Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Front Immunol ; 15: 1369536, 2024.
Article in English | MEDLINE | ID: mdl-38736881

ABSTRACT

Introduction: Chronic rejection is a major complication post-transplantation. Within lung transplantation, chronic rejection was considered as airway centred. Chronic Lung Allograft Dysfunction (CLAD), defined to cover all late chronic complications, makes it more difficult to understand chronic rejection from an immunological perspective. This study investigated the true nature, timing and location of chronic rejection as a whole, within mouse lung transplantation. Methods: 40 mice underwent an orthotopic left lung transplantation, were sacrificed at day 70 and evaluated by histology and in vivo µCT. For timing and location of rejection, extra grafts were sacrificed at day 7, 35, 56 and investigated by ex vivo µCT or single cell RNA (scRNA) profiling. Results: Chronic rejection originated as innate inflammation around small arteries evolving toward adaptive organization with subsequent end-arterial fibrosis and obliterans. Subsequently, venous and pleural infiltration appeared, followed by airway related bronchiolar folding and rarely bronchiolitis obliterans was observed. Ex vivo µCT and scRNA profiling validated the time, location and sequence of events with endothelial destruction and activation as primary onset. Conclusion: Against the current belief, chronic rejection in lung transplantation may start as an arterial response, followed by responses in venules, pleura, and, only in the late stage, bronchioles, as may be seen in some but not all patients with CLAD.


Subject(s)
Graft Rejection , Lung Transplantation , Animals , Lung Transplantation/adverse effects , Graft Rejection/immunology , Mice , Chronic Disease , Disease Models, Animal , Mice, Inbred C57BL , Lung/pathology , Lung/immunology , Male , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/pathology
2.
Semin Respir Crit Care Med ; 34(3): 352-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23821509

ABSTRACT

Lung transplantation is currently considered as an ultimate live-saving treatment for selected patients suffering from end-stage pulmonary disease. Long-term survival, however, is hampered by chronic rejection, or chronic lung allograft dysfunction (CLAD). Recently, various phenotypes within CLAD have been identified, challenging the established clinical definition of bronchiolitis obliterans syndrome (BOS). Some patients with presumed BOS, for instance, demonstrate an important improvement in forced expiratory volume in the first second of expiration (FEV1) after treatment with azithromycin. These patients are characterized by the presence of excess (≥ 15%) bronchoalveolar lavage (BAL) neutrophils, in absence of concurrent infection. This phenotype of CLAD has been redefined as neutrophilic reversible allograft dysfunction (NRAD), and these patients generally have a very good prognosis after diagnosis. Another group of patients with CLAD develop a restrictive rather than an obstructive pulmonary function defect (defined as a decline in total lung capacity of at least 10%) and demonstrate persistent interstitial and ground-glass opacities on chest computed tomographic (CT) scan. This phenotype is called restrictive allograft syndrome (RAS), and patients with RAS have a much worse prognosis after diagnosis. This review further discusses both of these CLAD phenotypes that do not fit the classical definition of BOS. Potential pathophysiological mechanisms, etiology, diagnosis, prognosis, and treatments are discussed.


Subject(s)
Bronchiolitis Obliterans/etiology , Lung Transplantation , Primary Graft Dysfunction/physiopathology , Animals , Bronchiolitis Obliterans/diagnosis , Bronchiolitis Obliterans/physiopathology , Chronic Disease , Forced Expiratory Volume , Graft Rejection/physiopathology , Humans , Lung Diseases/physiopathology , Lung Diseases/surgery , Neutrophils/metabolism , Prognosis , Survival , Syndrome , Tomography, X-Ray Computed
3.
Transplantation ; 95(9): 1167-72, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23425818

ABSTRACT

BACKGROUND: Chronic rejection is the major problem hampering long-term survival after lung transplantation. Recently, it became clear that patients may develop an obstructive (bronchiolitis obliterans syndrome [BOS]) or a restrictive lung function defect (restrictive allograft syndrome [RAS]), for which specific risk factors are unknown. METHODS: A retrospective analysis of our lung transplantation cohort was performed (n=380). Patients with an irreversible decline in forced expiratory volume in 1 second were identified and classified as BOS or RAS. Patient characteristics, bronchoalveolar lavage (BAL) cellularity, rates of respiratory tract infection, colonization, acute rejection, and lymphocytic bronchiolitis were compared between BOS, RAS, and stable patients. RESULTS: There were 103 patients suffering from chronic rejection, of which 79 had BOS and 24 were diagnosed with RAS. There were more patients with infection and pseudomonal colonizations in BOS and RAS compared with control (P=0.0090 and P=0.0034, respectively). More patients ever experienced acute and severe acute rejections (A≥2; both P<0.0001) and lymphocytic bronchiolitis (P=0.0006) in BOS and RAS versus control. There were more patients experiencing severe lymphocytic bronchiolitis in RAS compared with BOS (P=0.031). BAL neutrophilia in BOS and RAS were elevated at days 360, 540, and 720 versus control. BOS, but especially RAS patients, experienced more frequent episodes of increased BAL eosinophilia (≥2%; P<0.0001). CONCLUSION: Acute rejection, lymphocytic bronchiolitis, colonization with pseudomonas, infection, and BAL eosinophilia and neutrophilia are risk factors for the later development not only of RAS but also of BOS.


Subject(s)
Bronchiolitis Obliterans/etiology , Lung Diseases/etiology , Lung Transplantation/adverse effects , Adult , Azithromycin/therapeutic use , C-Reactive Protein/analysis , Female , Graft Rejection , Humans , Immunosuppressive Agents/therapeutic use , Interleukin-8/blood , Male , Middle Aged , Retrospective Studies , Risk Factors , Transplantation, Homologous/adverse effects
4.
J Heart Lung Transplant ; 32(4): 447-53, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23415729

ABSTRACT

BACKGROUND: Interleukin-17 (IL-17) is involved in autoimmune and chronic pulmonary diseases and linked with neutrophilic inflammation. Azithromycin reduces and prevents broncholaveolar lavage (BAL) neutrophilia after lung transplantation (LTx). In this investigation we assessed the involvement of IL-17 in different post-transplant complications in human LTx biopsies. METHODS: Immunohistochemical staining against IL-17A was performed on biopsies of LTx patients with either chronic rejection, acute A-grade rejection (A > 2B0), lymphocytic bronchiolitis (LB), infection, and stable patients. Biopsies of 7 patients with LB were stained before and after azithromycin treatment. IL-17+ cells were quantified as number per square millimeter of lamina propria. Double staining with CD4/CD8 was performed to determine the origin of IL-17. RESULTS: In the LB group, biopsies showed a significant presence of IL-17+ cells/mm2 of lamina propria compared with the stable, acute A-grade/chronic rejection and infection groups (p < 0.0001). The number of IL-17+ cells on biopsy correlated with percent BAL (%BAL) neutrophilia (r = 0.34, p = 0.0056). Azithromycin reduced both %BAL neutrophilia and IL-17+ cells (both p = 0.016) in the LB group. CD8+ cells were the major source of IL-17. CONCLUSIONS: IL-17+ / CD8+ cells are present in LB after LTx but not in acute A-grade/chronic rejection nor during infection. Moreover, azithromycin significantly decreased the number of IL-17+ cells in the airway wall, which may further explain its effect on BAL neutrophilia.


Subject(s)
Bronchiolitis/immunology , Interleukin-17/physiology , Lung Transplantation , Lymphocytes/immunology , Postoperative Complications/immunology , Adult , Azithromycin/pharmacology , Female , Humans , Male , Middle Aged , Neutrophils/drug effects , Retrospective Studies
5.
Cell Biochem Biophys ; 67(2): 331-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22205500

ABSTRACT

The airways in asthma and COPD are characterized by an increase in airway smooth muscle (ASM) mass and bronchial vascular changes associated with increased expression of pro-angiogenic growth factors, such as fibroblast growth factors (FGF-1 and FGF-2) and vascular endothelial growth factor (VEGF). We investigated the contribution of FGF-1/-2 in VEGF production in ASM cells and assessed the influence of azithromycin and dexamethasone and their underlying signaling mechanisms. Growth-synchronized human ASM cells were pre-treated with MAPK inhibitors, U0126 for ERK1/2(MAPK) and SB239063 for p38(MAPK) as well as with dexamethasone or azithromycin, 30 min before incubation with FGF-1 or FGF-2. Expression of VEGF (VEGF-A, VEGF121, and VEGF165) was assessed by quantitative PCR, VEGF release by ELISA and MAPK phosphorylation by Western blotting. Both FGF-1 and FGF-2 significantly induced mRNA levels of VEGF-A, VEGF121, and VEGF165. The VEGF protein release was increased 1.8-fold (FGF-1) and 5.5-fold (FGF-2) as compared to controls. Rapid transient increase in ERK1/2(MAPK) and p38(MAPK) phosphorylation and subsequent release of VEGF from FGF-1 or FGF-2-treated ASM cells were inhibited by respective blockers. Furthermore, azithromycin and dexamethasone significantly reduced both the VEGF release and the activation of p38(MAPK) pathway in response to FGF-1 or FGF-2 treatment. Our Results demonstrate that FGF-1 and FGF-2 up-regulate VEGF production via ERK1/2(MAPK) and p38(MAPK) pathways. Both azithromycin and dexamethasone elicited their anti-angiogenic effects via p38(MAPK) pathway in vitro, thereby suggesting a possible therapeutic approach to tackle VEGF-mediated vascular remodeling.


Subject(s)
Azithromycin/pharmacology , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/pharmacology , MAP Kinase Signaling System/drug effects , Trachea/cytology , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Dexamethasone/pharmacology , Enzyme Activation/drug effects , Fibroblast Growth Factor 1/antagonists & inhibitors , Fibroblast Growth Factor 2/antagonists & inhibitors , Gene Expression Regulation/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Time Factors , Vascular Endothelial Growth Factor A/genetics
6.
Transplantation ; 94(2): 101-9, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22461039

ABSTRACT

Chronic lung allograft rejection is the single most important cause of death in lung transplant recipients after the first postoperative year, resulting in a 5-year survival rate of approximately 50%, which is far behind that of other solid organ transplantations. Spirometry is routinely used as a clinical marker for assessing pulmonary allograft function and diagnosing chronic lung allograft rejection after lung transplantation (LTx). As such, a progressive obstructive decline in pulmonary allograft function (forced expiratory volume in 1 sec [FEV1]) in absence of all other causes (currently defined as bronchiolitis obliterans syndrome [BOS]) is considered to reflect the evolution of chronic lung allograft rejection. BOS has a 5-year prevalence of approximately 45% and is thought to be the final common endpoint of various alloimmunologic and nonalloimmunologic injuries to the pulmonary allograft, triggering different innate and adaptive immune responses. Most preventive and therapeutic strategies for this complex process have thus far been largely unsuccessful. However, the introduction of the neomacrolide antibiotic azithromycin (AZI) in the field of LTx as of 2003 made it clear that some patients with established BOS might in fact benefit from such therapy due to its various antiinflammatory and immunomodulatory properties, as summarized in this review. Particularly in patients with an increased bronchoalveolar lavage neutrophilia (i.e., 15%-20% or more), AZI treatment could result in an increase in FEV1 of at least 10%. More recently, it has become clear that prophylactic therapy with AZI actually may prevent BOS and improve FEV1 after LTx, most likely through its interactions with the innate immune system. However, one should always be aware of possible adverse effects related to AZI when implementing this drug as prophylactic or long-term treatment. Even so, AZI therapy after LTx can generally be considered as safe.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Azithromycin/therapeutic use , Graft Rejection/drug therapy , Immunologic Factors/therapeutic use , Lung Transplantation/adverse effects , Bronchiolitis Obliterans/drug therapy , Bronchiolitis Obliterans/etiology , Chronic Disease , Graft Rejection/etiology , Graft Rejection/prevention & control , Humans , Transplantation, Homologous
7.
PLoS One ; 7(1): e29802, 2012.
Article in English | MEDLINE | ID: mdl-22238655

ABSTRACT

Almost all animal models for chronic rejection (CR) after lung transplantation (LTx) fail to resemble the human situation. It was our attempt to develop a representative model of CR in mice. Orthotopic LTx was performed in allografts receiving daily immunosuppression with steroids and cyclosporine. Controls included isografts and mice only undergoing thoracotomy (SHAM). Allografts were sacrificed 2, 4, 6, 8, 10 or 12 weeks after LTx. Pulmonary function was measured repeatedly in the 12w allografts, isografts and SHAM mice. Histologically, all allografts demonstrated acute rejection (AR) around the blood vessels and airways two weeks after LTx. This decreased to 50-75% up to 10 weeks and was absent after 12 weeks. Obliterative bronchiolitis (OB) lesions were observed in 25-50% of the mice from 4-12 weeks. Isografts and lungs of SHAM mice were normal after 12 weeks. Pulmonary function measurements showed a decline in FEV(0.1), TLC and compliance in the allografts postoperatively (2 weeks) with a slow recovery over time. After this initial decline, lung function of allografts increased more than in isografts and SHAM mice indicating that pulmonary function measurement is not a good tool to diagnose CR in a mouse. We conclude that a true model for CR, with clear OB lesions in about one third of the animals, but without a decline in lung function, is possible. This model is an important step forward in the development of an ideal model for CR which will open new perspectives in unraveling CR pathogenesis and exploring new treatment options.


Subject(s)
Bronchiolitis Obliterans/pathology , Bronchiolitis Obliterans/therapy , Graft Rejection/pathology , Lung Transplantation/pathology , Animals , Chronic Disease , Disease Models, Animal , Humans , Lung Transplantation/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Syndrome , Time Factors , Transplantation, Homologous
8.
Transplantation ; 93(2): 224-9, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22134370

ABSTRACT

BACKGROUND: Vitamin D deficiency has been reported in different chronic pulmonary diseases like asthma and chronic obstructive pulmonary disease, but little is known in lung transplant recipients. METHODS: Serum 25-hydroxyvitamin D (25-OHD) levels and pulmonary function (forced expiratory volume in 1 sec [FEV(1)] %predicted) were measured in 131 lung transplant patients during their yearly posttransplant check-up hospital stay, and the total number of infections and perivascular/peribronchiolar rejections were assessed from transplantation on. RESULTS: Vitamin D deficiency (<30 ng/mL) occurred in 62 of 131 patients (47.3%), of whom 26 (19.8%) were severely deficient (<20 ng/mL). The FEV(1) was significantly lower in the deficient group compared with the group with normal levels (P=0.019). Moreover, we could find an association between FEV(1) and 25-OHD levels in univariate analysis (P=0.018), which remained significant in multivariate analysis (P=0.012). The same holds true for the association between 25-OHD levels and the peak postoperative FEV(1) (P=0.021 in multivariate analysis). We also identified significantly more patients with moderate to severe B-grade rejections in the deficient group (P=0.0038). CONCLUSION: Vitamin D deficiency is present in 47% of our lung transplant patients and seems independently associated with a lower FEV(1) and more severe B-grade rejections. This study raises the potential need for additional vitamin D treatment in lung transplantation and clearly indicates the role of a randomized placebo-controlled trial with vitamin D supplementation, which is ongoing in our center.


Subject(s)
Lung Transplantation/adverse effects , Vitamin D Deficiency/complications , Adult , Female , Follow-Up Studies , Forced Expiratory Volume , Graft Rejection/etiology , Humans , Lung Diseases/complications , Lung Diseases/physiopathology , Lung Diseases/surgery , Lung Transplantation/physiology , Male , Middle Aged , Vitamin D/administration & dosage , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...