an attractive target for treatment of chronic allograft rejection.
Collapse
Affiliation(s)
- G Schiechl
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - F J Hermann
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - M Rodriguez Gomez
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - S Kutzi
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - K Schmidbauer
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Y Talke
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - S Neumayer
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - N Goebel
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - K Renner
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - H Brühl
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - H Karasuyama
- Department of Immune Regulation, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - K Obata-Ninomiya
- Department of Immune Regulation, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - K Utpatel
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - M Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - S W Hirt
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - E K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - S Fichtner-Feigl
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.,RCI Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - M Mack
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany.,RCI Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Kremer J, Muschitz GK, Aumayr K, Moser P, Szabo G, Weymann A, Zuckermann A, Podesser BK. Influence of antithymocyte globulin treatment of brain-dead organ donor on inflammatory response in cardiac grafts: an experimental study in mice. Transpl Int 2016; 29:1329-1336. [PMID: 27571572 DOI: 10.1111/tri.12851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/05/2016] [Accepted: 08/25/2016] [Indexed: 11/27/2022]
Abstract
The expression of proinflammatory cytokines in donor hearts after antithymocyte globulin (ATG) treatment given prior to organ removal was evaluated to analyze changes in inflammatory response. Adult female OF-1 mice were randomized into brain death (BD) groups (BD Control, BD ATG) with or without treatment, and Controls (Control, ATG). BD induction was performed through gradual inflation of an intracranial positioned balloon catheter. At the end of a 6-h observation period, ATG (1 mg/kg BW) was given intravenously. After 45 min, the donor hearts were removed. Proinflammatory markers IL-2 and IL-6 were examined using ELISA and immunohistochemistry staining. After single administration of ATG, the inflammatory reaction in the myocardium showed a significant reduction in IL-2 expression (BD Control vs. BD ATG, P = 0.033). Our investigation showed expected increase in proinflammatory mediators after BD. This increase was abolished by single infusion of ATG, indicated by significant reduction in IL-2 levels in the myocardium. We observed a reduction of IL-6 deposition in media cells in ATG-treated specimens. Further research is necessary to evaluate the role of ATG in donor management considering a potentially positive effect of ATG on IL-2-directed inflammatory response and possible reduction of IL-6-mediated vascular changes.
Collapse
Affiliation(s)
- Jamila Kremer
- Department of Cardiac Surgery, Heart and Marfan Center, University of Heidelberg, Heidelberg, Germany.,Ludwig Boltzmann Cluster for Cardiovascular Research, Core Unit for Biomedical Research, Medical University Vienna, Vienna, Austria
| | - Gabriela K Muschitz
- Ludwig Boltzmann Cluster for Cardiovascular Research, Core Unit for Biomedical Research, Medical University Vienna, Vienna, Austria.,Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria
| | - Klaus Aumayr
- Department of Clinical Pathology, Medical University Vienna, Vienna, Austria
| | - Philipp Moser
- Ludwig Boltzmann Cluster for Cardiovascular Research, Core Unit for Biomedical Research, Medical University Vienna, Vienna, Austria.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Gabor Szabo
- Department of Cardiac Surgery, Heart and Marfan Center, University of Heidelberg, Heidelberg, Germany
| | - Alexander Weymann
- Department of Cardiac Surgery, Heart and Marfan Center, University of Heidelberg, Heidelberg, Germany
| | - Andreas Zuckermann
- Division of Cardiac Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research, Core Unit for Biomedical Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
18
|
Zhang R, Wang M, Xia N, Yu S, Chen Y, Wang N. Cloning and analysis of gene expression of interleukin-17 homolog in triangle-shell pearl mussel, Hyriopsis cumingii, during pearl sac formation. FISH & SHELLFISH IMMUNOLOGY 2016; 52:151-156. [PMID: 26994668 DOI: 10.1016/j.fsi.2016.03.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/10/2016] [Accepted: 03/13/2016] [Indexed: 06/05/2023]
Abstract
Successful allograft of mantle tissues in certain bivalve mollusks can form pearl sacs secreting nacre for pearl production. Little was known, however, about the immune consequences in response to the tissue transplantation. In the present study, interleukin (IL)-17, one of the key regulatory genes of alloimmunity, was cloned from the triangle-shell pearl mussel (HcIL-17) Hyriopsis cumingii by high-throughput sequencing of the mantle transcriptome. The sequence of HcIL-17 contains an open reading frame of 567 bp encoding a putative protein of 188 amino acid residues. Analysis of sequence characteristics, multiple sequence alignment and phylogenetic analysis indicated HcIL-17 was a novel member in the mollusk IL-17 family. Expression of the HcIL-17 gene in donor mantle tissues and in hemocytes of recipient mussel was up-regulated dramatically within 7 days in response to the mantle tissue allograft for pearl aquaculture, suggesting remarkable proinflammatory responses during pearl sac formation in triangle-shell pearl mussels. Analysis of the time-course expression of HcIL-17 gene revealed the induction of HcIL-17 was time-dependent, reflecting the different periods of alloimmune events in triangle-shell mussels. The results of this study provide essential background information for further investigation of mollusk alloimmunity.
Collapse
Affiliation(s)
- Rui Zhang
- School of Medicine, Jiangsu University, Zhenjiang City, 212013, China
| | - Meng Wang
- School of Food Science and Biological Engineering, Jiangsu University, Zhenjiang City, 212013, China
| | - Ni Xia
- School of Food Science and Biological Engineering, Jiangsu University, Zhenjiang City, 212013, China
| | - Shuang Yu
- School of Medicine, Jiangsu University, Zhenjiang City, 212013, China
| | - Yi Chen
- School of Medicine, Jiangsu University, Zhenjiang City, 212013, China
| | - Ning Wang
- School of Food Science and Biological Engineering, Jiangsu University, Zhenjiang City, 212013, China.
| |
Collapse
|
19
|
Khan MA, Hsu JL, Assiri AM, Broering DC. Targeted complement inhibition and microvasculature in transplants: a therapeutic perspective. Clin Exp Immunol 2015; 183:175-86. [PMID: 26404106 DOI: 10.1111/cei.12713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2015] [Indexed: 12/18/2022] Open
Abstract
Active complement mediators play a key role in graft-versus-host diseases, but little attention has been given to the angiogenic balance and complement modulation during allograft acceptance. The complement cascade releases the powerful proinflammatory mediators C3a and C5a anaphylatoxins, C3b, C5b opsonins and terminal membrane attack complex into tissues, which are deleterious if unchecked. Blocking complement mediators has been considered to be a promising approach in the modern drug discovery plan, and a significant number of therapeutic alternatives have been developed to dampen complement activation and protect host cells. Numerous immune cells, especially macrophages, develop both anaphylatoxin and opsonin receptors on their cell surface and their binding affects the macrophage phenotype and their angiogenic properties. This review discusses the mechanism that complement contributes to angiogenic injury, and the development of future therapeutic targets by antagonizing activated complement mediators to preserve microvasculature in rejecting the transplanted organ.
Collapse
Affiliation(s)
- M A Khan
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - J L Hsu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - A M Assiri
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - D C Broering
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
20
|
Thomas MN, Kalnins A, Andrassy M, Wagner A, Klussmann S, Rentsch M, Habicht A, Pratschke S, Stangl M, Bazhin AV, Meiser B, Fischereder M, Werner J, Guba M, Andrassy J. SDF-1/CXCR4/CXCR7 is pivotal for vascular smooth muscle cell proliferation and chronic allograft vasculopathy. Transpl Int 2015; 28:1426-35. [PMID: 26265085 DOI: 10.1111/tri.12651] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/09/2015] [Accepted: 07/20/2015] [Indexed: 01/29/2023]
Abstract
Chronic rejection remains a major obstacle in transplant medicine. Recent studies suggest a crucial role of the chemokine SDF-1 on neointima formation after injury. Here, we investigate the potential therapeutic effect of inhibiting the SDF-1/CXCR4/CXCR7 axis with an anti-SDF-1 Spiegelmer (NOX-A12) on the development of chronic allograft vasculopathy. Heterotopic heart transplants from H-2bm12 to B6 mice and aortic transplants from Balb/c to B6 were performed. Mice were treated with NOX-A12. Control animals received a nonfunctional Spiegelmer (revNOX-A12). Samples were retrieved at different time points and analysed by histology, RT-PCR and proliferation assay. Blockade of SDF-1 caused a significant decrease in neointima formation as measured by intima/media ratio (1.0 ± 0.1 vs. 1.8 ± 0.1, P < 0.001 AoTx; 0.35 ± 0.05 vs. 1.13 ± 0.27, P < 0.05 HTx). In vitro treatment of primary vascular smooth muscle cells with NOX-A12 showed a significant reduction in proliferation (0.42 ± 0.04 vs. 0.24 ± 0.03, P < 0.05). TGF-β, TNF-α and IL-6 levels were significantly reduced under SDF-1 inhibition (3.42 ± 0.37 vs. 1.67 ± 0.33, P < 0.05; 2.18 ± 0.37 vs. 1.0 ± 0.39, P < 0.05; 2.18 ± 0.26 vs. 1.6 ± 0.1, P < 0.05). SDF-1/CXCR4/CXCR7 plays a critical role in the development of chronic allograft vasculopathy (CAV). Therefore, pharmacological inhibition of SDF-1 with NOX-A12 may represent a therapeutic option to ameliorate chronic rejection changes.
Collapse
Affiliation(s)
- Michael N Thomas
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Aivars Kalnins
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Martin Andrassy
- Innere Medizin III: Kardiologie, Angiologie und Pneumologie, Universitätsklinikum Heidelberg, Rupprecht-Karls Universität, Heidelberg, Germany
| | - Anne Wagner
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | | | - Markus Rentsch
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Antje Habicht
- Transplantationszentrum, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Sebastian Pratschke
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Manfred Stangl
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Alexandr V Bazhin
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Bruno Meiser
- Transplantationszentrum, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Michael Fischereder
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Jens Werner
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Markus Guba
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| | - Joachim Andrassy
- Klinik für Allgemeine-,Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum der Universität München, Ludwig-Maximilians Universität, München, Germany
| |
Collapse
|
21
|
Hwang SM, Jin M, Shin YH, Ki Choi S, Namkoong E, Kim M, Park MY, Park K. Role of LPA and the Hippo pathway on apoptosis in salivary gland epithelial cells. Exp Mol Med 2014; 46:e125. [PMID: 25502757 PMCID: PMC4274396 DOI: 10.1038/emm.2014.77] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/11/2014] [Accepted: 09/19/2014] [Indexed: 12/11/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid involved in numerous physiological responses. However, the expression of LPA receptors and the role of the Hippo signaling pathway in epithelial cells have remained elusive. In this experiment, we studied the functional expression of LPA receptors and the associated signaling pathway using reverse transcriptase–PCR, microspectrofluorimetry, western blotting and immunocytochemistry in salivary gland epithelial cells. We found that LPA receptors are functionally expressed and involved in activating the Hippo pathway mediated by YAP/TAZ through Lats/Mob1 and RhoA/ROCK. Upregulation of YAP/TAZ-dependent target genes, including CTGF, ANKRD1 and CYR61, has also been observed in LPA-treated cells. In addition, based on data suggesting that tumor necrosis factor (TNF)-α induces cell apoptosis, LPA upregulates TNF-induced caspase-3 and cleaved Poly(ADP-ribose)polymerase (PARP). However, small interfering RNA treatment to Yes-associated protein (YAP) or transcriptional co-activator with a PDZ-binding motif (TAZ) significantly decreased TNF-α- and LPA-induced apoptosis, suggesting that YAP and TAZ modulate the apoptotic pathway in salivary epithelial cells.
Collapse
Affiliation(s)
- Sung-Min Hwang
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - MeiHong Jin
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - Yong Hwan Shin
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - Seul Ki Choi
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - Eun Namkoong
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - MinKyoung Kim
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - Moon-Yong Park
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| | - Kyungpyo Park
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, ChongnoKu, Seoul, Korea
| |
Collapse
|
22
|
Natural Killer Cells Play a Critical Role in Cardiac Allograft Vasculopathy in an Interleukin-6–Dependent Manner. Transplantation 2014; 98:1029-39. [DOI: 10.1097/tp.0000000000000405] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Shi X, Zhang M, Liu F, Wang Z, Zhang L, Cheng H, Zhang S, Fei T, Guo M, Bian J, Wang Q, Ding G. Tim-1-Fc suppresses chronic cardiac allograft rejection and vasculopathy by reducing IL-17 production. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:509-520. [PMID: 24551271 PMCID: PMC3925895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 12/31/2013] [Indexed: 06/03/2023]
Abstract
Previously, we demonstrated that Tim-1-Fc prevents acute cardiac graft rejection by inhibiting Th1 response. In the present report, we tackled the impact of Tim-1-Fc on Th17 cells in a model of cardiac chronic rejection. Administration of Tim-1-Fc did not result in a detectable impact on innate immunity and regulatory T cells, while it provided protection for Bm12-derive cardiac grafts against chronic rejection in B6 recipients, as manifested by the reduction of inflammatory infiltration along with less severity of vasculopathy. Studies in T-bet(-/-) recipients by implanting Bm12-derived cardiac grafts further revealed that Tim-1-Fc significantly protected cardiac grafts from chronic rejection along with attenuated production of IL-17 producing T cells. Depletion of CD4 and CD8 T cells or blockade of IL-17 in T-bet(-/-) recipients demonstrated that Tim-1-Fc selectively suppresses Th17 differentiation along with attenuated IL-17 secretion. Together, our data suggest that Tim-1-Fc protects cardiac grafts from chronic rejection by suppressing CD4 Th17 development and functionality. Therefore, Tim-1-Fc might be a potential immunosuppressive agent in the setting of cardiac transplantation.
Collapse
Affiliation(s)
- Xiaoming Shi
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| | - Mingjian Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical UniversityShanghai, China
| | - Fang Liu
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| | - Zhengxing Wang
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| | - Luding Zhang
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| | - Haifei Cheng
- Department of Pharmacology, 411 Naval Medical HospitalShanghai, China
| | - Shu Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Teng Fei
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| | - Meng Guo
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| | - Jun Bian
- Department of Pharmacology, 411 Naval Medical HospitalShanghai, China
| | - Quanxing Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical UniversityShanghai, China
| | - Guoshan Ding
- Institute of Organ Transplantation, Changzheng Hospital, Second Military Medical UniversityShanghai, China
| |
Collapse
|
24
|
Sun G, Xue R, Yao F, Liu D, Huang H, Chen C, Li Y, Zeng J, Zhang G, Dong Y, Liu C. The critical role of Sestrin 1 in regulating the proliferation of cardiac fibroblasts. Arch Biochem Biophys 2014; 542:1-6. [DOI: 10.1016/j.abb.2013.11.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 11/20/2013] [Accepted: 11/25/2013] [Indexed: 11/27/2022]
|
25
|
Single-target RNA interference for the blockade of multiple interacting proinflammatory and profibrotic pathways in cardiac fibroblasts. J Mol Cell Cardiol 2013; 66:141-56. [PMID: 24239602 DOI: 10.1016/j.yjmcc.2013.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 10/18/2013] [Accepted: 11/04/2013] [Indexed: 12/14/2022]
Abstract
Therapeutic targets of broad relevance are likely located in pathogenic pathways common to disorders of various etiologies. Screening for targets of this type revealed CCN genes to be consistently upregulated in multiple cardiomyopathies. We developed RNA interference (RNAi) to silence CCN2 and found this single-target approach to block multiple proinflammatory and profibrotic pathways in activated primary cardiac fibroblasts (PCFBs). The RNAi-strategy was developed in murine PCFBs and then investigated in "individual" human PCFBs grown from human endomyocardial biopsies (EMBs). Screening of short hairpin RNA (shRNA) sequences for high silencing efficacy and specificity yielded RNAi adenovectors silencing CCN2 in murine or human PCFBs, respectively. Comparison of RNAi with CCN2-modulating microRNA (miR) vectors expressing miR-30c or miR-133b showed higher efficacy of RNAi. In murine PCFBs, CCN2 silencing resulted in strongly reduced expression of stretch-induced chemokines (Ccl2, Ccl7, Ccl8), matrix metalloproteinases (MMP2, MMP9), extracellular matrix (Col3a1), and a cell-to-cell contact protein (Cx43), suggesting multiple signal pathways to be linked to CCN2. Immune cell chemotaxis towards CCN2-depleted PCFBs was significantly reduced. We demonstrate here that this RNAi strategy is technically applicable to "individual" human PCFBs, too, but that these display individually strikingly different responses to CCN2 depletion. Either genomically encoded factors or stable epigenetic modification may explain different responses between individual PCFBs. The new RNAi approach addresses a key regulator protein induced in cardiomyopathies. Investigation of this and other molecular therapies in individual human PCBFs may help to dissect differential pathogenic processes between otherwise similar disease entities and individuals.
Collapse
|
26
|
IL-13 signaling via IL-13Rα2 triggers TGF-β1-dependent allograft fibrosis. Transplant Res 2013; 2:16. [PMID: 24143891 PMCID: PMC4016099 DOI: 10.1186/2047-1440-2-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 10/10/2013] [Indexed: 11/25/2022] Open
Abstract
Background Allograft fibrosis still remains a critical problem in transplantation, including heart transplantation. The IL-13/TGF-β1 interaction has previously been identified as a key pathway orchestrating fibrosis in different inflammatory immune disorders. Here we investigate if this pathway is also responsible for allograft fibrosis and if interference with the IL-13/TGF-β1 interaction prevents allograft fibrosis. Methods FVB or control DBA/1 donor hearts were transplanted heterotopically into DBA/1 recipient mice and hearts were explanted at day 60 and 100 post-transplantation. Cardiac tissue was examined by Masson’s trichrome staining and immunohistochemistry for CD4, CD8, CD11b, IL-13, Fas ligand, matrix metalloproteinase (MMP)-1, MMP-13, β2-microglobulin, and Gremlin-1. Graft-infiltrating cells were isolated and analyzed by flow cytometry. IL-13 and TGF-β1 levels were determined by enzyme-linked immunosorbent assay (ELISA) and the amount of collagen was quantified using a Sircol assay; IL-13Rα2 expression was detected by Western blotting. In some experiments IL-13/ TGF-β1 signaling was blocked with specific IL-13Rα2 siRNA. Additionally, a PCR array of RNA isolated from the allografts was performed to analyze expression of multiple genes involved in fibrosis. Results Both groups survived long-term (>100 days). The allogeneic grafts were infiltrated by significantly increased numbers of CD4+ (P <0.0001), CD8+ (P <0.0001), and CD11b+ cells (P = 0.0065) by day 100. Furthermore, elevated IL-13 levels (P = 0.0003) and numbers of infiltrating IL-13+ cells (P = 0.0037), together with an expression of IL-13Rα2, were detected only within allografts. The expression of IL-13 and IL-13Rα2 resulted in significantly increased TGF-β1 levels (P <0.0001), higher numbers of CD11bhighGr1intermediateTGF-β1+ cells, and elevated cardiac collagen deposition (P = 0.0094). The allograft fibrosis found in these experiments was accompanied by upregulation of multiple profibrotic genes, which was confirmed by immunohistochemical stainings of allograft tissue. Blockage of the IL-13/TGF-β1 interaction by IL-13Rα2 siRNA led to lower numbers of CD11bhighGr1intermediateTGF-β1+, CD4+, CD8+, and CD11b+ cells, and prevented collagen deposition (P = 0.0018) within these allografts. Conclusions IL-13 signaling via IL-13Rα2 induces TGF-β1 and causes allograft fibrosis in a murine model of chronic transplant rejection. Blockage of this IL-13/TGF-β1 interaction by IL-13Rα2 siRNA prevents cardiac allograft fibrosis. Thus, IL-13Rα2 may be exploitable as a future target to reduce allograft fibrosis in organ transplantation.
Collapse
|
27
|
The C-terminal module IV of connective tissue growth factor is a novel immune modulator of the Th17 response. J Transl Med 2013; 93:812-24. [PMID: 23648563 DOI: 10.1038/labinvest.2013.67] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Connective tissue growth factor (CTGF/CCN2) is a matricellular protein susceptible to proteolytic degradation. CCN2 levels have been suggested as a potential risk biomarker in several chronic diseases. In body fluids, CCN2 full-length and its degradation fragments can be found; however, their in vivo effects are far from being elucidated. CCN2 was described as a profibrotic mediator, but this concept is changing to a proinflammatory cytokine. In vitro, CCN2 full-length and its C-terminal module IV (CCN2(IV)) exert proinflammatory properties. Emerging evidence suggest that Th17 cells, and its effector cytokine IL-17A, participate in chronic inflammatory diseases. Our aim was to explore whether CCN2(IV) could regulate the Th17 response. In vitro, stimulation of human naive CD4+ T lymphocytes with CCN2(IV) resulted in differentiation to Th17 phenotype. The in vivo effects of CCN2(IV) were studied in C57BL/6 mice. Intraperitoneal administration of recombinant CCN2(IV) did not change serum IL-17A levels, but caused an activation of the Th17 response in the kidney, characterized by interstitial infiltration of Th17 (IL17A+/CD4+) cells and upregulation of proinflammatory mediators. In CCN2(IV)-injected mice, elevated renal levels of Th17-related factors (IL-17A, IL-6, STAT3 and RORγt) were found, whereas Th1/Th2 cytokines or Treg-related factors (TGF-β and Foxp-3) were not modified. Treatment with an anti-IL-17A neutralizing antibody diminished CCN2(IV)-induced renal inflammation. Our findings unveil that the C-terminal module of CCN2 induces the Th17 differentiation of human Th17 cells and causes a renal Th17 inflammatory response. Furthermore, these data bear out that IL-17A targeting is a promising tool for chronic inflammatory diseases, including renal pathologies.
Collapse
|
28
|
Schiechl G, Brunner SM, Kesselring R, Martin M, Ruemmele P, Mack M, Hirt SW, Schlitt HJ, Geissler EK, Fichtner-Feigl S. Inhibition of innate co-receptor TREM-1 signaling reduces CD4(+) T cell activation and prolongs cardiac allograft survival. Am J Transplant 2013; 13:1168-80. [PMID: 23463907 DOI: 10.1111/ajt.12186] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 01/25/2023]
Abstract
The innate receptor "triggering-receptor-expressed-on-myeloid-cells-1" (TREM-1) enhances downstream signaling of "pattern recognition receptor" (PRR) molecules implicated in inflammatory responses. However the mechanistic role of TREM-1 in chronic heart rejection has yet to be elucidated. We examined the effect of TREM-1(+) antigen-presenting cells (APC) on alloreactive CD4(+) lymphocytes. Bm12 donor hearts were transplanted into wild-type MHC-class-II-mismatched C57BL/6J recipient mice. Progressive allograft rejection of bm12-donor hearts with decreased organ function, severe vasculopathy and allograft fibrosis was evident within 4 weeks. TREM-1(+) CD11b(+) MHC-II(+) F4/80(+) CCR2(+) APC and IFNγ-producing CD4(+) cells were detected during chronic rejection. Peptide inhibition of TREM-1 attenuated graft vasculopathy, reduced graft-infiltrating leukocytes and prolonged allograft survival, while being accompanied by sustained low levels of CD4(+) and CD8(+) cell infiltration. Remarkably, temporary inhibition of TREM-1 during early immune activation was sufficient for long-term allograft survival. Mechanistically, TREM-1 inhibition leads to reduced differentiation and proliferation of IFNγ-producing Th1 cells. In conclusion, TREM-1 influences chronic heart rejection by regulating the infiltration and differentiation of CD4(+) lymphocytes.
Collapse
Affiliation(s)
- G Schiechl
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lemaître PH, Vokaer B, Charbonnier LM, Iwakura Y, Field KA, Estenne M, Goldman M, Leo O, Remmelink M, Le Moine A. Cyclosporine A drives a Th17- and Th2-mediated posttransplant obliterative airway disease. Am J Transplant 2013; 13:611-20. [PMID: 23331973 DOI: 10.1111/ajt.12067] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 11/16/2012] [Accepted: 11/17/2012] [Indexed: 01/25/2023]
Abstract
Calcineurin-inhibitor refractory bronchiolitis obliterans (BO) represents the leading cause of late graft failure after lung transplantation. T helper (Th)2 and Th17 lymphocytes have been associated with BO development. Taking advantage of a fully allogeneic trachea transplantation model in mice, we addressed the pathogenicity of Th cells in obliterative airway disease (OAD) occurring in cyclosporine A (CsA)-treated recipients. We found that CsA prevented CD8(+) T cell infiltration into the graft and downregulated the Th1 response but affected neither Th2 nor Th17 responses in vivo. In secondary mixed lymphocyte cultures, CsA dramatically decreased donor-specific IFN-γ production, enhanced IL-17 production and did not affect IL-13. As CD4(+) depletion efficiently prevented OAD in CsA-treated recipients, we further explored the role of Th2 and Th17 immunity in vivo. Although IL-4 and IL-17 deficient untreated mice developed an OAD comparable to wild-type recipients, a single cytokine deficiency afforded significant protection in CsA-treated recipients. In conclusion, CsA treatment unbalances T helper alloreactivity and favors Th2 and Th17 as coexisting pathways mediating chronic rejection of heterotopic tracheal allografts.
Collapse
Affiliation(s)
- P H Lemaître
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wen P, Wang X, Li T, Zhang B, Sun X, Qiu G, Fan J, Xing T, Luo Q, Tang H, Peng Z. Establishment of a novel volume-loaded heterotopic heart transplantation model in rats. J Surg Res 2013; 183:435-41. [PMID: 23498339 DOI: 10.1016/j.jss.2013.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/04/2013] [Accepted: 01/10/2013] [Indexed: 11/24/2022]
Abstract
BACKGROUND Non-volume-loaded (NL) donor hearts in the heterotopic heart transplantation model in rats undergo atrophy and thrombus formation in graft cavities after transplantation. The present study aimed to establish a novel model with volume-loaded donor hearts. METHODS We used Sprague-Dawley rats as donors and recipients. We established an NL model by anastomosing the donor ascending aorta and pulmonary artery end-to-side to the recipient abdominal aorta and inferior vena cava, respectively, and ligating the superior and inferior vena cava on the donor right atrium. The method of the volume-loaded (VL) model was the same as described above, except we performed an anastomosis of the donor left atrium to the recipient abdominal aorta to allow volume loading of the donor's left ventricle. We assessed the characteristics of the grafts by the surgical success rate, echocardiography, and histologic evaluation between the two models. RESULTS Echocardiography showed that donor left ventricle in VL models was volume loaded and had normal systolic and diastolic function compared with the NL models. The mean weight of NL hearts was significantly less than that of VL hearts. Morphologic observation revealed that thrombus formation in donor heart cavities in NL model was significantly higher than that in the VL model. The area of cardiomyocytes per high-power field in the NL model was significantly lower than that in the VL model. CONCLUSIONS We provide a novel VL heterotopic heart transplantation model in rats, in which hemodynamic performance of grafts is close to the normal cardiac physiologic situation; thus, the novel model will be more suitable for clinical research.
Collapse
Affiliation(s)
- Peihao Wen
- Department of General Surgery, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nguyen H, Chiasson VL, Chatterjee P, Kopriva SE, Young KJ, Mitchell BM. Interleukin-17 causes Rho-kinase-mediated endothelial dysfunction and hypertension. Cardiovasc Res 2012; 97:696-704. [PMID: 23263331 DOI: 10.1093/cvr/cvs422] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
AIMS Elevated levels of pro-inflammatory cytokine interleukin-17A (IL-17) are associated with hypertensive autoimmune diseases; however, the connection between IL-17 and hypertension is unknown. We hypothesized that IL-17 increases blood pressure by decreasing endothelial nitric oxide production. METHODS AND RESULTS Acute treatment of endothelial cells with IL-17 caused a significant increase in phosphorylation of the inhibitory endothelial nitric oxide (NO) synthase residue threonine 495 (eNOS Thr495). Of the kinases known to phosphorylate eNOS Thr495, only inhibition of Rho-kinase prevented the IL-17-induced increase. IL-17 caused a threefold increase in the Rho-kinase activator RhoA, and this was prevented by an IL-17 neutralizing antibody. In isolated mouse aortas, IL-17 significantly increased eNOS Thr495 phosphorylation, induced RhoA expression, and decreased NO-dependent relaxation responses, all of which were prevented by either an IL-17 neutralizing antibody or inhibition of Rho-kinase. In mice, IL-17 treatment for 1 week significantly increased systolic blood pressure and this was associated with decreased aortic NO-dependent relaxation responses, increased eNOS Thr495 phosphorylation, and increased RhoA expression. Inhibition of Rho-kinase prevented the hypertension caused by IL-17. CONCLUSION These data demonstrate that IL-17 activates RhoA/Rho-kinase leading to endothelial dysfunction and hypertension. Inhibitors of IL-17 or Rho-kinase may prove useful as anti-hypertensive drugs in IL-17-associated autoimmune diseases.
Collapse
Affiliation(s)
- Hoanglan Nguyen
- Division of Nephrology & Hypertension, Department of Internal Medicine, Texas A&M Health Science Center College of Medicine/Scott & White Memorial Hospital, 702 SW HK Dodgen Loop, Temple, TX 76504, USA
| | | | | | | | | | | |
Collapse
|
32
|
Vanhoutte D, van Almen GC, Van Aelst LNL, Van Cleemput J, Droogné W, Jin Y, Van de Werf F, Carmeliet P, Vanhaecke J, Papageorgiou AP, Heymans S. Matricellular proteins and matrix metalloproteinases mark the inflammatory and fibrotic response in human cardiac allograft rejection. Eur Heart J 2012; 34:1930-41. [PMID: 23139380 PMCID: PMC4051259 DOI: 10.1093/eurheartj/ehs375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Aims The cardiac extracellular matrix is highly involved in regulating inflammation, remodelling, and function of the heart. Whether matrix alterations relate to the degree of inflammation, fibrosis, and overall rejection in the human transplanted heart remained, until now, unknown. Methods and results Expression of matricellular proteins, proteoglycans, and metalloproteinases (MMPs) and their inhibitors (TIMPs) were investigated in serial endomyocardial biopsies (n = 102), in a cohort of 39 patients within the first year after cardiac transplantation. Out of 15 matrix-related proteins, intragraft transcript and protein levels of syndecan-1 and MMP-9 showed a strong association with the degree of cardiac allograft rejection (CAR), the expression of pro-inflammatory cytokines tumour necrosis factor (TNF)-α, interleukin (IL)-6 and transforming growth factor (TGF)-β, and with infiltrating CD3+T-cells and CD68+monocytes. In addition, SPARC, CTGF, TSP-2, MMP-14, TIMP-1, Testican-1, TSP-1, Syndecan-1, MMP-2, -9, and -14, as well as IL-6 and TGF-β transcript levels and inflammatory infiltrates all strongly relate to collagen expression in the transplanted heart. More importantly, receiver operating characteristic curve analysis demonstrated that syndecan-1 and MMP-9 transcript levels had the highest area under the curve (0.969 and 0.981, respectively), thereby identifying both as a potential decision-making tool to discriminate rejecting from non-rejecting hearts. Conclusion Out of 15 matrix-related proteins, we identified synd-1 and MMP-9 intragraft transcript levels of as strong predictors of human CAR. In addition, a multitude of non-structural matrix-related proteins closely associate with collagen expression in the transplanted heart. Therefore, we are convinced that these findings deserve further investigation and are likely to be of clinical value to prevent human CAR.
Collapse
Affiliation(s)
- Davy Vanhoutte
- Cardiovascular Diseases, University Hospitals Leuven, and Department of Cardiovascular Sciences, KU Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Significant Reduction of Acute Cardiac Allograft Rejection by Selective Janus Kinase-1/3 Inhibition Using R507 and R545. Transplantation 2012; 94:695-702. [DOI: 10.1097/tp.0b013e3182660496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
34
|
Immunosuppression With 4SC-101, a Novel Inhibitor of Dihydroorotate Dehydrogenase, in a Rat Model of Renal Transplantation. Transplantation 2012; 93:1101-7. [DOI: 10.1097/tp.0b013e31824fd861] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Bruneau S, Woda CB, Daly KP, Boneschansker L, Jain NG, Kochupurakkal N, Contreras AG, Seto T, Briscoe DM. Key Features of the Intragraft Microenvironment that Determine Long-Term Survival Following Transplantation. Front Immunol 2012; 3:54. [PMID: 22566935 PMCID: PMC3342046 DOI: 10.3389/fimmu.2012.00054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 03/02/2012] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss how changes in the intragraft microenvironment serve to promote or sustain the development of chronic allograft rejection. We propose two key elements within the microenvironment that contribute to the rejection process. The first is endothelial cell proliferation and angiogenesis that serve to create abnormal microvascular blood flow patterns as well as local tissue hypoxia, and precedes endothelial-to-mesenchymal transition. The second is the overexpression of local cytokines and growth factors that serve to sustain inflammation and, in turn, function to promote a leukocyte-induced angiogenesis reaction. Central to both events is overexpression of vascular endothelial growth factor (VEGF), which is both pro-inflammatory and pro-angiogenic, and thus drives progression of the chronic rejection microenvironment. In our discussion, we focus on how inflammation results in angiogenesis and how leukocyte-induced angiogenesis is pathological. We also discuss how VEGF is a master control factor that fosters the development of the chronic rejection microenvironment. Overall, this review provides insight into the intragraft microenvironment as an important paradigm for future direction in the field.
Collapse
Affiliation(s)
- Sarah Bruneau
- The Division of Nephrology, Transplantation Research Center, Children's Hospital Boston Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Booth AJ, Wood SC, Cornett AM, Dreffs AA, Lu G, Muro AF, White ES, Bishop DK. Recipient-derived EDA fibronectin promotes cardiac allograft fibrosis. J Pathol 2012; 226:609-18. [PMID: 21960174 DOI: 10.1002/path.3010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/25/2011] [Accepted: 09/23/2011] [Indexed: 11/12/2022]
Abstract
Advances in donor matching and immunosuppressive therapies have decreased the prevalence of acute rejection of cardiac grafts; however, chronic rejection remains a significant obstacle for long-term allograft survival. While initiating elements of anti-allograft immune responses have been identified, the linkage between these factors and the ultimate development of cardiac fibrosis is not well understood. Tissue fibrosis resembles an exaggerated wound healing response, in which extracellular matrix (ECM) molecules are central. One such ECM molecule is an alternatively spliced isoform of the ubiquitous glycoprotein fibronectin (FN), termed extra domain A-containing cellular fibronectin (EDA cFN). EDA cFN is instrumental in fibrogenesis; thus, we hypothesized that it might also regulate fibrotic remodelling associated with chronic rejection. We compared the development of acute and chronic cardiac allograft rejection in EDA cFN-deficient (EDA(-/-)) and wild-type (WT) mice. While EDA(-/-) mice developed acute cardiac rejection in a manner indistinguishable from WT controls, cardiac allografts in EDA(-/-) mice were protected from fibrosis associated with chronic rejection. Decreased fibrosis was not associated with differences in cardiomyocyte hypertrophy or intra-graft expression of pro-fibrotic mediators. Further, we examined expression of EDA cFN and total FN by whole splenocytes under conditions promoting various T-helper lineages. Conditions supporting regulatory T-cell (Treg) development were characterized by greatest production of total FN and EDA cFN, though EDA cFN to total FN ratios were highest in Th1 cultures. These findings indicate that recipient-derived EDA cFN is dispensable for acute allograft rejection responses but that it promotes the development of fibrosis associated with chronic rejection. Further, conditions favouring the development of regulatory T cells, widely considered graft-protective, may drive production of ECM molecules which enhance deleterious remodelling responses. Thus, EDA cFN may be a therapeutic target for ameliorating fibrosis associated with chronic cardiac allograft rejection.
Collapse
Affiliation(s)
- Adam J Booth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Booth AJ, Grabauskiene S, Wood SC, Lu G, Burrell BE, Bishop DK. IL-6 promotes cardiac graft rejection mediated by CD4+ cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:5764-71. [PMID: 22025555 DOI: 10.4049/jimmunol.1100766] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
IL-6 mediates numerous immunologic effects relevant to transplant rejection; however, its specific contributions to these processes are not fully understood. To this end, we neutralized IL-6 in settings of acute cardiac allograft rejection associated with either CD8(+) or CD4(+) cell-dominant responses. In a setting of CD8(+) cell-dominant graft rejection, IL-6 neutralization delayed the onset of acute rejection while decreasing graft infiltrate and inverting anti-graft Th1/Th2 priming dominance in recipients. IL-6 neutralization markedly prolonged graft survival in the setting of CD4(+) cell-mediated acute rejection and was associated with decreased graft infiltrate, altered Th1 responses, and reduced serum alloantibody. Furthermore, in CD4(+) cell-dominated rejection, IL-6 neutralization was effective when anti-IL-6 administration was delayed by as many as 6 d posttransplant. Finally, IL-6-deficient graft recipients were protected from CD4(+) cell-dominant responses, suggesting that IL-6 production by graft recipients, rather than grafts, is necessary for this type of rejection. Collectively, these observations define IL-6 as a critical promoter of graft infiltration and a shaper of T cell lineage development in cardiac graft rejection. In light of these findings, the utility of therapeutics targeting IL-6 should be considered for preventing cardiac allograft rejection.
Collapse
Affiliation(s)
- Adam Jared Booth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
38
|
Brunner SM, Schiechl G, Falk W, Schlitt HJ, Geissler EK, Fichtner-Feigl S. Interleukin-33 prolongs allograft survival during chronic cardiac rejection. Transpl Int 2011; 24:1027-39. [PMID: 21797940 DOI: 10.1111/j.1432-2277.2011.01306.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Interleukin-33 (IL-33) stimulates the generation of cells and cytokines characteristic of a Th2 immune response. We examined the effects of IL-33 on allografted heart tissue in a chronic cardiac rejection model, including analysis of the peripheral myeloid and lymphoid compartments. B6.C-H2bm12/KhEg hearts were transplanted into MHC class II-mismatched C57Bl/6J mice; IL-33 was administered daily. Cells from allografts and spleens were isolated for flow cytometry and cultured for cytokine production; some tissues were used for immunohistochemistry. Animals treated with IL-33 showed significantly longer allograft survival, which was associated with a distinct cytokine profile produced by graft-infiltrating cells. Proinflammatory IL-17A production was decreased with IL-33 treatment, while increased levels of IL-5, IL-10, and IL-13 were observed. After IL-33 therapy, flow cytometry showed a direct induction of CD4(+) Foxp3(+) Treg, whereas the number of B220(+) CD19(+) B cells, and circulating, as well as allograft deposited, alloantibodies was reduced. Following IL-33 treatment, a significant decrease in graft-infiltrating CD11b(high) Gr1(high) granulocytes coincided with a significant increase in CD11b(high) Gr1(intermediate) myeloid-derived suppressor cells (MDSC). In conclusion, IL-33 treatment in the setting of chronic rejection promotes the development of a Th2-type immune response that favors MDSC and Treg expansion, reduces antibody-mediated rejection (AMR), and ultimately, prolongs allograft survival.
Collapse
Affiliation(s)
- Stefan M Brunner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|