Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Am J Transplant ; 23(2): 214-222, 2023 02.
Article in English | MEDLINE | ID: mdl-36695698

ABSTRACT

Fractures negatively impact quality of life and survival. We hypothesized that recipient frailty score and genetic profile measured before transplant would predict risk of fracture after lung transplant. We conducted a retrospective cohort study of bone mineral density (BMD) and fracture among lung transplant recipients at a single center. The association between predictors and outcomes were assessed by multivariable time-dependent Cox models or regression analysis. Among the 284 participants, osteoporosis and fracture were highly prevalent. Approximately 59% of participants had posttransplant osteopenia, and 35% of participants developed at least 1 fracture. Low BMD was associated with a polygenic osteoporosis risk score, and the interaction between genetic score and BMD predicted fracture. Pretransplant frailty was associated with risk for spine and hip fracture, which were not associated with chronic lung allograft dysfunction or death. Chest fractures were the most frequent type of fracture and conferred a 2.2-fold increased risk of chronic lung allograft dysfunction or death (time-dependent P < .001). Pneumonia, pleural effusions, and acute rejection frequently occurred surrounding chest fracture. Pretransplant frailty and recipient genotype may aid clinical risk stratification for fracture after transplant. Fracture carries significant morbidity, underscoring the importance of surveillance and osteoporosis prevention.


Subject(s)
Fractures, Bone , Frailty , Lung Transplantation , Osteoporosis , Humans , Retrospective Studies , Frailty/complications , Quality of Life , Fractures, Bone/genetics , Fractures, Bone/complications , Osteoporosis/genetics , Osteoporosis/complications , Bone Density , Lung Transplantation/adverse effects , Risk Factors
2.
Am J Transplant ; 22(3): 843-852, 2022 03.
Article in English | MEDLINE | ID: mdl-34859569

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been implicated to play a role in chronic lung allograft dysfunction (CLAD). Liver kinase B1 (LKB1), a tumor suppressor gene, can regulate EMT. However, its role in CLAD development following lung transplantation remains unknown. Using qRT-PCR, biopsies from lung transplant recipients with bronchiolitis obliterans syndrome (BOS) demonstrated significant downregulation of LKB1 (p = .0001), compared to stable biopsies. To determine the role of LKB1 in EMT development, we analyzed EMT in human bronchial epithelial cell line BEAS-2B. Knockdown of LKB1 by siRNA significantly dysregulated mesenchymal markers expression in BEAS-2B cells. Following incubation of human primary bronchial epithelial cell or BEAS-2B cells with exosomes isolated from BOS or stable lung transplant recipients, LKB1 expression was inhibited when incubated with BOS-exosome. Incubation with BOS-exosomes also decreased LKB1 expression and induced EMT markers in air-liquid interface culture method. Our results provide novel evidence that exosomes released from transplanted lungs undergoing chronic rejection are associated with inactivated tumor suppressor gene LKB1 and this loss induces EMT leading to the pathogenesis of CLAD following human lung transplantation.


Subject(s)
Bronchiolitis Obliterans , Graft vs Host Disease , Lung Transplantation , Allografts , Biomarkers , Bronchiolitis Obliterans/etiology , Epithelial-Mesenchymal Transition , Genes, Tumor Suppressor , Humans , Liver , Lung , Lung Transplantation/adverse effects
3.
Am J Transplant ; 22(11): 2560-2570, 2022 11.
Article in English | MEDLINE | ID: mdl-35729715

ABSTRACT

Respiratory viral infection (RVI) in lung transplant recipients (LTRs) is a risk for chronic lung allograft dysfunction (CLAD). We hypothesize that donor-derived cell-free DNA (%ddcfDNA), at the time of RVI predicts CLAD progression. We followed 39 LTRs with RVI enrolled in the Genomic Research Alliance for Transplantation for 1 year. Plasma %ddcfDNA was measured by shotgun sequencing, with high %ddcfDNA as ≥1% within 7 days of RVI. We examined %ddcfDNA, spirometry, and a composite (progression/failure) of CLAD stage progression, re-transplant, and death from respiratory failure. Fifty-nine RVI episodes, 38 low and 21 high %ddcfDNA were analyzed. High %ddcfDNA subjects had a greater median %FEV1 decline at RVI (-13.83 vs. -1.83, p = .007), day 90 (-7.97 vs. 0.91, p = .04), and 365 (-20.05 vs. 1.09, p = .047), compared to those with low %ddcfDNA and experienced greater progression/failure within 365 days (52.4% vs. 21.6%, p = .01). Elevated %ddcfDNA at RVI was associated with an increased risk of progression/failure adjusting for symptoms and days post-transplant (HR = 1.11, p = .04). No difference in %FEV1 decline was seen at any time point when RVIs were grouped by histopathology result at RVI. %ddcfDNA delineates LTRs with RVI who will recover lung function and who will experience sustained decline, a utility not seen with histopathology.


Subject(s)
Cell-Free Nucleic Acids , Lung Transplantation , Respiration Disorders , Virus Diseases , Humans , Allografts , Lung , Lung Transplantation/adverse effects , Transplantation, Homologous
4.
Am J Transplant ; 21(7): 2360-2371, 2021 07.
Article in English | MEDLINE | ID: mdl-33249747

ABSTRACT

Histopathologic examination of lungs afflicted by chronic lung allograft dysfunction (CLAD) consistently shows both mononuclear cell (MNC) inflammation and mesenchymal cell (MC) fibroproliferation. We hypothesize that interleukin 6 (IL-6) trans-signaling may be a critical mediator of MNC-MC crosstalk and necessary for the pathogenesis of CLAD. Bronchoalveolar lavage (BAL) fluid obtained after the diagnosis of CLAD has approximately twofold higher IL-6 and soluble IL-6 receptor (sIL-6R) levels compared to matched pre-CLAD samples. Human BAL-derived MCs do not respond to treatment with IL-6 alone but have rapid and prolonged JAK2-mediated STAT3 Tyr705 phosphorylation when exposed to the combination of IL-6 and sIL-6R. STAT3 phosphorylation within MCs upregulates numerous genes causing increased invasion and fibrotic differentiation. MNC, a key source of both IL-6 and sIL-6R, produce minimal amounts of these proteins at baseline but significantly upregulate production when cocultured with MCs. Finally, the use of an IL-6 deficient recipient in a murine orthotopic transplant model of CLAD reduces allograft fibrosis by over 50%. Taken together these results support a mechanism where infiltrating MNCs are stimulated by resident MCs to release large quantities of IL-6 and sIL-6R which then feedback onto the MCs to increase invasion and fibrotic differentiation.


Subject(s)
Interleukin-6 , Lung Transplantation , Allografts , Animals , Fibrosis , Humans , Lung/pathology , Lung Transplantation/adverse effects , Mice , Receptors, Interleukin-6
5.
Am J Transplant ; 21(6): 2145-2160, 2021 06.
Article in English | MEDLINE | ID: mdl-33078555

ABSTRACT

Chronic lung allograft dysfunction (CLAD) remains the major complication limiting long-term survival among lung transplant recipients (LTRs). Limited understanding of CLAD immunopathogenesis and a paucity of biomarkers remain substantial barriers for earlier detection and therapeutic interventions for CLAD. We hypothesized the airway transcriptome would reflect key immunologic changes in disease. We compared airway brush-derived transcriptomic signatures in CLAD (n = 24) versus non-CLAD (n = 21) LTRs. A targeted assessment of the proteome using concomitant bronchoalveolar lavage (BAL) fluid for 24 cytokines/chemokines and alloimmune T cell responses was performed to validate the airway transcriptome. We observed an airway transcriptomic signature of differential genes expressed (DGEs) in CLAD marked by Type-1 immunity and striking upregulation of two endogenous immune regulators: indoleamine 2, 3 dioxygenase 1 (IDO-1) and tumor necrosis factor receptor superfamily 6B (TNFRSF6B). Advanced CLAD staging was associated with a more intense airway transcriptome signature. In a validation cohort using the identified signature, we found an area under the curve (AUC) of 0.77 for CLAD LTRs. Targeted proteomic analyses revealed a predominant Type-1 profile with detection of IFN-γ, TNF-α, and IL-1ß as dominant CLAD cytokines, correlating with the airway transcriptome. The airway transcriptome provides novel insights into CLAD immunopathogenesis and biomarkers that may impact diagnosis of CLAD.


Subject(s)
Bronchiolitis Obliterans , Lung Transplantation , Allografts , Graft Rejection/genetics , Humans , Lung , Lung Transplantation/adverse effects , Proteomics , Transcriptome/genetics
6.
Am J Transplant ; 20(3): 825-833, 2020 03.
Article in English | MEDLINE | ID: mdl-31665560

ABSTRACT

Chronic lung allograft dysfunction (CLAD), a condition of excess matrix deposition and airways fibrosis, limits survival after lung transplantation. Amphiregulin (Areg) is an epidermal growth factor receptor (EGFR) ligand suggested to regulate airway injury and repair. We sought to determine whether Areg expression increases in CLAD, localize the cellular source of Areg induction in CLAD, and assess its effects on airway matrix deposition. Lung fluid Areg protein was quantified in patients with or without CLAD. In situ hybridization was performed to localize Areg and EGFR transcript in CLAD and normal lung tissue. Expression of hyaluronan, a matrix constituent that accumulates in CLAD, was measured in Areg-exposed bronchial epithelial cells in the presence or absence of an EGFR inhibitor. We demonstrated that lung fluid Areg protein was significantly increased in CLAD in a discovery and replication cohort. Areg and EGFR transcripts were abundantly expressed within CLAD tissue, localized to basally distributed airway epithelial cells overlying fibrotic regions. Areg-exposed bronchial epithelial cells increased hyaluronan and hyaluronan synthase expression in an EGFR-dependent manner. Collectively, these novel observations suggest that Areg contributes to airway remodeling and CLAD. Moreover these data implicate a role for EGFR signaling in CLAD pathogenesis, suggesting novel therapeutic targets.


Subject(s)
Airway Remodeling , Lung Transplantation , Allografts , Amphiregulin/genetics , Humans , Lung , Lung Transplantation/adverse effects
7.
Am J Transplant ; 20(10): 2644-2651, 2020 10.
Article in English | MEDLINE | ID: mdl-32185874

ABSTRACT

Although chronic lung allograft dysfunction (CLAD) remains the major life-limiting factor following lung transplantation, much of its pathophysiology remains unknown. The discovery that CLAD can manifest both clinically and morphologically in vastly different ways led to the definition of distinct subtypes of CLAD. In this review, recent advances in our understanding of the pathophysiological mechanisms of the different phenotypes of CLAD will be discussed with a particular focus on tissue-based and molecular studies. An overview of the current knowledge on the mechanisms of the airway-centered bronchiolitis obliterans syndrome, as well as the airway and alveolar injuries in the restrictive allograft syndrome and also the vascular compartment in chronic antibody-mediated rejection is provided. Specific attention is also given to morphological and molecular markers for early CLAD diagnosis or histological changes associated with subsequent CLAD development. Evidence for a possible overlap between different forms of CLAD is presented and discussed. In the end, "tissue remains the (main) issue," as we are still limited in our knowledge about the actual triggers and specific mechanisms of all late forms of posttransplant graft failure, a shortcoming that needs to be addressed in order to further improve the outcome of lung transplant recipients.


Subject(s)
Bronchiolitis Obliterans , Lung Transplantation , Allografts , Bronchiolitis Obliterans/etiology , Graft Rejection/etiology , Humans , Lung , Lung Transplantation/adverse effects , Transplantation, Homologous
8.
Am J Transplant ; 19(11): 3162-3175, 2019 11.
Article in English | MEDLINE | ID: mdl-31305014

ABSTRACT

Bronchiolitis obliterans syndrome is the main limitation for long-term survival after lung transplantation. Some specific B cell populations are associated with long-term graft acceptance. We aimed to monitor the B cell profile during early development of bronchiolitis obliterans syndrome after lung transplantation. The B cell longitudinal profile was analyzed in peripheral blood mononuclear cells from patients with bronchiolitis obliterans syndrome and patients who remained stable over 3 years of follow-up. CD24hi CD38hi transitional B cells were increased in stable patients only, and reached a peak 24 months after transplantation, whereas they remained unchanged in patients who developed a bronchiolitis obliterans syndrome. These CD24hi CD38hi transitional B cells specifically secrete IL-10 and express CD9. Thus, patients with a total CD9+ B cell frequency below 6.6% displayed significantly higher incidence of bronchiolitis obliterans syndrome (AUC = 0.836, PPV = 0.75, NPV = 1). These data are the first to associate IL-10-secreting CD24hi CD38hi transitional B cells expressing CD9 with better allograft outcome in lung transplant recipients. CD9-expressing B cells appear as a contributor to a favorable environment essential for the maintenance of long-term stable graft function and as a new predictive biomarker of bronchiolitis obliterans syndrome-free survival.


Subject(s)
B-Lymphocytes/metabolism , Biomarkers/metabolism , Bronchiolitis Obliterans/diagnosis , Graft Rejection/diagnosis , Lung Transplantation/adverse effects , Postoperative Complications/diagnosis , Tetraspanin 29/metabolism , Adolescent , Adult , Aged , Bronchiolitis Obliterans/etiology , Cohort Studies , Female , Follow-Up Studies , Graft Rejection/etiology , Graft Survival , Humans , Leukocytes, Mononuclear , Male , Middle Aged , Postoperative Complications/etiology , Prognosis , Risk Factors , Survival Rate , Syndrome , Transplantation, Homologous , Young Adult
9.
Am J Transplant ; 19(1): 247-258, 2019 01.
Article in English | MEDLINE | ID: mdl-30378739

ABSTRACT

Chronic lung allograft dysfunction (CLAD) is a fatal condition that limits survival after lung transplantation (LTx). The pathological hallmark of CLAD is obliterative bronchiolitis (OB). A subset of patients present with a more aggressive CLAD phenotype, called restrictive allograft syndrome (RAS), characterized by lung parenchymal fibrosis (PF). The mouse orthotopic single LTx model has proven relevant to the mechanistic study of allograft injury. The minor-alloantigen-mismatched strain combination using C57BL/10(B10) donors and C57BL/6(B6) recipients reportedly leads to OB. Recognizing that OB severity is a spectrum that may coexist with other pathologies, including PF, we aimed to characterize and quantify pathologic features of CLAD in this model. Left LTx was performed in the following combinations: B10→B6, B6→B10, B6→B6. Four weeks posttransplant, blinded pathologic semi-quantitative assessment showed that OB was present in 66% of B10→B6 and 30% of B6→B10 grafts. Most mice with OB also had PF with a pattern of pleuroparenchymal fibroelastosis, reminiscent of human RAS-related pathology. Grading of pathologic changes demonstrated variable severity of airway fibrosis, PF, acute rejection, vascular fibrosis, and epithelial changes, similar to those seen in human CLAD. These assessments can make the murine LTx model a more useful tool for further mechanistic studies of CLAD pathogenesis.


Subject(s)
Bronchiolitis Obliterans/etiology , Graft Rejection/etiology , Lung Diseases/surgery , Lung Transplantation/adverse effects , Lung Transplantation/methods , Allografts/pathology , Animals , Disease Models, Animal , Fibrosis , Inflammation , Isoantigens , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Male , Mice , Mice, Inbred C57BL , Phenotype , Postoperative Complications/pathology , Tissue Donors , Transplantation, Homologous/adverse effects
10.
Am J Transplant ; 19(10): 2705-2718, 2019 10.
Article in English | MEDLINE | ID: mdl-31278849

ABSTRACT

Despite standardized postoperative care, some lung transplant patients suffer multiple episodes of acute and chronic rejection while others avoid graft problems for reasons that are poorly understood. Using an established model of C57BL/10 to C57BL/6 minor antigen mismatched single lung transplantation, we now demonstrate that the recipient microbiota contributes to variability in the alloimmune response. Specifically, mice from the Envigo facility in Frederick, Maryland contain nearly double the number of CD4+ Foxp3+ regulatory T cells (Tregs ) than mice from the Jackson facility in Bar Harbor, Maine or the Envigo facility in Indianapolis, Indiana (18 vs 9 vs 7%). Lung graft recipients from the Maryland facility thus do not develop acute or chronic rejection. Treatment with broad-spectrum antibiotics decreases Tregs and increases both acute and chronic graft rejection in otherwise tolerant strains of mice. Constitutive depletion of regulatory T cells, using Foxp3-driven expression of diphtheria toxin receptor, leads to the development of chronic rejection and supports the role of Tregs in both acute and chronic alloimmunity. Taken together, our data demonstrate that the microbiota of certain individuals may contribute to tolerance through Treg -dependent mechanisms and challenges the practice of indiscriminate broad-spectrum antibiotic use in the perioperative period.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Commerce/standards , Forkhead Transcription Factors/physiology , Graft Rejection/prevention & control , Lung Diseases/immunology , Lung Transplantation/adverse effects , Microbiota , T-Lymphocytes, Regulatory/immunology , Allografts , Animals , CD4-Positive T-Lymphocytes/microbiology , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Survival/immunology , Lung Diseases/microbiology , Lung Diseases/surgery , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/microbiology , Transplant Recipients
11.
Am J Transplant ; 19(12): 3377-3389, 2019 12.
Article in English | MEDLINE | ID: mdl-31365766

ABSTRACT

Chronic lung allograft dysfunction (CLAD) limits long-term survival after lung transplant (LT). Ischemia-reperfusion injury (IRI) promotes chronic rejection (CR) and CLAD, but the underlying mechanisms are not well understood. To examine mechanisms linking IRI to CR, a mouse orthotopic LT model using a minor alloantigen strain mismatch (C57BL/10 [B10, H-2b ] → C57BL/6 [B6, H-2b ]) and isograft controls (B6→B6) was used with antecedent minimal or prolonged graft storage. The latter resulted in IRI with subsequent airway and parenchymal fibrosis in prolonged storage allografts but not isografts. This pattern of CR after IRI was associated with the formation of B cell-rich tertiary lymphoid organs within the grafts and circulating autoantibodies. These processes were attenuated by B cell depletion, despite preservation of allograft T cell content. Our observations suggest that IRI may promote B cell recruitment that drives CR after LT. These observations have implications for the mechanisms leading to CLAD after LT.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Fibrosis/pathology , Graft Rejection/pathology , Graft Survival/immunology , Lung Transplantation/adverse effects , Reperfusion Injury/complications , Allografts , Animals , Chronic Disease , Disease Models, Animal , Fibrosis/etiology , Graft Rejection/etiology , Male , Mice , Reperfusion Injury/pathology
12.
Am J Transplant ; 18(4): 810-820, 2018 04.
Article in English | MEDLINE | ID: mdl-28941323

ABSTRACT

Graft-versus-host disease (GVHD) is the major cause of nonrelapse morbidity and mortality after allogeneic stem cell transplantation (allo-SCT). Prevention and treatment of GVHD remain inadequate and commonly lead to end-organ dysfunction and opportunistic infection. The role of interleukin (IL)-17 and IL-22 in GVHD remains uncertain, due to an apparent lack of lineage fidelity and variable and contextually determined protective and pathogenic effects. We demonstrate that donor T cell-derived IL-22 significantly exacerbates cutaneous chronic GVHD and that IL-22 is produced by highly inflammatory donor CD4+ T cells posttransplantation. IL-22 and IL-17A derive from both independent and overlapping lineages, defined as T helper (Th)22 and IL-22+ Th17 cells. Donor Th22 and IL-22+ Th17 cells share a similar IL-6-dependent developmental pathway, and while Th22 cells arise independently of the IL-22+ Th17 lineage, IL-17 signaling to donor Th22 directly promotes their development in allo-SCT. Importantly, while both IL-22 and IL-17 mediate skin GVHD, Th17-induced chronic GVHD can be attenuated by IL-22 inhibition in preclinical systems. In the clinic, high levels of both IL-17A and IL-22 expression are present in the skin of patients with GVHD after allo-SCT. Together, these data demonstrate a key role for donor-derived IL-22 in patients with chronic skin GVHD and confirm parallel but symbiotic developmental pathways of Th22 and Th17 differentiation.


Subject(s)
Graft vs Host Disease/etiology , Interleukin-17/metabolism , Interleukins/metabolism , Skin Diseases/etiology , Stem Cell Transplantation/adverse effects , Tissue Donors , Animals , Chronic Disease , Female , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Prognosis , Skin Diseases/metabolism , Skin Diseases/pathology , Transplantation, Homologous , Interleukin-22
13.
Am J Transplant ; 18(1): 136-144, 2018 01.
Article in English | MEDLINE | ID: mdl-28637080

ABSTRACT

The clinical significance and treatment strategies for minimal acute rejection (grade A1), the most common form of acute rejection (AR), remain controversial. In this retrospective single-center cohort study of 441 lung transplant recipients, we formally evaluate the association between minimal AR and chronic lung allograft dysfunction (CLAD) and test a novel hypothesis using bronchoalveolar lavage (BAL) CXCL9 concentration during minimal AR as a biomarker of subsequent CLAD development. In univariable and multivariable models adjusted for all histopathologic injury patterns, minimal AR was not associated with CLAD development. However, minimal AR with elevated BAL CXCL9 concentrations markedly increased CLAD risk in a dose-response manner. Minimal AR with CXCL9 concentrations greater than the 25th, 50th, and 75th percentile had adjusted hazard ratios (HRs) for CLAD of 1.1 (95% confidence interval [CI] 0.8-1.6), 1.6 (95% CI 1.1-2.3), and 2.2 (95% CI 1.4-3.4), respectively. Thus we demonstrate the utility of BAL CXCL9 measurement as a prognostic biomarker that allows discrimination of recipients at increased risk of CLAD development after minimal AR. BAL CXCL9 measurement during transbronchial biopsies may provide clinically useful prognostic data and guide treatment decisions for this common form of AR, as a possible strategy to minimize CLAD development.


Subject(s)
Acute Lung Injury/diagnosis , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL9/metabolism , Graft Rejection/diagnosis , Lung Transplantation/adverse effects , Postoperative Complications/diagnosis , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Female , Follow-Up Studies , Graft Rejection/etiology , Graft Rejection/metabolism , Graft Survival , Humans , Male , Middle Aged , Postoperative Complications/etiology , Postoperative Complications/metabolism , Prognosis , Retrospective Studies , Transplantation, Homologous
14.
Am J Transplant ; 18(8): 2050-2060, 2018 08.
Article in English | MEDLINE | ID: mdl-29607606

ABSTRACT

Chronic lung allograft dysfunction (CLAD) remains the leading cause of morbidity and mortality after lung transplantation. Diagnosis requires spirometric change, which becomes increasingly difficult with advancing CLAD. Fourier decomposition magnetic resonance imaging (FD-MRI) permits acquisition of ventilated-weighted images during free-breathing. This study evaluates FD-MRI in detecting CLAD in selected patients after bilateral lung transplantation (DLTx). DLTx recipients demonstrating CLAD at various stages participated. Radiologists remained blinded to clinical status until completion of image analysis. Image acquisition used a 1.5-T MR scanner using a spoiled gradient echo sequence. After FD processing and regional fractional ventilation (RFV) quantification, the volume defect percentage at 2 thresholds (VDP1,2 ), median lung RFV and quartile coefficient of dispersion (QCD) were calculated. Sixty-two patients participated. CLAD was present in 29/62 (47%) patients, of whom 17/62 (27%) had forced expiratory volume in 1 second ≤65% at image acquisition. VDP1 was higher among these participants compared to other groups (P < .001). Increased VDP1 was associated with subsequent graft loss, with values >2% showing reduced survival, independent of degree of graft dysfunction (P = .005). VDP2 discriminated between presence or absence of CLAD (area under the curve = 0.71; P = .03). QCD increased significantly with advancing disease (P < .001). In conclusion, FD-MRI-derived parameters demonstrate potential in quantitative CLAD diagnosis and assessment after DLTx.


Subject(s)
Bronchiolitis Obliterans/surgery , Graft Rejection/diagnosis , Lung Transplantation/adverse effects , Magnetic Resonance Imaging/methods , Postoperative Complications , Primary Graft Dysfunction/diagnosis , Adult , Allografts , Chronic Disease , Cross-Sectional Studies , Female , Follow-Up Studies , Graft Rejection/etiology , Graft Survival , Humans , Male , Middle Aged , Primary Graft Dysfunction/etiology , Prognosis , Retrospective Studies , Risk Factors , Young Adult
15.
Am J Transplant ; 18(6): 1527-1533, 2018 06.
Article in English | MEDLINE | ID: mdl-29513387

ABSTRACT

Calcineurin inhibitors (CNIs) are the backbone of traditional immunosuppressive regimens for lung transplant recipients (LTR). The CNIs are both narrow therapeutic index drugs with significant interpatient and intrapatient variability that require therapeutic drug monitoring to ensure safety and effectiveness. We hypothesized that tacrolimus time-in-therapeutic range (TTR) affects acute and chronic rejection rates in LTRs. This was a single-center, observational, cross-sectional study of 292 adult LTRs. Subjects who received tacrolimus posttransplant for the first year were included. TTR was calculated at 1 year using protocol goal ranges (12-15 mg/mL months 0-6; 10-12 mg/mL for months 7-12). The primary outcome was acute cellular rejection (ACR) burden at 1 year. Chronic lung allograft dysfunction (CLAD), mortality, and infection rate were assessed as secondary outcomes at 1 year. Primary and secondary outcomes were assessed using logistic regression. Increasing TTR by 10% was associated with a significantly lower likelihood of high-burden ACR at 1 year on univariable (OR 0.46, 95% CI 0.40-0.54, P < .001) and multivariable (OR 0.64, 95% CI 0.47-0.86, P = .003) assessment, controlling for age and induction agent. Increasing TTR by 10% was also associated with lower rates of CLAD (P < .001) and mortality (P < .001) at 1 year. Prospective studies confirming these findings appear warranted.


Subject(s)
Immunosuppressive Agents/therapeutic use , Lung Transplantation , Tacrolimus/therapeutic use , Treatment Outcome , Adult , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged
16.
Am J Transplant ; 17(9): 2338-2349, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28251796

ABSTRACT

Bronchiolitis obliterans after lung transplantation is a major cause of postoperative mortality in which T cell-mediated immunity is known to play an important role. However, the exact contribution of natural killer (NK) cells, which have functions similar to CD8+ T cells, has not been defined. Here, we assessed the role of NK cells in murine bronchiolitis obliterans through heterotopic tracheal transplantations and found a greater percentage of NK cells in allografts than in isografts. Depletion of NK cells using an anti-NK1.1 antibody attenuated bronchiolitis obliterans in transplant recipients compared with controls. In terms of NK cell effector functions, an improvement in bronchiolitis obliterans was observed in perforin-KO recipient mice compared to wild type (WT). Furthermore, we found upregulation of NKG2D-ligand in allografts and demonstrated the significance of this using grafts expressing Rae-1, a murine NKG2D-ligand, which induced severe bronchiolitis obliterans in WT and Rag-1 KO recipients. This effect was ameliorated by injection of anti-NKG2D blocking antibody. Together, these results suggest that cytotoxicity resulting from activation of NK cells through NKG2D leads to the development of murine bronchiolitis obliterans.


Subject(s)
Bronchiolitis Obliterans/etiology , Disease Models, Animal , Graft Rejection/etiology , Killer Cells, Natural/pathology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Trachea/transplantation , Transplantation, Heterotopic/adverse effects , Animals , Bronchiolitis Obliterans/metabolism , Bronchiolitis Obliterans/pathology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Graft Rejection/metabolism , Graft Rejection/pathology , Homeodomain Proteins/physiology , Immunity, Cellular , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID
18.
Am J Transplant ; 17(2): 474-484, 2017 02.
Article in English | MEDLINE | ID: mdl-27278097

ABSTRACT

The immunological role of exosomes in allograft rejection remains unknown. We sought to determine whether exosomes are induced during lung allograft rejection and to define the antigenic compositions of HLA, lung-associated self-antigens (SAgs) and microRNAs (miRNAs). Exosomes were isolated from sera and bronchoalveolar lavage fluid from 30 lung transplant recipients (LTxRs) who were stable or who had acute rejection (AR) or bronchiolitis obliterans syndrome (BOS). Exosomes were defined by flow cytometry for CD63 and western blotting for annexin V SAgs, collagen V (Col-V) and Kα1 tubulin were examined by electron microscopy; miRNAs were profiled by a miRNA array. Donor HLA and SAgs were detected on exosomes from LTxRs with AR and BOS but not from stable LTxRs. Exosomes expressing Col-V were isolated from sera from LTxRs 3 mo before AR and 6 mo before BOS diagnosis, suggesting that exosomes with SAgs may be a noninvasive rejection biomarker. Exosomes isolated from LTxRs with AR or BOS also contained immunoregulatory miRNAs. We concluded that exosomes expressing donor HLA, SAgs and immunoregulatory miRNAs are present in the circulation and local site after human lung transplantation and play an important role in the immune pathogenesis of acute allograft rejection and BOS.


Subject(s)
Autoantigens/immunology , Bronchiolitis Obliterans/etiology , Exosomes/genetics , Graft Rejection/etiology , Lung Transplantation/adverse effects , MicroRNAs/genetics , Tissue Donors , Case-Control Studies , Exosomes/immunology , Female , Follow-Up Studies , Graft Survival , Humans , Male , Middle Aged , Postoperative Complications , Prognosis , Risk Factors , Transplantation, Homologous
19.
Am J Transplant ; 17(5): 1286-1293, 2017 May.
Article in English | MEDLINE | ID: mdl-27664940

ABSTRACT

Mobile health interventions may help transplant recipients follow their complex medical regimens. Pocket Personal Assistant for Tracking Health (Pocket PATH) is one such intervention tailored for lung transplant recipients. A randomized controlled trial showed Pocket PATH's superiority to usual care for promoting the self-management behaviors of adherence, self-monitoring and communication with clinicians during posttransplant year 1. Its long-term impact was unknown. In this study, we examined associations between Pocket PATH exposure during year 1 and longer term clinical outcomes-mortality and bronchiolitis obliterans syndrome (BOS)-among 182 recipients who survived the original trial. Cox regression assessed whether (a) original group assignment and (b) performance of self-management behaviors during year 1 predicted time to outcomes. Median follow-up was 5.7 years after transplant (range 4.2-7.2 years). Pocket PATH exposure had no direct effect on outcomes (p-values >0.05). Self-monitoring was associated with reduced mortality risk (hazard ratio [HR] 0.45; 95% confidence interval [CI] 0.22-0.91; p = 0.027), and reporting abnormal health indicators to clinicians was associated with reduced risks of mortality (HR 0.15; 95% CI 0.04-0.65; p = 0.011) and BOS (HR 0.27; 95% CI 0.08-0.86; p = 0.026), regardless of intervention group assignment. Although Pocket PATH did not have a direct impact on long-term outcomes, early improvements in self-management facilitated by Pocket PATH may be associated with long-term clinical benefit.


Subject(s)
Bronchiolitis Obliterans/surgery , Lung Transplantation/rehabilitation , Quality of Life , Self-Management , Telemedicine/statistics & numerical data , Transplant Recipients , Female , Follow-Up Studies , Graft Survival , Humans , Male , Middle Aged , Prognosis
20.
Am J Transplant ; 17(6): 1502-1514, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27982503

ABSTRACT

Chronic lung allograft dysfunction (CLAD) is the major limitation of long-term survival after lung transplantation. CLAD manifests as bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS). Alloimmune reactions and epithelial-to-mesenchymal transition have been suggested in BOS. However, little is known regarding the role of allogenicity in epithelial cell differentiation. Primary human bronchial epithelial cells (BECs) were treated with activated T cells in the presence or absence of transforming growth factor (TGF)-ß. The expression of epithelial and mesenchymal markers was investigated. The secretion of inflammatory cytokines and matrix metalloproteinase (MMP)-9 was measured in culture supernatants and in plasma from lung transplant recipients (LTRs): 49 stable, 29 with BOS, and 16 with RAS. We demonstrated that C-C motif chemokine 2 secreted by T cells supports TGF-ß-induced MMP-9 production by BECs after binding to C-C chemokine receptor type 2. Longitudinal investigation in LTRs revealed a rise in plasma MMP-9 before CLAD onset. Multivariate analysis showed that plasma MMP-9 was independently associated with BOS (odds ratio [OR] = 6.19, p = 0.002) or RAS (OR = 3.9, p = 0.024) and predicted the occurrence of CLAD 12 months before the functional diagnosis. Thus, immune cells support airway remodeling through the production of MMP-9. Plasma MMP-9 is a potential predictive biomarker of CLAD.


Subject(s)
Biomarkers/blood , Epithelial Cells/immunology , Graft Rejection/diagnosis , Lung Diseases/complications , Lung Transplantation/adverse effects , Matrix Metalloproteinase 9/blood , Receptors, CCR2/metabolism , T-Lymphocytes/immunology , Adult , Allografts , Bronchi/immunology , Bronchi/metabolism , Bronchi/pathology , Chronic Disease , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Follow-Up Studies , Graft Rejection/blood , Graft Rejection/etiology , Graft Survival/immunology , Humans , Longitudinal Studies , Lung Diseases/surgery , Male , Middle Aged , Postoperative Complications , Prognosis , Risk Factors , T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL