Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Heart Lung Transplant ; 42(6): 807-818, 2023 06.
Article in English | MEDLINE | ID: mdl-36754701

ABSTRACT

INTRODUCTION: Heart transplant results have constantly improved but primary left ventricle graft dysfunction (LV-PGD) remains a devastating complication early after transplantation. Donor and recipient systemic inflammatory response may be involved in immune activation of the transplant, and LV-PGD development. Here, we investigated donor and recipient plasma and intragraft cytokine profiles preoperatively and during LV-PGD and searched for predictive markers for LV-PGD. METHODS: Donor and recipient plasma samples (n = 74) and myocardial biopsies of heart transplants (n = 64) were analyzed. Plasma and intragraft cytokine levels were determined by multiplexed and next-generation sequencing platforms, respectively. The development of LV-PGD during the first 24 hours, and graft function and mortality up to 1 year after transplantation, were examined. RESULTS: Severe LV-PGD, but not mild or moderate LV-PGD, was significantly associated with early mortality, plasma high-sensitivity troponin elevation, and an increase in intragraft and plasma proinflammatory cytokines during reperfusion. Preoperative donor and recipient plasma cytokine levels failed to predict LV-PGD. Cytokine network analysis identified interleukins -6, -8, -10, and -18 as key players during reperfusion. Prolonged cold and total ischemia time, and increased need for red blood cell transfusions during operation were identified as clinical risk factors for severe LV-PGD. CONCLUSIONS: Severe LV-PGD was associated with a poor clinical outcome. Donor and recipient plasma cytokine profile failed to predict LV-PGD, but severe LV-PGD was associated with an increase in post-reperfusion intragraft and recipient plasma proinflammatory cytokines. Identified key cytokines may be potential therapeutic targets to improve early and long-term outcomes after heart transplantation.


Subject(s)
Heart Transplantation , Lung Transplantation , Primary Graft Dysfunction , Humans , Cytokines , Primary Graft Dysfunction/etiology , Heart Transplantation/adverse effects , Lung Transplantation/adverse effects , Risk Factors , Retrospective Studies
2.
Int J Cancer ; 150(11): 1779-1791, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35041762

ABSTRACT

Cancer is a significant cause of morbidity and mortality after solid organ transplantation (SOT) and related to lifelong immunosuppression. This retrospective registry study assessed for the first time in Finland population-based cancer risk and cancer mortality after all SOTs (lung and childhood transplantations included) as standardized incidence ratios (SIRs) and standardized mortality ratios (SMRs). Data from transplant registries were linked with the data of Finnish Cancer Registry and Statistics Finland. We followed 6548 consecutive first SOT recipients from 1 January 1987 to 31 December 2016 translating to 66 741 person-years (median follow-up time 8.9 years [interquartile range 4.0-15.1]). In total, 2096 cancers were found in 1483 patients (23% of all patients). Majority of cancers (53%) were nonmelanoma skin cancers (NMSCs). The overall SIR was 3.6 (95% confidence interval [CI]: 3.5-3.8) and the SIR excluding NMSCs was 2.2 (95% CI: 2.0-2.3). SIR for all cancers was highest for heart (5.0) and lowest for liver (2.7) recipients. Most common cancer types after NMSCs were non-Hodgkin lymphoma (NHL), SIR 9.9 (95% CI: 8.5-11.4) and kidney cancer, SIR 7.3 (95% CI: 6.0-8.8). Cancer-related deaths were 17% (n = 408) of all deaths after first month post transplantation. SMR for all cancers was 2.5 (95% CI: 2.2-2.7) and for NHL 13.6 (95% CI: 10.7-16.8). Notably, overall SIR for cancer was lower in later period (2000-2016), 3.0 (95% CI: 2.8-3.2), than in earlier period (1987-1999), 4.3 (95% CI: 4.0-4.5), P < .001. Decrease in cancer incidence was temporally associated with major changes in immunosuppression in the 2000s.


Subject(s)
Neoplasms , Organ Transplantation , Skin Neoplasms , Child , Cohort Studies , Finland/epidemiology , Humans , Incidence , Neoplasms/epidemiology , Neoplasms/etiology , Organ Transplantation/adverse effects , Registries , Retrospective Studies , Risk Factors , Skin Neoplasms/complications
3.
Am J Pathol ; 192(2): 254-269, 2022 02.
Article in English | MEDLINE | ID: mdl-34774518

ABSTRACT

Obliterative bronchiolitis (OB) after lung transplantation is a nonreversible, life-threatening complication. Herein, the role of vascular endothelial growth factor receptor (Vegfr)-1 and -2 was investigated in the development of obliterative airway disease (OAD), an experimental model for OB. The nonimmunosuppressed recipients underwent transplantation with fully major histocompatibility complex mismatched heterotopic tracheal allografts and received Vegfr1 and -2-specific monoclonal antibodies either alone or in combination, or rat IgG as a control. The treatment with Vegfr1- or -2-blocking antibody significantly decreased intragraft mRNA expression of natural killer cell activation markers early after transplantation. This was followed by reduced infiltration of Cd11b+ cells and Cd4+ T cells as well as down-regulated mRNA expression of proinflammatory chemokines and profibrotic growth factors. However, blocking of both Vegfr1 and -2 was necessary to reduce luminal occlusion. Furthermore, concomitant inhibition of the calcineurin activation pathway almost totally abolished the development of OAD. This study proposes that blocking of Vegf receptors blunted natural killer cell and innate immune responses early after transplantation and attenuated the development of OAD. The results of this study suggest that further studies on the role of Vegfr1 and -2 blocking in development of obliterative airway lesions might be rewarding.


Subject(s)
Bronchiolitis Obliterans/immunology , Immunity, Innate , Killer Cells, Natural/immunology , Lung Transplantation , Vascular Endothelial Growth Factor Receptor-1/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/genetics , Bronchiolitis Obliterans/pathology , Calcineurin/genetics , Calcineurin/immunology , Killer Cells, Natural/pathology , Male , Mice , Mice, Inbred BALB C , Rats , Signal Transduction/genetics , Signal Transduction/immunology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
J Heart Lung Transplant ; 41(3): 311-324, 2022 03.
Article in English | MEDLINE | ID: mdl-34933799

ABSTRACT

BACKGROUND: The pathophysiological changes related to brain death may affect the quality of the transplanted organs and expose the recipients to risks. We probed systemic changes reflected in donor plasma proteome and investigated their relationship to heart transplant outcomes. METHODS: Plasma samples from brain-dead multi-organ donors were analyzed by label-free protein quantification using high-definition mass spectrometry. Unsupervised and supervised statistical models were used to determine proteome differences between brain-dead donors and healthy controls. Proteome variation and the corresponding biological pathways were analyzed and correlated with transplant outcomes. RESULTS: Statistical models revealed that donors had a unique but heterogeneous plasma proteome with 237 of 463 proteins being changed compared to controls. Pathway analysis showed that coagulation, gluconeogenesis, and glycolysis pathways were upregulated in donors, while complement, LXR/RXR activation, and production of nitric oxide and reactive oxygen species in macrophages pathways were downregulated. In point-biserial correlation analysis, lysine-specific demethylase 3A was moderately correlated with any grade and severe PGD. In univariate and multivariate Cox regression analyses myosin Va and proteasome activator complex subunit 2 were significantly associated with the development of acute rejections with hemodynamic compromise within 30 days. Finally, we found that elevated levels of lysine-specific demethylase 3A and moesin were identified as predictors for graft-related 1-year mortality in univariate analysis. CONCLUSIONS: We show that brain death significantly changed plasma proteome signature Donor plasma protein changes related to endothelial cell and cardiomyocyte function, inflammation, and vascular growth and arteriogenesis could predict transplant outcome suggesting a role in donor evaluation.


Subject(s)
Brain Death/blood , Heart Transplantation , Proteome/analysis , Adult , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Treatment Outcome
5.
J Cardiovasc Dev Dis ; 8(12)2021 Dec 12.
Article in English | MEDLINE | ID: mdl-34940535

ABSTRACT

The median survival of patients with heart transplants is relatively limited, implying one of the most relevant questions in the field-how to expand the lifespan of a heart allograft? Despite optimal transplantation conditions, we do not anticipate a rise in long-term patient survival in near future. In order to develop novel strategies for patient monitoring and specific therapies, it is critical to understand the underlying pathological mechanisms at cellular and molecular levels. These events are driven by innate immune response and allorecognition driven inflammation, which controls both tissue damage and repair in a spatiotemporal context. In addition to immune cells, also structural cells of the heart participate in this process. Novel single cell methods have opened new avenues for understanding the dynamics driving the events leading to allograft failure. Here, we review current knowledge on the cellular composition of a normal heart, and cellular mechanisms of ischemia-reperfusion injury (IRI), acute rejection and cardiac allograft vasculopathy (CAV) in the transplanted hearts. We highlight gaps in current knowledge and suggest future directions, in order to improve cellular and molecular understanding of failing heart allografts.

7.
Clin Transplant ; 34(9): e13984, 2020 09.
Article in English | MEDLINE | ID: mdl-32445429

ABSTRACT

BACKGROUND: Cardiac allograft vasculopathy (CAV) is characterized by diffuse thickening of the arterial intima. Statins reduce the incidence of CAV, but despite the use of statins, CAV remains one of the leading causes of long-term death after heart transplant. Inhibitors of proprotein convertase subtilisin-kexin type 9 (PCSK9) substantially reduce cholesterol levels but have not been tested in heart transplant recipients. METHODS: The Cholesterol lowering with EVOLocumab to prevent cardiac allograft Vasculopathy in De-novo heart transplant recipients (EVOLVD) trial (ClinicalTrials.gov Identifier: NCT03734211) is a randomized, double-blind trial designed to test the effect of the PCSK9 inhibitor evolocumab on coronary intima thickness in heart transplant recipients. Adults who have received a cardiac transplant within the past 4-8 weeks are eligible. Exclusion criteria include an estimated glomerular filtration rate < 20 mL/min/1.73 m2 , renal replacement therapy, or contraindications to coronary angiography with intravascular ultrasound. 130 patients will be randomized (1:1) to 12-month treatment with evolocumab or matching placebo. The primary endpoint is the coronary artery intima thickness as measured by intravascular ultrasound. CONCLUSION: The EVOLVD trial is a randomized clinical trial designed to show whether treatment with the PCSK9 inhibitor evolocumab can ameliorate CAV over the first year after heart transplant.


Subject(s)
Anticholesteremic Agents , Heart Transplantation , Adult , Allografts , Antibodies, Monoclonal, Humanized , Anticholesteremic Agents/therapeutic use , Cholesterol , Cholesterol, LDL , Heart Transplantation/adverse effects , Humans , Proprotein Convertase 9
8.
Eur Heart J Acute Cardiovasc Care ; 9(4): 333-341, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32004079

ABSTRACT

BACKGROUND: The use of venoarterial extracorporeal membrane oxygenation in cardiogenic shock keeps increasing, but its cost-utility is unknown. METHODS: We studied retrospectively the cost-utility of venoarterial extracorporeal membrane oxygenation in a five-year cohort of consequent patients treated due to refractory cardiogenic shock or cardiac arrest in a transplant centre in 2013-2017. In our centre, venoarterial extracorporeal membrane oxygenation is considered for all cardiogenic shock patients potentially eligible for heart transplantation, and for selected postcardiotomy patients. We assessed the costs of the index hospitalization and of the one-year hospital costs, and the patients' health-related quality of life (response rate 71.7%). Based on the data and the population-based life expectancies, we calculated the amount and the costs of quality-adjusted life years gained both without discount and with an annual discount of 3.5%. RESULTS: The cohort included 102 patients (78 cardiogenic shock; 24 cardiac arrest) of whom 67 (65.7%) survived to discharge and 66 (64.7%) to one year. The effective costs per one hospital survivor were 242,303€. Median in-hospital costs of the index hospitalization per patient were 129,967€ (interquartile range 150,340€). Mean predicted number of quality-adjusted life years gained by the treatment was 20.9 (standard deviation 9.7) without discount, and the median cost per quality-adjusted life year was 7474€ (interquartile range 10,973€). With the annual discount of 3.5%, 13.0 (standard deviation 4.8) quality-adjusted life years were gained with the cost of 12,642€ per quality-adjusted life year (interquartile range 15,059€). CONCLUSIONS: We found the use of venoarterial extracorporeal membrane oxygenation in refractory cardiogenic shock and cardiac arrest justified from the cost-utility point of view in a transplant centre setting.


Subject(s)
Extracorporeal Membrane Oxygenation/economics , Heart Arrest/therapy , Patient Acceptance of Health Care , Shock, Cardiogenic/therapy , Cost-Benefit Analysis , Female , Heart Arrest/economics , Humans , Male , Middle Aged , Patient Selection , Retrospective Studies , Shock, Cardiogenic/economics
9.
Circulation ; 140(8): 627-640, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31352795

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury may compromise the short-term and long-term prognosis after heart transplantation. Experimental studies show that simvastatin administered to the organ donor is vasculoprotective and inhibits cardiac allograft ischemia-reperfusion injury. METHODS: Eighty-four multiorgan donors were randomly assigned to receive 80 mg of simvastatin (42 donors) via nasogastric tube after declaration of brain death and upon acceptance as a cardiac donor, or to receive no simvastatin (42 donors). The primary efficacy end point was postoperative plasma troponin T and I levels during the first 24 hours after heart transplantation. Secondary end points included postoperative hemodynamics, inflammation, allograft function, rejections and rejection treatments, and mortality. Results: Organ donor simvastatin treatment significantly reduced the heart recipient plasma levels of troponin T by 34% (14 900 ± 12 100 ng/L to 9800 ± 7900 ng/L, P=0.047), and troponin I by 40% (171 000 ± 151 000 ng/L to 103 000 ± 109 000 ng/L, P=0.023) at 6 hours after reperfusion, the levels of NT-proBNP (N-terminal pro-B-type natriuretic peptide) by 36% (32 800 ± 24 300 ng/L to 20 900 ± 15 900 ng/L; P=0.011) at 1 week, and the number of rejection treatments with hemodynamic compromise by 53% within the first 30 days (P=0.046). Donor simvastatin treatment did not affect donor lipid levels but was associated with a specific transplant myocardial biopsy gene expression profile, and a decrease in recipient postoperative plasma levels of CXCL10 (C-X-C motif chemokine 10), interleukin-1α, placental growth factor, and platelet-derived growth factor-BB. Postoperative hemodynamics, biopsy-proven acute rejections, and mortality were similar. No adverse effects were seen in recipients receiving noncardiac solid organ transplants from simvastatin-treated donors. CONCLUSIONS: Donor simvastatin treatment reduces biomarkers of myocardial injury after heart transplantation, and-also considering its documented general safety profile-may be used as a novel, safe, and inexpensive adjunct therapy in multiorgan donation. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT01160978.


Subject(s)
Allografts/drug effects , Graft Rejection/drug therapy , Heart Transplantation , Inflammation/drug therapy , Reperfusion Injury/drug therapy , Simvastatin/therapeutic use , Adolescent , Adult , Aged , Chemokine CXCL10/blood , Double-Blind Method , Female , Graft Rejection/mortality , Hemodynamics/drug effects , Humans , Male , Middle Aged , Reperfusion Injury/mortality , Tissue Donors , Transplantation, Homologous , Troponin T/blood , Young Adult
10.
Transpl Int ; 32(1): 95-106, 2019 01.
Article in English | MEDLINE | ID: mdl-29953680

ABSTRACT

Hypoxia-inducible factors (HIFs) play a critical role in inflammatory properties of myeloid-derived cells. The effect of HIFs on myeloid-derived cell functions in organ transplantation remains unknown, however. We transplanted hearts into transgenic mice with myeloid cell-targeted deletions of HIF-1α or its negative regulator von Hippel-Lindau (VHL) to investigate the effects of HIF-1α inactivation or HIF pathway activation, respectively, on ischemia-reperfusion injury (IRI) and acute rejection. Deletion of VHL in myeloid cells enhanced mRNA expression of anti-inflammatory genes IDO, Arg-1, and HO-1 in vitro. In vivo, VHL-/- myeloid-derived cells of allograft recipients alleviated IRI and acute rejection, evidenced by reduced cardiomyocyte damage, decreased proinflammatory cytokine mRNA levels, and absence of inflammatory infiltrate at 5 days after transplantation. Ultimately, allograft survival was significantly prolonged. In vitro, VHL-/- myeloid-derived cells dose-dependently inhibited T-cell proliferation. Myeloid cells with HIF-1α-deletion retained proinflammatory qualities in vitro and in vivo. Deletion of VHL in myeloid cells of nonimmunosuppressed cardiac allograft recipients reduced myocardial injury and acute rejection. We suggest that HIF transcription factors induce a regulatory phenotype in myeloid-derived cells, which may be harnessed as a novel therapeutic strategy to regulate immune responses after heart transplantation.


Subject(s)
Heart Transplantation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloid Cells/cytology , Myocytes, Cardiac/drug effects , Vascular Endothelial Growth Factor A/metabolism , Allografts , Animals , Cell Proliferation , Female , Graft Survival , Inflammation , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , RNA, Messenger/metabolism , Reperfusion Injury , T-Lymphocytes/cytology , Transplantation, Homologous
11.
Eur J Cardiothorac Surg ; 55(4): 780-787, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30534984

ABSTRACT

OBJECTIVES: Our goal was to study the outcome of patients with cardiogenic shock who were treated with venoarterial extracorporeal membrane oxygenation (VA ECMO), including the subsequent long-term health-related quality of life (HRQoL). METHODS: We conducted a retrospective study of 133 consecutive patients treated in a single centre from 2007 to 2016. The HRQoL was studied using the EuroQuol-5 dimensions-3 level questionnaire and the RAND 36-Item Short Form Health Survey at a minimum of 1 year after VA ECMO. RESULTS: Of all patients, 66 (49.6%) were weaned from VA ECMO and 16 (12.0%) patients were bridged directly to a transplant, 15 (11.3%) to a ventricular assist device and 1 (0.8%) to a total artificial heart. Survival to discharge was 63.9% and to 1 year, 60.9%. A higher in-hospital mortality rate was independently associated with lower HCO3 at VA ECMO implantation [odds ratio (OR) 1.2/decrease of 1 mmol/l in HCO3 (95% confidence interval 1.1-1.3, P < 0.001)] and with increased need of red blood cells transfused during intensive care [OR 1.9/unit of red blood cells needed/day (95% confidence interval 1.4-2.6, P < 0.001)]. HRQoL measured with the EuroQuol-5 dimensions-3 level questionnaire was equal to the HRQoL of the general population. In the 36-Item Short Form questionnaire, patients reported better emotional well-being and equal energy, pain and general health perception compared to the general population. Limitations were experienced only in physical health. In total, 56% of the patients ≤ 60 years had returned to work. CONCLUSIONS: VA ECMO can provide acceptable long-term survival with good HRQoL for selected patients with refractory cardiogenic shock. Timing of patient assessment and of VA ECMO implantation is essential because deeper acidosis is associated with a higher in-hospital mortality rate.


Subject(s)
Extracorporeal Membrane Oxygenation , Quality of Life , Shock, Cardiogenic/therapy , Extracorporeal Membrane Oxygenation/mortality , Female , Humans , Male , Middle Aged , Retrospective Studies , Shock, Cardiogenic/mortality , Surveys and Questionnaires , Survival Analysis
12.
Exp Clin Transplant ; 15(2): 203-212, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27588416

ABSTRACT

OBJECTIVES: Cardiac vascular endothelial growth factor-B transgene limits myocardial damage in rat infarction models. We investigated whether heart transplant vascular endothelial growth factor-B overexpression protected against ischemia-reperfusion injury. MATERIALS AND METHODS: We transplanted hearts heterotopically from Dark Agouti to Wistar Furth rats. To characterize the role of vascular endothelial growth factor-B in ischemia-reperfusion injury, we transplanted either long-term human vascular endothelial growth factor-B transgene overexpressing hearts from Wistar Furth rats or short-term adeno-associated virus 9-human vascular endothelial growth factor-B-transduced hearts from Dark Agouti rats into Wistar Furth rats. Heart transplants were subjected to 2 hours of cold and 1 hour of warm ex vivo ischemia. Samples were collected 6 hours after reperfusion. RESULTS: Two hours of cold and 1 hour of warm ischemia increased vascular endothelial growth factor-B mRNA levels 2-fold before transplant and 6 hours after reperfusion. Transgenic vascular endothelial growth factor-B overexpression caused mild cardiac hypertrophy and elevated cardiac troponin T levels 6 hours after reperfusion. Laser Doppler measurements indicated impaired epicardial tissue perfusion in these transgenic transplants. Recombinant human vascular endothelial growth factor-B increased mRNA levels of cytochrome c oxidase and extracellular ATPase CD39, suggesting active oxidative phosphorylation and high ATP production. Adeno-associated virus 9-mediated vascular endothelial growth factor-B overexpression in transplanted hearts increased intragraft macrophages 1.5-fold and proinflammatory cytokine interleukin 12 p35 mRNA 1.6-fold, without affecting recipient serum cardiac troponin T concentration. CONCLUSIONS: Vascular endothelial growth factor-B expression in transplanted hearts is linked to ischemia and ischemia-reperfusion injury. Cardiac transgenic vascular endothelial growth factor-B overexpression failed to protect heart transplants from ischemia-reperfusion injury.


Subject(s)
Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Vascular Endothelial Growth Factor B/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/metabolism , Apoptosis , Apyrase/metabolism , Cold Ischemia/adverse effects , Coronary Circulation , Dependovirus/genetics , Disease Models, Animal , Electron Transport Complex IV/metabolism , Genetic Vectors , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/metabolism , Macrophages/metabolism , Male , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/pathology , Oxidative Phosphorylation , Rats, Inbred WF , Rats, Transgenic , Time Factors , Transduction, Genetic , Troponin T/metabolism , Vascular Endothelial Growth Factor B/genetics , Warm Ischemia/adverse effects
13.
Pharmacology ; 98(5-6): 204-208, 2016.
Article in English | MEDLINE | ID: mdl-27359361

ABSTRACT

BACKGROUND: Bone morphogenetic protein (BMP)-7 mediates ischemic tolerance and anti-fibrotic effects in various organs such as kidney and heart. Recently, reno- and podocyte-protective effects of a potent HMG-CoA reductase inhibitor, pitavastatin, were accompanied by BMP-7 upregulation. METHODS: Here, we investigated the effect of simvastatin treatment on BMP-7 expression in major MHC-mismatched rat cardiac allografts subjected to ischemia-reperfusion injury and adaptive immune activation at 10 days. RESULTS: We localized Smad2 activity and Reca-1+ fibroblast specific protein-1+ immunoreactivity, suggesting endothelial-to-mesenchymal transition, at fibrotic borderline of cardiac allografts at 10 days. Simvastatin donor and recipient combination treatment significantly upregulated cardiac allograft BMP-7 expression when compared to nontreated controls at 10 days. CONCLUSION: The beneficial effect of statin treatment on cardiac allograft may in part be mediated through the upregulation of BMP-7.


Subject(s)
Allografts/drug effects , Bone Morphogenetic Protein 7/biosynthesis , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Simvastatin/pharmacology , Up-Regulation/drug effects , Allografts/metabolism , Animals , Gene Expression Regulation , Graft Rejection/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Rats , Rats, Wistar , Simvastatin/therapeutic use , Up-Regulation/physiology
14.
Transpl Immunol ; 37: 40-45, 2016 07.
Article in English | MEDLINE | ID: mdl-27155462

ABSTRACT

BACKGROUND: In transplantation-associated ischemia/reperfusion injury (Tx-IRI), tumor necrosis factor alpha and damage-associated molecular patterns promote caspase-8 and -9 apoptotic and receptor-interacting protein kinase-1 and -3 (RIPK1/3) necroptotic pathway activation. The extent of cell death and the counterbalance between apoptosis and regulated necrosis eventually determine the immune response of the allograft. Although simvastatin prevents Tx-IRI, its role in apoptotic and necroptotic activity remains unsolved. METHODS: Rat allograft donors and recipients were treated with a single-dose of simvastatin 2h prior to allograft procurement and reperfusion, respectively. Intragraft caspase-3, -8, and -9 and RIPK1 and -3 mRNA expression was analysed by quantitative RT-PCR and protein activity measured by immunohistochemistry and luminescent assays 6h after reperfusion. Lactate and lactate dehydrogenase (LDH) levels were analysed from allograft recipient and from hypoxic endothelial cell cultures having treated with activated simvastatin. RESULTS: When compared to without cold ischemia, prolonged 4-hour cold ischemia significantly enhanced intragraft mRNA expression of caspase-3 and -9, and RIPK1 and -3, and elevated protein activity of caspase-9 and RIPK1 in the allografts. Simvastatin pretreatment decreased mRNA expression of caspase-3 and -9, and RIPK1 and -3 and protein activity of caspase-9 and RIPK1 in the allografts. Intragraft caspase-8 mRNA expression remained constant regardless of cold ischemia or simvastatin pretreatment. Simvastatin pretreatment attenuated lactate and LDH levels, both in the allograft recipients and in hypoxic endothelial cell cultures. CONCLUSIONS: The beneficial effects of simvastatin pretreatment in cardiac allograft IRI may involve prevention of apoptosis and necroptosis.


Subject(s)
Caspase 9/metabolism , Endothelial Cells/drug effects , Graft Rejection/prevention & control , Heart Transplantation , Protein Serine-Threonine Kinases/metabolism , Reperfusion Injury/drug therapy , Simvastatin/therapeutic use , Animals , Apoptosis/drug effects , Caspase 9/genetics , Cells, Cultured , Endothelial Cells/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Inbred WF , Receptor-Interacting Protein Serine-Threonine Kinases , Transplantation, Homologous
15.
J Heart Lung Transplant ; 35(5): 671-8, 2016 05.
Article in English | MEDLINE | ID: mdl-26856676

ABSTRACT

BACKGROUND: Obliterative bronchiolitis after lung transplantation is characterized by chronic airway inflammation leading to the obliteration of small airways. Hypoxia-inducible factor-1 (HIF-1) is a master regulator of cellular responses to hypoxia and inflammation. The Von Hippel-Lindau protein (pVHL) drives the degradation of oxygen-sensitive subunit HIF-1α that controls the activity of HIF-1. We investigated the effect of myeloid cell-targeted gene deletion of HIF-1α or its negative regulator pVHL on the development of obliterative airway disease (OAD) in the recipients of tracheal allografts, a mouse model for obliterative bronchiolitis after lung transplantation. METHODS: Tracheal allografts were heterotopically transplanted from BALB/c donor mice to fully major histocompatibility complex-mismatched recipient mice with HIF-1α or VHL gene deletion in myeloid cells. The recipients were left non-immunosuppressed or received tacrolimus daily. Histologic, immunohistochemical, and real-time reverse transcription polymerase chain reaction analyses were performed at 3, 10, and 30 days. RESULTS: In the absence of immunosuppression, myeloid cell-specific VHL deficiency of the recipient mice improved epithelial recovery, decreased inflammatory cell infiltration and expression of pro-inflammatory cytokines, increased regulatory forkhead box P3 messenger RNA expression, and reduced OAD development in tracheal allografts. In the presence of tacrolimus immunosuppression, loss of HIF-1α activity in myeloid cells of the recipient by HIF-1α gene deletion accelerated OAD development in mouse tracheal allografts. CONCLUSIONS: Activity of the HIF-pathway affects the development of allograft rejection, and our results suggest that myeloid cell-specific VHL-deficiency that potentially increases HIF-activity decreases allograft inflammation and the subsequent development of OAD in mouse tracheal allografts.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Bronchiolitis Obliterans , Cell Hypoxia , Graft Rejection , Lung Transplantation , Mice , Transplantation, Homologous
16.
Transplantation ; 100(2): 303-13, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26371596

ABSTRACT

BACKGROUND: Microvascular dysfunction and cardiomyocyte injury are hallmarks of ischemia-reperfusion injury (IRI) after heart transplantation. Platelet-derived growth factors (PDGF) have an ambiguous role in this deleterious cascade. On one hand, PDGF may exert vascular stabilizing and antiapoptotic actions through endothelial-pericyte and endothelial-cardiomyocyte crosstalk in the heart; and on the other hand, PDGF signaling mediates neointimal formation and exacerbates chronic rejection in cardiac allografts. The balance between these potentially harmful and beneficial actions determines the final outcome of cardiac allografts. METHODS AND RESULTS: We transplanted cardiac allografts from Dark Agouti rat and Balb mouse donors to fully major histocompatibility complex-mismatched Wistar Furth rat or C57 mouse recipients with a clinically relevant 2-hour cold ischemia and 1-hour warm ischemia. Ex vivo intracoronary delivery of adenovirus-mediated gene transfer of recombinant human PDGF-BB upregulated messenger RNA expression of anti-mesenchymal transition and survival factors BMP-7 and Bcl-2 and preserved capillary density in rat cardiac allografts at day 10. In mouse cardiac allografts PDGF receptor-ß, but not -α intragraft messenger RNA levels were reduced and capillary protein localization was lost during IRI. The PDGF receptor tyrosine kinase inhibitor imatinib mesylate and a monoclonal antibody against PDGF receptor-α enhanced myocardial damage evidenced by serum cardiac troponin T release in the rat and mouse cardiac allografts 6 hours after reperfusion, respectively. Moreover, imatinib mesylate enhanced rat cardiac allograft vasculopathy, cardiac fibrosis, and late allograft loss at day 56. CONCLUSIONS: Our results suggest that PDGF-B signaling may play a role in endothelial and cardiomyocyte recovery from IRI after heart transplantation.


Subject(s)
Genetic Therapy/methods , Heart Transplantation/adverse effects , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-sis/biosynthesis , Adenoviridae/genetics , Allografts , Animals , Becaplermin , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Genetic Vectors , Imatinib Mesylate/pharmacology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-sis/genetics , Rats, Wistar , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Time Factors
17.
Microcirculation ; 23(1): 5-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26190445

ABSTRACT

Cardiac lymphatic system is a rare focus of the modern cardiovascular research. Nevertheless, the growing body of evidence is depicting lymphatic endothelium as an important functional unit in healthy and diseased myocardium. Since the discovery of angiogenic VEGF-A in 1983 and lymphangiogenic VEGF-C in 1997, an increasing amount of knowledge has accumulated on the essential roles of VEGF ligands and receptors in physiological and pathological angiogenesis and lymphangiogenesis. Tissue adaptation to several stimuli such as hypoxia, pathogen invasion, degenerative process and inflammation often involves coordinated changes in both blood and lymphatic vessels. As lymphatic vessels are involved in the initiation and resolution of inflammation and regulation of tissue edema, VEGF family members may have important roles in myocardial lymphatics in healthy and in cardiac disease. We will review the properties of VEGF ligands and receptors concentrating on their lymphatic vessel effects first in normal myocardium and then in cardiac disease.


Subject(s)
Heart Diseases/metabolism , Lymphangiogenesis , Myocardium/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , Animals , Edema/metabolism , Edema/pathology , Edema/therapy , Heart Diseases/pathology , Heart Diseases/therapy , Humans , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy
18.
Transpl Int ; 28(12): 1416-25, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26174800

ABSTRACT

Obliterative bronchiolitis (OB) involves airway epithelial detachment, fibroproliferation, and inflammation, resulting in chronic rejection and transplant failure. Cysteine-rich 61 (CCN1) is an integrin receptor antagonist with a context-dependent role in inflammatory and fibroproliferative processes. We used a mouse tracheal OB model to investigate the role of CCN1 in the development of lung allograft OB. C57Bl/6 mice received a systemic injection of CCN1-expressing adenoviral vectors 2 days prior to subcutaneous implantation of tracheal allografts from major MHC-mismatched BALB/c mice. We treated another group of tracheal allograft recipients with cyclic arginine-glycine-aspartic acid peptide to dissect the role of αvß3-integrin signaling in mediating CCN1 effects in tracheal allografts. Allografts were removed 4 weeks after transplantation and analyzed for luminal occlusion, inflammation, and vasculogenesis. CCN1 overexpression induced luminal occlusion (P < 0.05), fibroproliferation, and smooth muscle cell proliferation (P < 0.05). Selective activation of αvß3-integrin receptor failed to mimic the actions of CCN1, and blocking failed to inhibit the effects of CCN1 in tracheal allografts. In conclusion, CCN1 exacerbates tracheal OB by enhancing fibroproliferation via an αvß3-integrin-independent pathway. Further experiments are required to uncover its potentially harmful role in the development of OB after lung transplantation.


Subject(s)
Bronchiolitis Obliterans/etiology , Cysteine-Rich Protein 61/metabolism , Trachea/transplantation , Allografts , Animals , Bronchiolitis Obliterans/metabolism , Bronchiolitis Obliterans/pathology , Cell Proliferation , Cysteine-Rich Protein 61/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Rejection/pathology , Immunohistochemistry , Integrin alphaVbeta3/agonists , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Peptides, Cyclic/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Up-Regulation
19.
Transplantation ; 95(9): 1084-91, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23466635

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury (IRI) and allograft dysfunction remain as two of the major clinical challenges after heart transplantation. Here, we investigated the effect of donor treatment with simvastatin and methylprednisolone on microvascular dysfunction and immunomodulation during IRI in rat cardiac allografts subjected to prolonged ischemia time. METHODS: The DA rats received simvastatin, methylprednisolone, or both 2 hr before heart donation. The allografts were subjected to 4-hr hypothermic preservation and transplanted to the fully major histocompatibility complex-mismatched WF rat recipients. RESULTS: Six hours after reperfusion, donor treatment either with simvastatin alone or with high dose of methylprednisolone alone or in combination with simvastatin and methylprednisolone significantly reduced cardiac troponin T release and the number of allograft infiltrating ED1 macrophages MPO neutrophils. However, the combination donor treatment was superior in the prevention of IRI and significantly prolonged allograft survival. Donor simvastatin treatment inhibited allograft microvascular RhoA GTPase pathway activation, whereas methylprednisolone prevented activation of innate immune response and mRNA expression of hypoxia-inducible factor-1α and its multiple target genes. CONCLUSIONS: Our results show that donor treatment in combination with simvastatin and methylprednisolone prevents IRI and has beneficial effect on allograft survival in rat cardiac allografts. Minimizing microvascular injury and the activation of innate immunity may offer a novel therapeutic strategy to expand the donor pool and furthermore improve the function of the marginal donor organs.


Subject(s)
Heart Transplantation/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Methylprednisolone/administration & dosage , Reperfusion Injury/prevention & control , Simvastatin/administration & dosage , Tissue Donors , Animals , Glucuronosyltransferase/genetics , Graft Survival , Hyaluronan Synthases , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunomodulation , RNA, Messenger/analysis , Rats , Rats, Inbred WF , Transplantation, Homologous , rac GTP-Binding Proteins/genetics , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/physiology
20.
Trends Cardiovasc Med ; 23(3): 85-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23295079

ABSTRACT

Ischemia/reperfusion injury of a transplanted heart may result in serious early and late adverse effects such as primary graft dysfunction, increased allograft immunogenicity, and initiation of fibroproliferative cascades that compromise the survival of the recipient. Microvascular dysfunction has a central role in ischemia/reperfusion injury through increased vascular permeability, leukocyte adhesion and extravasation, thrombosis, vasoconstriction, and the no-reflow phenomenon. Here we review the involvement of microvascular endothelial cells and their surrounding pericytes in ischemia/reperfusion injury, and the pleiotropic, cholesterol-independent effects of statins on microvascular dysfunction. In addition, we delineate how the rapid vasculoprotective effects of statins could be used to protect cardiac allografts against ischemia/reperfusion injury by administering statins to the organ donor before graft removal and transplantation.


Subject(s)
Graft Survival/drug effects , Heart Transplantation/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Primary Graft Dysfunction/prevention & control , Simvastatin/therapeutic use , Tissue Donors , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Humans , Microvessels/drug effects , Microvessels/metabolism , Microvessels/transplantation , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Pericytes/drug effects , Pericytes/metabolism , Pericytes/transplantation , Primary Graft Dysfunction/etiology , Primary Graft Dysfunction/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...