Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Transplant ; 38(1): e15235, 2024 01.
Article in English | MEDLINE | ID: mdl-38289893

ABSTRACT

INTRODUCTION: Tacrolimus forms the backbone of immunosuppression regimens in lung transplant recipients (LTRs). It is extensively metabolized by cytochrome P450 (CYP) 3A5 enzymes, of which polymorphisms can significantly affect tacrolimus dose requirements. It is unknown how coadministration of tacrolimus with voriconazole, a potent CYP3A5 inhibitor, affects rejection rates or empiric dose adjustments needed after voriconazole discontinuation. METHODS: This retrospective cohort study compares LTRs with poor (PR) versus intermediate/extensive (IE) CYP3A5 metabolizer phenotypes. The primary endpoint is cumulative immune outcomes within three months of voriconazole discontinuation; secondary endpoints include change in tacrolimus dose-to-concentration ratios after voriconazole discontinuation. RESULTS: Thirty-four patients underwent full analysis: 13 IE and 21 PR metabolizers. A higher proportion of IE metabolizers were African American (46.2% vs. 9.5%, p = .03). There was no significant difference in composite immune outcomes, though there was a proportionally higher frequency of new donor-specific antibody development in PR metabolizers (14.3% vs 7.7%, p = .56). Both groups required approximately 2.5 to 3-fold tacrolimus dose increases post-voriconazole discontinuation to re-attain therapeutic levels. CONCLUSION: This novel investigation sheds light on how CYP3A5 phenotype could be used to guide tacrolimus dosing, with the goal of preventing both toxicity and organ rejection.


Subject(s)
Immunosuppressive Agents , Tacrolimus , Humans , Voriconazole/therapeutic use , Antifungal Agents/therapeutic use , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Retrospective Studies , Transplant Recipients , Genotype , Phenotype , Lung
2.
Surg Endosc ; 38(4): 2134-2141, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38443500

ABSTRACT

INTRODUCTION: A history of lung transplantation is a risk factor for poor outcomes in patients undergoing laparoscopic fundoplication. We wanted to determine whether enhanced recovery after a robotic-assisted surgery program would mitigate these risks. METHODS: We performed a single-center retrospective analysis of the Society of Thoracic Surgery database for patients who underwent elective antireflux procedures from 1/2018 to 2/2021 under the enhanced recovery after surgery program using robotic assistance. We identified the patient and surgical characteristics, morbidity, length of stay, and 30-day readmission rates. RESULTS: Among 386 patients who underwent barrier creation, 41 had previously undergone a lung transplant, either bilateral (n = 28) or single (n = 13). There were no significant differences in postoperative complications (9.8% vs. 5.2%, p = 0.27), median hospital length of stay (1 d vs. 1 d, p = 0.28), or 30-day readmission (7.3% vs. 4.9%, p = 0.46). Bivariate analysis showed that older age (p = 0.03), history of DVT/PE (p < 0.001), history of cerebrovascular events (p = 0.03), opioid dependence (p = 0.02), neurocognitive dysfunction (p < 0.001), and dependent functional status (p = 0.02) were associated with postoperative complications. However, lung transplantation was not associated with an increased risk of postoperative complications (p = 0.28). DISCUSSION: The risk of surgical complications in patients with a history of lung transplantation may be mitigated by the combination of ERAS and minimally invasive surgery such as robot-assisted surgery.


Subject(s)
Enhanced Recovery After Surgery , Laparoscopy , Lung Transplantation , Robotic Surgical Procedures , Humans , Fundoplication/methods , Robotic Surgical Procedures/adverse effects , Robotic Surgical Procedures/methods , Retrospective Studies , Lung Transplantation/adverse effects , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Laparoscopy/adverse effects , Laparoscopy/methods , Length of Stay
3.
Am J Respir Crit Care Med ; 208(9): 930-943, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37695863

ABSTRACT

Rationale: CFTR (cystic fibrosis transmembrane conductance regulator) modulator drugs restore function to mutant channels in patients with cystic fibrosis (CF) and lead to improvements in body mass index and lung function. Although it is anticipated that early childhood treatment with CFTR modulators will significantly delay or even prevent the onset of advanced lung disease, lung neutrophils and inflammatory cytokines remain high in patients with CF with established lung disease despite modulator therapy, underscoring the need to identify and ultimately target the sources of this inflammation in CF lungs. Objectives: To determine whether CF lungs, like chronic obstructive pulmonary disease (COPD) lungs, harbor potentially pathogenic stem cell "variants" distinct from the normal p63/Krt5 lung stem cells devoted to alveolar fates, to identify specific variants that might contribute to the inflammatory state of CF lungs, and to assess the impact of CFTR genetic complementation or CFTR modulators on the inflammatory variants identified herein. Methods: Stem cell cloning technology developed to resolve pathogenic stem cell heterogeneity in COPD and idiopathic pulmonary fibrosis lungs was applied to end-stage lungs of patients with CF (three homozygous CFTR:F508D, one CFTR F508D/L1254X; FEV1, 14-30%) undergoing therapeutic lung transplantation. Single-cell-derived clones corresponding to the six stem cell clusters resolved by single-cell RNA sequencing of these libraries were assessed by RNA sequencing and xenografting to monitor inflammation, fibrosis, and mucin secretion. The impact of CFTR activity on these variants after CFTR gene complementation or exposure to CFTR modulators was assessed by molecular and functional studies. Measurements and Main Results: End-stage CF lungs display a stem cell heterogeneity marked by five predominant variants in addition to the normal lung stem cell, of which three are proinflammatory both at the level of gene expression and their ability to drive neutrophilic inflammation in xenografts in immunodeficient mice. The proinflammatory functions of these three variants were unallayed by genetic or pharmacological restoration of CFTR activity. Conclusions: The emergence of three proinflammatory stem cell variants in CF lungs may contribute to the persistence of lung inflammation in patients with CF with advanced disease undergoing CFTR modulator therapy.


Subject(s)
Cystic Fibrosis , Pulmonary Disease, Chronic Obstructive , Humans , Child, Preschool , Animals , Mice , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Lung/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Inflammation/metabolism
4.
Am J Transplant ; 22(7): 1884-1892, 2022 07.
Article in English | MEDLINE | ID: mdl-35286760

ABSTRACT

The development of donor-specific antibodies (DSA) after lung transplantation is common and results in adverse outcomes. In kidney transplantation, Belatacept has been associated with a lower incidence of DSA, but experience with Belatacept in lung transplantation is limited. We conducted a two-center pilot randomized controlled trial of de novo immunosuppression with Belatacept after lung transplantation to assess the feasibility of conducting a pivotal trial. Twenty-seven participants were randomized to Control (Tacrolimus, Mycophenolate Mofetil, and prednisone, n = 14) or Belatacept-based immunosuppression (Tacrolimus, Belatacept, and prednisone until day 89 followed by Belatacept, Mycophenolate Mofetil, and prednisone, n = 13). All participants were treated with rabbit anti-thymocyte globulin for induction immunosuppression. We permanently stopped randomization and treatment with Belatacept after three participants in the Belatacept arm died compared to none in the Control arm. Subsequently, two additional participants in the Belatacept arm died for a total of five deaths compared to none in the Control arm (log rank p = .016). We did not detect a significant difference in DSA development, acute cellular rejection, or infection between the two groups. We conclude that the investigational regimen used in this study is associated with increased mortality after lung transplantation.


Subject(s)
Lung Transplantation , Tacrolimus , Abatacept/therapeutic use , Antilymphocyte Serum/therapeutic use , Graft Rejection/drug therapy , Graft Rejection/etiology , Graft Rejection/prevention & control , Graft Survival , Humans , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Lung Transplantation/adverse effects , Mycophenolic Acid/therapeutic use , Pilot Projects , Prednisone
5.
Am J Transplant ; 22(12): 3137-3142, 2022 12.
Article in English | MEDLINE | ID: mdl-35869809

ABSTRACT

A recent study concluded that SARS-CoV-2 mRNA vaccine responses were improved among transplant patients taking mTOR inhibitors (mTORi). This could have profound implications for vaccine strategies in transplant patients; however, limitations in the study design raise concerns about the conclusions. To address this issue more robustly, in a large cohort with appropriate adjustment for confounders, we conducted various regression- and machine learning-based analyses to compare antibody responses by immunosuppressive agents in a national cohort (n = 1037). MMF was associated with significantly lower odds of positive antibody response (aOR = 0.09 0.130.18 ). Consistent with the recent mTORi study, the odds tended to be higher with mTORi (aOR = 1.00 1.452.13 ); however, importantly, this seemingly protective tendency disappeared (aOR = 0.47 0.731.12 ) after adjusting for MMF. We repeated this comparison by combinations of immunosuppression agents. Compared to MMF + tacrolimus, MMF-free regimens were associated with higher odds of positive antibody response (aOR = 2.39 4.267.92 for mTORi+tacrolimus; 2.34 5.5415.32 for mTORi-only; and 6.78 10.2515.93 for tacrolimus-only), whereas MMF-including regimens were not, regardless of mTORi use (aOR = 0.81 1.542.98 for MMF + mTORi; and 0.81 1.512.87 for MMF-only). We repeated these analyses in an independent cohort (n = 512) and found similar results. Our study demonstrates that the recently reported findings were confounded by MMF, and that mTORi is not independently associated with improved vaccine responses.


Subject(s)
COVID-19 , Kidney Transplantation , Humans , Tacrolimus , Mycophenolic Acid/therapeutic use , Antibody Formation , MTOR Inhibitors , COVID-19 Vaccines , SARS-CoV-2 , Graft Rejection/prevention & control , COVID-19/prevention & control , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Transplant Recipients , TOR Serine-Threonine Kinases , mRNA Vaccines
6.
Respir Res ; 23(1): 349, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36522710

ABSTRACT

BACKGROUND: Despite causing increased morbidity and mortality, pulmonary hypertension (PH) in chronic obstructive pulmonary disease (COPD) patients (COPD-PH) lacks treatment, due to incomplete understanding of its pathogenesis. Hypertrophy of pulmonary arterial walls and pruning of the microvasculature with loss of capillary beds are known features of pulmonary vascular remodeling in COPD. The remodeling features of pulmonary medium- and smaller vessels in COPD-PH lungs are less well described and may be linked to maladaptation of endothelial cells to chronic cigarette smoking (CS). MicroRNA-126 (miR126), a master regulator of endothelial cell fate, has divergent functions that are vessel-size specific, supporting the survival of large vessel endothelial cells and inhibiting the proliferation of microvascular endothelial cells. Since CS decreases miR126 in microvascular lung endothelial cells, we set out to characterize the remodeling by pulmonary vascular size in COPD-PH and its relationship with miR126 in COPD and COPD-PH lungs. METHODS: Deidentified lung tissue was obtained from individuals with COPD with and without PH and from non-diseased non-smokers and smokers. Pulmonary artery remodeling was assessed by ⍺-smooth muscle actin (SMA) abundance via immunohistochemistry and analyzed by pulmonary artery size. miR126 and miR126-target abundance were quantified by qPCR. The expression levels of ceramide, ADAM9, and endothelial cell marker CD31 were assessed by immunofluorescence. RESULTS: Pulmonary arteries from COPD and COPD-PH lungs had significantly increased SMA abundance compared to non-COPD lungs, especially in small pulmonary arteries and the lung microvasculature. This was accompanied by significantly fewer endothelial cell markers and increased pro-apoptotic ceramide abundance. miR126 expression was significantly decreased in lungs of COPD individuals. Of the targets tested (SPRED1, VEGF, LAT1, ADAM9), lung miR126 most significantly inversely correlated with ADAM9 expression. Compared to controls, ADAM9 was significantly increased in COPD and COPD-PH lungs, predominantly in small pulmonary arteries and lung microvasculature. CONCLUSION: Both COPD and COPD-PH lungs exhibited significant remodeling of the pulmonary vascular bed of small and microvascular size, suggesting these changes may occur before or independent of the clinical development of PH. Decreased miR126 expression with reciprocal increase in ADAM9 may regulate endothelial cell survival and vascular remodeling in small pulmonary arteries and lung microvasculature in COPD and COPD-PH.


Subject(s)
Hypertension, Pulmonary , MicroRNAs , Pulmonary Disease, Chronic Obstructive , Humans , Hypertension, Pulmonary/pathology , Vascular Remodeling , Endothelial Cells/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Artery/metabolism , Lung/metabolism , Ceramides/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Membrane Proteins/metabolism , ADAM Proteins/metabolism
7.
Clin Transplant ; 36(5): e14600, 2022 05.
Article in English | MEDLINE | ID: mdl-35083796

ABSTRACT

Response to two doses of a nucleoside-modified messenger ribonucleic acid (mRNA) vaccine was evaluated in a large solid-organ transplant program. mRNA COVID-19 vaccine was administered to transplant candidates and recipients who met study inclusion criteria. Qualitative anti-SARS-CoV-2 Spike Total Immunoglobulin (Ig) and IgG-specific assays, and a semi-quantitative test for anti-SARS-CoV-2 Spike protein IgG were measured in 241 (17.2%) transplant candidates and 1163 (82.8%) transplant recipients; 55.2% of whom were non-Hispanic White and 44.8% identified as another race. Transplant recipients were a median (IQR) of 3.2 (1.1, 6.8) years from transplantation. Response differed by transplant status: 96.0% versus 43.2% by the anti-SARS-CoV-2 Total Ig (candidates vs. recipients, respectively), 93.5% versus 11.6% by the anti-SARS-CoV-2 IgG assay, and 91.9% versus 30.1% by anti-spike titers after two doses of vaccine. Multivariable analysis revealed candidates had higher likelihood of response versus recipients (odds ratio [OR], 14.6; 95 %CI 2.19, 98.11; P = .02). A slightly lower response was demonstrated in older patients (OR .96; 95 %CI .94, .99; P = .002), patients taking antimetabolites (OR, .21; 95% CI .08, .51; P = .001). Vaccination prior to transplantation should be encouraged.


Subject(s)
COVID-19 , Organ Transplantation , Aged , Antibodies, Viral , BNT162 Vaccine , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Immunity, Humoral , Immunoglobulin G , RNA, Messenger , SARS-CoV-2 , Transplant Recipients
8.
Lung ; 200(1): 11-18, 2022 02.
Article in English | MEDLINE | ID: mdl-35066606

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease with a variable clinical course. Biomarkers that predict patient outcomes are needed. We leveraged data from 300 patients in the multicenter IPF-PRO Registry to determine associations between circulating proteins and the composite outcome of respiratory death or lung transplant. Plasma collected at enrollment was analyzed using aptamer-based proteomics (1305 proteins). Over a median follow-up of 30.4 months, there were 76 respiratory deaths and 26 lung transplants. In unadjusted univariable analyses, 61 proteins were significantly associated with the outcome (hazard ratio > 2 or < 0.5, corrected p ≤ 0.05). In multivariable analyses, a set of 4 clinical measures and 47 unique proteins predicted the probability of respiratory death or lung transplant with an optimism-corrected C-index of 0.76. Our results suggest that select circulating proteins strongly associate with the risk of mortality in patients with IPF and confer information independent of clinical measures.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Transplantation , Cohort Studies , Humans , Proteomics , Registries
9.
Pharmacogenomics J ; 20(1): 69-79, 2020 02.
Article in English | MEDLINE | ID: mdl-30992538

ABSTRACT

Single-nucleotide polymorphisms (SNPs) in genes involved in mycophenolic acid (MPA) metabolism have been shown to contribute to variable MPA exposure, but their clinical effects are unclear. We aimed to determine if SNPs in key genes in MPA metabolism affect outcomes after lung transplantation. We performed a retrospective cohort study of 275 lung transplant recipients, 228 receiving mycophenolic acid and a control group of 47 receiving azathioprine. Six SNPs known to regulate MPA exposure from the SLCO, UGT and MRP2 families were genotyped. Primary outcome was 1-year survival. Secondary outcomes were 3-year survival, nonminimal (≥A2 or B2) acute rejection, and chronic lung allograft dysfunction (CLAD). Statistical analyses included time-to-event Kaplan-Meier with log-rank test and Cox regression modeling. We found that SLCO1B3 SNPs rs4149117 and rs7311358 were associated with decreased 1-year survival [rs7311358 HR 7.76 (1.37-44.04), p = 0.021; rs4149117 HR 7.28 (1.27-41.78), p = 0.026], increased risk for nonminimal acute rejection [rs4149117 TT334/T334G: OR 2.01 (1.06-3.81), p = 0.031; rs7311358 GG699/G699A: OR 2.18 (1.13-4.21) p = 0.019] and lower survival through 3 years for MPA patients but not for azathioprine patients. MPA carriers of either SLCO1B3 SNP had shorter survival after CLAD diagnosis (rs4149117 p = 0.048, rs7311358 p = 0.023). For the MPA patients, Cox regression modeling demonstrated that both SNPs remained independent risk factors for death. We conclude that hypofunctional SNPs in the SLCO1B3 gene are associated with an increased risk for acute rejection and allograft failure in lung transplant recipients treated with MPA.


Subject(s)
Allografts/drug effects , Lung Transplantation/adverse effects , Mycophenolic Acid/administration & dosage , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Adult , Allografts/transplantation , Azathioprine/administration & dosage , Female , Genotype , Humans , Immunosuppressive Agents/administration & dosage , Lung/metabolism , Lung/pathology , Lung Transplantation/methods , Male , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , Mycophenolic Acid/metabolism , Polymorphism, Single Nucleotide/genetics , Transplant Recipients
10.
Am J Transplant ; 19(5): 1464-1477, 2019 05.
Article in English | MEDLINE | ID: mdl-30582269

ABSTRACT

Primary graft dysfunction (PGD) is a major limitation in short- and long-term lung transplant survival. Recent work has shown that mitochondrial damage-associated molecular patterns (mtDAMPs) can promote solid organ injury, but whether they contribute to PGD severity remains unclear. We quantitated circulating plasma mitochondrial DNA (mtDNA) in 62 patients, before lung transplantation and shortly after arrival to the intensive care unit. Although all recipients released mtDNA, high levels were associated with severe PGD development. In a mouse orthotopic lung transplant model of PGD, we detected airway cell-free damaged mitochondria and mtDNA in the peripheral circulation. Pharmacologic inhibition or genetic deletion of formylated peptide receptor 1 (FPR1), a chemotaxis sensor for N-formylated peptides released by damaged mitochondria, inhibited graft injury. An analysis of intragraft neutrophil-trafficking patterns reveals that FPR1 enhances neutrophil transepithelial migration and retention within airways but does not control extravasation. Using donor lungs that express a mitochondria-targeted reporter protein, we also show that FPR1-mediated neutrophil trafficking is coupled with the engulfment of damaged mitochondria, which in turn triggers reactive oxygen species (ROS)-induced pulmonary edema. Therefore, our data demonstrate an association between mtDAMP release and PGD development and suggest that neutrophil trafficking and effector responses to damaged mitochondria are drivers of graft damage.


Subject(s)
Alarmins/metabolism , Lung Diseases/immunology , Lung Diseases/surgery , Lung Transplantation/adverse effects , Mitochondria/metabolism , Primary Graft Dysfunction , Aged , Animals , Cell Separation , DNA, Mitochondrial/blood , Female , Flow Cytometry , Graft Survival , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophils/metabolism , Pulmonary Edema/complications , Pulmonary Edema/immunology , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/metabolism , Reperfusion Injury , Retrospective Studies , Tissue Donors
11.
Respir Res ; 20(1): 227, 2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31640794

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease for which diagnosis and management remain challenging. Defining the circulating proteome in IPF may identify targets for biomarker development. We sought to quantify the circulating proteome in IPF, determine differential protein expression between subjects with IPF and controls, and examine relationships between protein expression and markers of disease severity. METHODS: This study involved 300 patients with IPF from the IPF-PRO Registry and 100 participants without known lung disease. Plasma collected at enrolment was analysed using aptamer-based proteomics (1305 proteins). Linear regression was used to determine differential protein expression between participants with IPF and controls and associations between protein expression and disease severity measures (percent predicted values for forced vital capacity [FVC] and diffusion capacity of the lung for carbon monoxide [DLco]; composite physiologic index [CPI]). Multivariable models were fit to select proteins that best distinguished IPF from controls. RESULTS: Five hundred fifty one proteins had significantly different levels between IPF and controls, of which 47 showed a |log2(fold-change)| > 0.585 (i.e. > 1.5-fold difference). Among the proteins with the greatest difference in levels in patients with IPF versus controls were the glycoproteins thrombospondin 1 and von Willebrand factor and immune-related proteins C-C motif chemokine ligand 17 and bactericidal permeability-increasing protein. Multivariable classification modelling identified nine proteins that, when considered together, distinguished IPF versus control status with high accuracy (area under receiver operating curve = 0.99). Among participants with IPF, 14 proteins were significantly associated with FVC % predicted, 23 with DLco % predicted, 14 with CPI. Four proteins (roundabout homolog-2, spondin-1, polymeric immunoglobulin receptor, intercellular adhesion molecule 5) demonstrated the expected relationship across all three disease severity measures. When considered in pathways analyses, proteins associated with the presence or severity of IPF were enriched in pathways involved in platelet and haemostatic responses, vascular or platelet derived growth factor signalling, immune activation, and extracellular matrix organisation. CONCLUSIONS: Patients with IPF have a distinct circulating proteome and can be distinguished using a nine-protein profile. Several proteins strongly associate with disease severity. The proteins identified may represent biomarker candidates and implicate pathways for further investigation. TRIAL REGISTRATION: ClinicalTrials.gov (NCT01915511).


Subject(s)
Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/genetics , Proteogenomics/methods , Registries , Aged , Biomarkers/blood , Cohort Studies , Female , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Male , Middle Aged , Proteomics/methods
13.
BMC Bioinformatics ; 16: 235, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26220471

ABSTRACT

BACKGROUND: Despite the tremendous drop in the cost of nucleotide sequencing in recent years, many research projects still utilize sequencing of pools containing multiple samples for the detection of sequence variants as a cost saving measure. Various software tools exist to analyze these pooled sequence data, yet little has been reported on the relative accuracy and ease of use of these different programs. RESULTS: In this manuscript we evaluate five different variant detection programs-The Genome Analysis Toolkit (GATK), CRISP, LoFreq, VarScan, and SNVer-with regard to their ability to detect variants in synthetically pooled Illumina sequencing data, by creating simulated pooled binary alignment/map (BAM) files using single-sample sequencing data from varying numbers of previously characterized samples at varying depths of coverage per sample. We report the overall runtimes and memory usage of each program, as well as each program's sensitivity and specificity to detect known true variants. CONCLUSIONS: GATK, CRISP, and LoFreq all gave balanced accuracy of 80% or greater for datasets with varying per-sample depth of coverage and numbers of samples per pool. VarScan and SNVer generally had balanced accuracy lower than 80%. CRISP and LoFreq required up to four times less computational time and up to ten times less physical memory than GATK did, and without filtering, gave results with the highest sensitivity. VarScan and SNVer had generally lower false positive rates, but also significantly lower sensitivity than the other three programs.


Subject(s)
Genetic Variation , Genomics , Sequence Analysis, DNA/methods , Software/standards , Algorithms , DNA/chemistry , DNA/metabolism , High-Throughput Nucleotide Sequencing , Humans , Polymorphism, Single Nucleotide
14.
Magn Reson Med ; 73(5): 1970-8, 2015 May.
Article in English | MEDLINE | ID: mdl-24954886

ABSTRACT

PURPOSE: To demonstrate that longitudinal, noninvasive monitoring via MRI can characterize acute cellular rejection in mouse orthotopic lung allografts. METHODS: Nineteen Balb/c donor to C57BL/6 recipient orthotopic left lung transplants were performed, further divided into control-Ig versus anti-CD4/anti-CD8 treated groups. A two-dimensional multislice gradient-echo pulse sequence synchronized with ventilation was used on a small-animal MR scanner to acquire proton images of lung at postoperative days 3, 7, and 14, just before sacrifice. Lung volume and parenchymal signal were measured, and lung compliance was calculated as volume change per pressure difference between high and low pressures. RESULTS: Normalized parenchymal signal in the control-Ig allograft increased over time, with statistical significance between day 14 and day 3 posttransplantation (0.046→0.789; P < 0.05), despite large intermouse variations; this was consistent with histopathologic evidence of rejection. Compliance of the control-Ig allograft decreased significantly over time (0.013→0.003; P < 0.05), but remained constant in mice treated with anti-CD4/anti-CD8 antibodies. CONCLUSION: Lung allograft rejection in individual mice can be monitored by lung parenchymal signal changes and by lung compliance through MRI. Longitudinal imaging can help us better understand the time course of individual lung allograft rejection and response to treatment.


Subject(s)
Graft Rejection/pathology , Lung Transplantation , Lung/pathology , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Allografts , Animals , Antibodies, Monoclonal/pharmacology , CD4 Antigens/immunology , CD8 Antigens/immunology , Graft Rejection/immunology , Longitudinal Studies , Lung/immunology , Lung Compliance/drug effects , Lung Compliance/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organ Size/drug effects , Organ Size/physiology
15.
Am J Respir Crit Care Med ; 189(5): 556-66, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24471427

ABSTRACT

RATIONALE: Although innate immunity is increasingly recognized to contribute to lung allograft rejection, the significance of endogenous innate ligands, such as hyaluronan (HA) fragments, in clinical or experimental lung transplantation is uncertain. OBJECTIVES: To determine if HA is associated with clinical bronchiolitis obliterans syndrome (BOS) in lung transplant recipients, and evaluate the effect of low- or high-molecular-weight HA on experimental lung allograft rejection, including dependence on innate signaling pathways or effector cells. METHODS: HA concentrations were measured in bronchoalveolar lavage and plasma samples from lung recipients with or without established BOS. BOS and normal lung tissues were assessed for HA localization and expression of HA synthases. Murine orthotopic lung recipients with established tolerance were treated with low- or high-molecular-weight HA under varied experimental conditions, including Toll-like receptor (TLR) 2/4 and myeloid differentiation protein 88 deficiency and neutrophil depletion. MEASUREMENTS AND MAIN RESULTS: HA localized within areas of intraluminal small airways fibrosis in BOS lung tissue. Moreover, transcripts for HA synthase enzymes were significantly elevated in BOS versus normal lung tissues and both lavage fluid and plasma HA concentrations were increased in recipients with BOS. Treatment with low-molecular-weight HA abrogated tolerance in murine orthotopic lung recipients in a TLR2/4- and myeloid differentiation protein 88-dependent fashion and drove expansion of alloantigen-specific T lymphocytes. Additionally, TLR-dependent signals stimulated neutrophilia that promoted rejection. In contrast, high-molecular-weight HA attenuated basal allograft inflammation. CONCLUSIONS: These data suggest that accumulation of HA could contribute to BOS by directly activating innate immune signaling pathways that promote allograft rejection and neutrophilia.


Subject(s)
Allografts/immunology , Bronchiolitis Obliterans/etiology , Graft Rejection/immunology , Hyaluronic Acid/immunology , Immunity, Innate , Lung Transplantation , Postoperative Complications/immunology , Adult , Allografts/metabolism , Allografts/transplantation , Animals , Biomarkers/metabolism , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/metabolism , Bronchoalveolar Lavage Fluid/immunology , Female , Follow-Up Studies , Graft Rejection/metabolism , Humans , Hyaluronic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neutrophil Activation , Neutrophils/metabolism , Postoperative Complications/metabolism , Syndrome
16.
J Immunol ; 189(9): 4221-5, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23018463

ABSTRACT

The mechanisms that link bacterial infection to solid organ rejection remain unclear. In this study, we show that following the establishment of lung allograft acceptance in mice, Pseudomonas aeruginosa airway infection induces a G-CSF-dependent neutrophilia that stimulates acute rejection. Graft-infiltrating neutrophils sharply upregulate the B7 molecules CD80 and CD86, but they do not express CD40 or MHC class II in response to P. aeruginosa infection. Neutrophil B7 promotes naive CD4(+) T cell activation and intragraft IL-2(+), IFN-γ(+), and IL-17(+) T lymphocyte accumulation. Intravital two-photon microscopy reveals direct interactions between neutrophils and CD4(+) T cells within pulmonary allografts. Importantly, lung rejection in P. aeruginosa-infected recipients is triggered by CD80/86 on neutrophils and can be prevented by B7 blockade without affecting clearance of this pathogen. These data show that neutrophils enhance T cell activation through B7 trans-costimulation and suggest that inhibiting neutrophil-mediated alloimmunity can be accomplished without compromising bacterial immune surveillance.


Subject(s)
B7-1 Antigen/physiology , Graft Rejection/etiology , Lung Transplantation/adverse effects , Neutrophils/immunology , Neutrophils/microbiology , Pseudomonas Infections/immunology , Transplantation Tolerance/immunology , Up-Regulation/immunology , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , Graft Rejection/immunology , Graft Rejection/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology
17.
Pharmacotherapy ; 44(3): 283-289, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38304955

ABSTRACT

A growing body of evidence supports the use of bivalirudin as an alternative to unfractionated heparin (UFH) for the prevention of thrombotic events in patients on venovenous (VV) extracorporeal membrane oxygenation (ECMO). However, data in patients bridged to lung transplantation are limited. In this case series, we describe the outcomes of six patients who were transitioned from UFH to bivalirudin during their course of VV ECMO support as a bridge to lung transplantation. All six patients were on VV ECMO support until transplant, with a median duration of 73 days. Bivalirudin demonstrated a shorter time to first therapeutic activated thromboplastin time (aPTT) level. Additionally, time in therapeutic range was longer while patients were receiving bivalirudin compared to UFH (median 92.9% vs. 74.6%). However, major bleeding and thrombotic events occurred while patients were receiving either anticoagulant. Based on our experience, bivalirudin appears to be a viable option for anticoagulation in VV ECMO patients bridged to lung transplantation. Larger studies evaluating the optimal anticoagulation strategy in patients bridged to transplant are needed.


Subject(s)
Lung Transplantation , Thrombosis , Adult , Humans , Heparin/adverse effects , Retrospective Studies , Anticoagulants/adverse effects , Hirudins/adverse effects , Peptide Fragments/therapeutic use , Thrombosis/etiology , Thrombosis/prevention & control , Recombinant Proteins/therapeutic use
18.
Transplantation ; 108(3): 777-786, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37899481

ABSTRACT

BACKGROUND: Chronic lung allograft dysfunction (CLAD) is the leading cause of death beyond the first year after lung transplantation. The development of donor-specific antibodies (DSA) is a recognized risk factor for CLAD. Based on experience in kidney transplantation, we hypothesized that belatacept, a selective T-cell costimulatory blocker, would reduce the incidence of DSA after lung transplantation, which may ameliorate the risk of CLAD. METHODS: We conducted a pilot randomized controlled trial (RCT) at 2 sites to assess the feasibility and inform the design of a large-scale RCT. All participants were treated with rabbit antithymocyte globulin for induction immunosuppression. Participants in the control arm were treated with tacrolimus, mycophenolate mofetil, and prednisone, and participants in the belatacept arm were treated with tacrolimus, belatacept, and prednisone through day 89 after transplant then converted to belatacept, mycophenolate mofetil, and prednisone for the remainder of year 1. RESULTS: After randomizing 27 participants, 3 in the belatacept arm died compared with none in the control arm. As a result, we stopped enrollment and treatment with belatacept, and all participants were treated with standard-of-care immunosuppression. Overall, 6 participants in the belatacept arm died compared with none in the control arm (log rank P = 0.008). We did not observe any differences in the incidence of DSA, acute cellular rejection, antibody-mediated rejection, CLAD, or infections between the 2 groups. CONCLUSIONS: We conclude that the investigational regimen used in this pilot RCT is associated with increased mortality after lung transplantation.


Subject(s)
Lung Transplantation , Tacrolimus , Humans , Abatacept/therapeutic use , Tacrolimus/adverse effects , Mycophenolic Acid/therapeutic use , Prednisone/therapeutic use , Pilot Projects , Immunosuppressive Agents/adverse effects , Immunosuppression Therapy , Antibodies , Lung Transplantation/adverse effects , Graft Rejection/prevention & control , Graft Rejection/etiology , Graft Survival
19.
Blood ; 118(23): 6172-82, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21972291

ABSTRACT

The mechanisms by which innate immune signals regulate alloimmune responses remain poorly understood. In the present study, we show by intravital 2-photon microscopy direct interactions between graft-infiltrating neutrophils and donor CD11c(+) dendritic cells (DCs) within orthotopic lung allografts immediately after reperfusion. Neutrophils isolated from the airways of lung transplantation recipients stimulate donor DCs in a contact-dependent fashion to augment their production of IL-12 and expand alloantigen-specific IFN-γ(+) T cells. DC IL-12 expression is largely regulated by degranulation and induced by TNF-α associated with the neutrophil plasma membrane. Extended cold ischemic graft storage enhances G-CSF-mediated granulopoiesis and neutrophil graft infiltration, resulting in exacerbation of ischemia-reperfusion injury after lung transplantation. Ischemia reperfusion injury prevents immunosuppression-mediated acceptance of mouse lung allografts unless G-CSF-mediated granulopoiesis is inhibited. Our findings identify granulopoiesis-mediated augmentation of alloimmunity as a novel link between innate and adaptive immune responses after organ transplantation.


Subject(s)
Dendritic Cells/cytology , Graft Rejection/immunology , Leukopoiesis/immunology , Lung Transplantation/immunology , Neutrophils/cytology , Acute Disease , Animals , Cell Degranulation/immunology , Cell Membrane/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Interleukin-12/metabolism , Leukopoiesis/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/metabolism , Reperfusion Injury/immunology , Signal Transduction/immunology , Transplantation Immunology/immunology , Transplantation, Homologous , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Transplant Direct ; 9(5): e1482, 2023 May.
Article in English | MEDLINE | ID: mdl-37096152

ABSTRACT

Combined liver-lung transplantation is an uncommon, although vital, procedure for patients with simultaneous end-stage lung and liver disease. The utility of lung-liver transplant has been questioned because of initial poor survival outcomes, particularly when compared with liver-alone transplant recipients. Methods: A single-center, retrospective review of the medical records of 19 adult lung-liver transplant recipients was conducted, comparing early recipients (2009-2014) with a recent cohort (2015-2021). Patients were also compared with the center's single lung or liver transplant recipients. Results: Recent lung-liver recipients were older (P = 0.004), had a higher body mass index (P = 0.03), and were less likely to have ascites (P = 0.02), reflecting changes in the etiologies of lung and liver disease. Liver cold ischemia time was longer in the modern cohort (P = 0.004), and patients had a longer posttransplant length of hospitalization (P = 0.048). Overall survival was not statistically different between the 2 eras studied (P = 0.61), although 1-y survival was higher in the more recent group (90.9% versus 62.5%). Overall survival after lung-liver transplant was equivalent to lung-alone recipients and was significantly lower than liver-alone recipients (5-y survival: 52%, 51%, and 75%, respectively). Lung-liver recipient mortality was primarily driven by deaths within 6 mo of transplant due to infection and sepsis. Graft failure was not significantly different (liver: P = 0.06; lung: P = 0.74). Conclusions: The severity of illness in lung-liver recipients combined with the infrequency of the procedure supports its continued use. However, particular attention should be paid to patient selection, immunosuppression, and prophylaxis against infection to ensure proper utilization of scarce donor organs.

SELECTION OF CITATIONS
SEARCH DETAIL