Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 55
Filtrer
1.
Am J Transplant ; 24(6): 944-953, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38403187

RÉSUMÉ

Chronic lung allograft dysfunction (CLAD) remains one of the major limitations to long-term survival after lung transplantation. We modified a murine model of CLAD and transplanted left lungs from BALB/c donors into B6 recipients that were treated with intermittent cyclosporine and methylprednisolone postoperatively. In this model, the lung allograft developed acute cellular rejection on day 15 which, by day 30 after transplantation, progressed to severe pleural and peribronchovascular fibrosis, reminiscent of changes observed in restrictive allograft syndrome. Lung transplantation into splenectomized B6 alymphoplastic (aly/aly) or splenectomized B6 lymphotoxin-ß receptor-deficient mice demonstrated that recipient secondary lymphoid organs, such as spleen and lymph nodes, are necessary for progression from acute cellular rejection to allograft fibrosis in this model. Our work uncovered a critical role for recipient secondary lymphoid organs in the development of CLAD after pulmonary transplantation and may provide mechanistic insights into the pathogenesis of this complication.


Sujet(s)
Modèles animaux de maladie humaine , Rejet du greffon , Transplantation pulmonaire , Souris de lignée BALB C , Souris de lignée C57BL , Animaux , Souris , Rejet du greffon/étiologie , Rejet du greffon/anatomopathologie , Transplantation pulmonaire/effets indésirables , Allogreffes , Évolution de la maladie , Fibrose , Maladie chronique , Survie du greffon , Mâle , Tissu lymphoïde/anatomopathologie
2.
Am J Transplant ; 23(2): 202-213, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36804130

RÉSUMÉ

Heart transplant and recipient survival are limited by immune cell-mediated injury of the graft vasculature. We examined the role of the phosphoinositide 3-kinase-ß (PI3Kß) isoform in endothelial cells (EC) during coronary vascular immune injury and repair in mice. In minor histocompatibility-antigen mismatched allogeneic heart grafts, a robust immune response was mounted to each wild-type, PI3Kß inhibitor-treated, or endothelial-selective PI3Kß knockout (ECßKO) graft transplanted to wild-type recipients. However, microvascular EC loss and progressive occlusive vasculopathy only developed in control, but not PI3Kß-inactivated hearts. We observed a delay in inflammatory cell infiltration of the ECßKO grafts, particularly in the coronary arteries. Surprisingly, this was accompanied by an impaired display of proinflammatory chemokine and adhesion molecules by the ECßKO ECs. In vitro, tumor necrosis factor α-stimulated endothelial ICAM1 and VCAM1 expression was blocked by PI3Kß inhibition or RNA interference. Selective PI3Kß inhibition also blocked tumor necrosis factor α-stimulated degradation of inhibitor of nuclear factor kappa Bα and nuclear translocation of nuclear factor kappa B p65 in EC. These data identify PI3Kß as a therapeutic target to reduce vascular inflammation and injury.


Sujet(s)
Cellules endothéliales , Lésions du système vasculaire , Souris , Animaux , Cellules endothéliales/anatomopathologie , Phosphatidylinositol 3-kinase , Phosphatidylinositol 3-kinases , Lésions du système vasculaire/anatomopathologie , Facteur de nécrose tumorale alpha
3.
Am J Transplant ; 22 Suppl 4: 12-17, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-36453706

RÉSUMÉ

Outcomes following heart transplantation remain suboptimal with acute and chronic rejection being major contributors to poor long-term survival. IL-6 is increasingly recognized as a critical pro-inflammatory cytokine involved in allograft injury and has been shown to play a key role in regulating the inflammatory and alloimmune responses following heart transplantation. Therapies that inhibit IL-6 signaling have emerged as promising strategies to prevent allograft rejection. Here, we review experimental and pre-clinical evidence that supports the potential use of IL-6 signaling blockade to improve outcomes in heart transplant recipients.


Sujet(s)
Transplantation cardiaque , Interleukine-6 , Coeur , Transplantation cardiaque/effets indésirables , Cytokines , Allogreffes
5.
Am J Transplant ; 21(11): 3550-3560, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34014614

RÉSUMÉ

Chronic rejection is among the most pressing clinical challenges in solid organ transplantation. Interestingly, in a mouse model of heterotopic heart transplantation, antibody-dependent, natural killer (NK) cell-mediated chronic cardiac allograft vasculopathy occurs in some donor-recipient strain combinations, but not others. In this study, we sought to identify the mechanism underlying this unexplained phenomenon. Cardiac allografts from major histocompatibility complex (MHC) mismatched donors were transplanted into immune-deficient C57Bl/6.rag-/- recipients, followed by administration of a monoclonal antibody against the donor MHC class I antigen. We found marked allograft vasculopathy in hearts from C3H donors, but near-complete protection of BALB/c allografts from injury. We found no difference in recipient NK cell phenotype or intrinsic responsiveness to activating signals between recipients of C3H versus BALB/c allografts. However, cardiac endothelial cells from C3H allografts showed an approximately twofold higher expression of Rae-1, an activating ligand of the NK cell receptor natural killer group 2D (NKG2D). Importantly, the administration of a neutralizing antibody against NKG2D abrogated the development of allograft vasculopathy in recipients of C3H allografts, even in the presence of donor-specific antibodies. Therefore, the activating NK cell receptor NKG2D is necessary in this model of chronic cardiac allograft vasculopathy, and strain-dependent expression of NK activating ligands correlates with the development of this disease.


Sujet(s)
Transplantation cardiaque , Sous-famille K des récepteurs de cellules NK de type lectine , Animaux , Anticorps monoclonaux , Cellules endothéliales , Rejet du greffon/étiologie , Transplantation cardiaque/effets indésirables , Souris , Souris de lignée BALB C , Souris de lignée C3H , Souris de lignée C57BL , Récepteurs de cellules tueuses naturelles
6.
Am J Transplant ; 21(9): 3088-3100, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-33445219

RÉSUMÉ

The 2007 Banff working classification of skin-containing Tissue Allograft Pathology addressed only acute T cell-mediated rejection in skin. We report the longitudinal long-term histological follow-up of six face transplant recipients, focusing on chronic and mucosal rejection. We identified three patterns suggestive of chronic rejection (lichen planus-like, vitiligo-like and scleroderma-like). Four patients presented lichen planus-like and vitiligo-like chronic rejection at 52 ± 17 months posttransplant with severe concomitant acute T cell-mediated rejection. After lichen planus-like rejection, two patients developed scleroderma-like alterations. Graft vasculopathy with C4d deposits and de novo DSA led to subsequent graft loss in one patient. Chronic active rejection was frequent and similar patterns were noted in mucosae. Concordance between 124 paired skin and mucosal biopsies acute rejection grades was low (κ = 0.2, p = .005) but most grade 0/I mucosal rejections were associated with grade 0/I skin rejections. We defined discordant (grade≥II mucosal rejection and grade 0/I skin rejection) (n = 55 [70%]) and concordant (grade≥II rejection in both biopsies) groups. Mucosal biopsies of the discordant group displayed lower intra-epithelial GranzymeB/FoxP3 ratios suggesting a less aggressive phenotype (p = .08). The grading system for acute rejection in mucosa may require phenotyping. Whether discordant infiltrates reflect a latent allo-immune reaction leading to chronic rejection remains an open question.


Sujet(s)
Transplantation de la face , Transplantation rénale , Biopsie , Études de suivi , Rejet du greffon/étiologie , Humains , Muqueuse
7.
Am J Transplant ; 21(1): 362-371, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32885581

RÉSUMÉ

Chronic lung allograft dysfunction (CLAD) is the major barrier to long-term survival following lung transplantation, and new mechanistic biomarkers are needed. Lymphocytic bronchitis (LB) precedes CLAD and has a defined molecular signature. We hypothesized that this LB molecular signature would be associated with CLAD in small airway brushings independent of infection. We quantified RNA expression from small airway brushings and transbronchial biopsies, using RNAseq and digital RNA counting, respectively, for 22 CLAD cases and 27 matched controls. LB metagene scores were compared across CLAD strata by Wilcoxon rank sum test. We performed unbiased host transcriptome pathway and microbial metagenome analysis in airway brushes and compared machine-learning classifiers between the two tissue types. This LB metagene score was increased in CLAD airway brushes (p = .002) and improved prediction of graft failure (p = .02). Gene expression classifiers based on airway brushes outperformed those using transbronchial biopsies. While infection was associated with decreased microbial alpha-diversity (p ≤ .04), neither infection nor alpha-diversity was associated with LB gene expression. In summary, CLAD was associated with small airway gene expression changes not apparent in transbronchial biopsies in this cohort. Molecular analysis of airway brushings for diagnosing CLAD merits further examination in multicenter cohorts.


Sujet(s)
Rejet du greffon , Transplantation pulmonaire , Allogreffes , Rejet du greffon/génétique , Humains , Inflammation/génétique , Poumon , Transplantation pulmonaire/effets indésirables
9.
Am J Transplant ; 21(4): 1641-1649, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33141487

RÉSUMÉ

Antibody-mediated rejection (AMR) that resists to standard of care (SOC) therapy remains a major challenge after kidney transplantation and leads to graft failure in a majority of cases. The use of anti-IL6 receptor antibodies was suggested to treat chronic antibody-mediated rejection (cAMR) after failure of classical treatments. We treated nine patients with AMR resistant to apheresis, rituximab, and intravenous immunoglobulins, with a monthly infusion of tocilizumab and compared them with a historical cohort of 37 patients with similar clinical, immunological, and histological characteristics. The 1-year graft survival and the decline in renal function did not differ between patients who received tocilizumab and those who did not. Histological follow-up showed that despite a decrease in inflammation and tubulitis scores after tocilizumab, the course of antibody-mediated lesions and chronic glomerulopathy were similar in both groups. In our study, the addition of monthly infusions of tocilizumab did not alter the course of AMR that resist to SOC therapy. Large randomized studies are urgently needed to assess the effect of tocilizumab in this context.


Sujet(s)
Transplantation rénale , Rejet du greffon/traitement médicamenteux , Rejet du greffon/prévention et contrôle , Survie du greffon , Humains , Immunoglobulines par voie veineuse/usage thérapeutique , Alloanticorps , Transplantation rénale/effets indésirables , Rituximab , Norme de soins
10.
Am J Transplant ; 20(10): 2644-2651, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32185874

RÉSUMÉ

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.


Sujet(s)
Bronchiolite oblitérante , Transplantation pulmonaire , Allogreffes , Bronchiolite oblitérante/étiologie , Rejet du greffon/étiologie , Humains , Poumon , Transplantation pulmonaire/effets indésirables , Transplantation homologue
11.
Am J Transplant ; 20(1): 241-249, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31397939

RÉSUMÉ

Acute cellular rejection (ACR) is a significant risk factor for chronic lung allograft dysfunction (CLAD). Although clinically manifest and higher grade (≥A2) ACR is generally treated with augmented immunosuppression, management of minimal (grade A1) ACR remains controversial. In our program, patients with subclinical and spirometrically stable A1 rejection (StA1R) are routinely not treated with augmented immunosuppression. We hypothesized that an untreated first StA1R does not increase the risk of CLAD or death compared to episodes of spirometrically stable no ACR (StNAR). The cohort was drawn from all consecutive adult, first, bilateral lung transplantations performed between 1999 and 2017. Biopsies obtained in the first-year posttransplant were paired with (forced expiratory volume in 1 second FEV1 ). The first occurrence of StA1R was compared to a time-matched StNAR. The risk of CLAD or death was assessed using univariable and multivariable Cox proportional hazards models. The analyses demonstrated no significant difference in risk of CLAD or death in patients with a first StA1R compared to StNAR. This largest study to date shows that, in clinically stable patients, an untreated first A1 ACR in the first-year posttransplant is not significantly associated with an increased risk for CLAD or death. Watchful-waiting approach may be an acceptable tactic for stable A1 episodes in lung transplant recipients.


Sujet(s)
Rejet du greffon/mortalité , Survie du greffon , Maladies pulmonaires/mortalité , Transplantation pulmonaire/mortalité , Complications postopératoires/mortalité , Sujet âgé , Allogreffes , Femelle , Études de suivi , Rejet du greffon/étiologie , Rejet du greffon/anatomopathologie , Humains , Maladies pulmonaires/chirurgie , Transplantation pulmonaire/effets indésirables , Mâle , Adulte d'âge moyen , Complications postopératoires/étiologie , Complications postopératoires/anatomopathologie , Pronostic , Études rétrospectives , Facteurs de risque , Taux de survie
12.
Am J Transplant ; 20(5): 1431-1438, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-31811777

RÉSUMÉ

Cardiac allograft vasculopathy (CAV) is associated with intragraft B cell infiltrates. Here, we studied the clonal composition of B cell infiltrates using 4 graft specimens with CAV. Using deep sequencing, we analyzed the immunoglobulin heavy chain variable region repertoire in both graft and blood. Results showed robust B cell clonal expansion in the graft but not in the blood for all cases. Several expanded B cell clones, characterized by their uniquely rearranged complementarity-determining region 3, were detected in different locations in the graft. Sequences from intragraft B cells also showed elevated levels of mutated rearrangements in the graft compared to blood B cells. The number of somatic mutations per rearrangement was also higher in the graft than in the blood, suggesting that B cells continued maturing in situ. Overall, our studies demonstrated B cell clonal expansion in human cardiac allografts with CAV. This local B cell response may contribute to the pathophysiology of CAV through a mechanism that needs to be identified.


Sujet(s)
Cardiopathies , Transplantation cardiaque , Allogreffes , Lymphocytes B , Rejet du greffon/étiologie , Transplantation cardiaque/effets indésirables , Humains
13.
Am J Transplant ; 20(3): 825-833, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31665560

RÉSUMÉ

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.


Sujet(s)
Remodelage des voies aériennes , Transplantation pulmonaire , Allogreffes , Amphiréguline/génétique , Humains , Poumon , Transplantation pulmonaire/effets indésirables
14.
Am J Transplant ; 19(10): 2705-2718, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31278849

RÉSUMÉ

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.


Sujet(s)
Lymphocytes T CD4+/immunologie , Commerce/normes , Facteurs de transcription Forkhead/physiologie , Rejet du greffon/prévention et contrôle , Maladies pulmonaires/immunologie , Transplantation pulmonaire/effets indésirables , Microbiote , Lymphocytes T régulateurs/immunologie , Allogreffes , Animaux , Lymphocytes T CD4+/microbiologie , Rejet du greffon/étiologie , Rejet du greffon/métabolisme , Survie du greffon/immunologie , Maladies pulmonaires/microbiologie , Maladies pulmonaires/chirurgie , Mâle , Souris , Souris de lignée C57BL , Lymphocytes T régulateurs/microbiologie , Receveurs de transplantation
15.
Am J Transplant ; 19(12): 3356-3366, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31152474

RÉSUMÉ

We determined peripheral blood (PB) and biopsy (Bx) RNA expression signatures in a Brazilian and US cohort of kidney transplant patients. Phenotypes assigned by precise histology were: acute rejection (AR), interstitial fibrosis/tubular atrophy/chronic rejection (CR), excellent functioning transplants (TX), and glomerulonephritis recurrence (GN). Samples were analyzed on microarrays and profiles from each cohort were cross-validated on the other cohort with similar phenotypes. We discovered signatures for each tissue: (1) AR vs TX, (2) CR vs TX, and (3) GN vs TX using the Random Forests algorithm. We validated biopsies signatures of AR vs TX (area under the curve [AUC] 0.97) and CR vs TX (AUC 0.87). We also validated both PB and Bx signatures of AR vs TX and CR vs TX with varying degrees of accuracy. Several biological pathways were shared between AR and CR, suggesting similar rejection mechanisms in these 2 clinical phenotypes. Thus, we identified gene expression signatures for AR and CR in transplant patients and validated them in independent cohorts of significantly different racial/ethnic backgrounds. These results reveal that there are strong unifying immune mechanisms driving transplant diseases and identified in the signatures discovered in each cohort, suggesting that molecular diagnostics across populations are feasible despite ethnic and environmental differences.


Sujet(s)
Marqueurs biologiques/analyse , Ethnies/génétique , Rejet du greffon/diagnostic , Défaillance rénale chronique/chirurgie , Transplantation rénale/effets indésirables , Agranulocytes/métabolisme , Transcriptome , Adolescent , Adulte , Sujet âgé , Biopsie , Études de cohortes , Femelle , Études de suivi , Analyse de profil d'expression de gènes , Rejet du greffon/sang , Rejet du greffon/étiologie , Survie du greffon , Humains , Mâle , Adulte d'âge moyen , Pronostic , Jeune adulte
16.
Am J Transplant ; 19(4): 1168-1177, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30312535

RÉSUMÉ

Face vascularized composite allografts (FVCAs) have helped patients with severe facial disfigurement, with acute rejection now largely controlled through iatrogenic immunosuppression. However, little is known regarding the incidence and mechanism(s) of more long-term pathologic alterations in FVCAs that may affect function and graft durability. Protocol surveillance biopsy specimens for up to an 8-year interval in 7 patients who received FVCAs at our institution revealed histopathologic evidence of chronic rejection. Clinical manifestations included features of premature aging, mottled leukoderma accentuating suture lines, telangiectasia, and dryness of nasal mucosa. Pathologic changes consisted of epidermal thinning accompanied by discrete foci of lymphocyte-mediated cytotoxicity, hyperkeratosis, follicular plugging, vascular ectasia, and sclerosis beneath the epidermal layer associated with collagen type I deposition. Genomic interrogation and immunohistochemistry of sclerotic zones revealed upregulation of the AP-1 pathway components, JunB and c-Fos, previously implicated in overproduction of type I dermal collagen in the setting of systemic sclerosis. We conclude that some patients develop chronic rejection in FVCAs with striking similarities to alterations seen in certain autoimmune cutaneous disorders (lupus erythematosus and scleroderma/chronic sclerodermoid graft-versus-host disease). Identification of relevant pathways and genes, such as JunB and c-Fos, may provide new targets for preventative therapies for chronic immune-mediated changes in vascularized composite allografts.


Sujet(s)
Allogreffes de tissus composites/immunologie , Transplantation de la face/méthodes , Rejet du greffon , Adulte , Maladie chronique , Femelle , Analyse de profil d'expression de gènes , Humains , Immunosuppresseurs/usage thérapeutique , Mâle , Adulte d'âge moyen
17.
Am J Transplant ; 19(6): 1671-1683, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30412654

RÉSUMÉ

We aimed to determine the long-term outcomes of eculizumab-treated, positive crossmatch (+XM) kidney transplant recipients compared with +XM and age-matched negative crossmatch (-XM) controls. We performed an observational retrospective study and examined allograft survival, histologic findings, long-term B-cell flow cytometric XM (BFXM), and allograft-loss-associated factors. The mean (SD) posttransplant follow-up was 6.3 (2.5) years in the eculizumab group; 7.6 (3.5), +XM control group; 7.9 (2.5), -XM control group. The overall and death-censored allograft survival rates were similar in +XM groups (P = .73, P = .48) but reduced compared with -XM control patients (P < .001, P < .001). In the eculizumab-treated group, 57.9% (11/19) of the allografts had chronic antibody-mediated rejection, but death-censored allograft survival was 76.6%, 5 years; 75.4%, 7 years. Baseline IgG3 positivity and BFXM ≥300 were associated with allograft loss. C1q positivity was also associated with allograft loss but did not reach statistical significance. Donor-specific antibodies appeared to decrease in eculizumab-treated patients. After excluding patients with posttransplant plasmapheresis, 42.3% (9/21) had negative BFXMs; 31.8% (7/22), completely negative single-antigen beads 1 year posttransplant. Eculizumab-treated +XM patients had reduced allograft survival compared with -XM controls but similar survival to +XM controls. BFXM and complement-activating donor-specific antibodies (by IgG3 and C1q testing) may be used for risk stratification in +XM transplantation.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Inhibiteurs du complément/usage thérapeutique , Transplantation rénale/méthodes , Adulte , Allogreffes , Lymphocytes B/immunologie , Études cas-témoins , Complément C1q/métabolisme , Femelle , Rejet du greffon/immunologie , Rejet du greffon/prévention et contrôle , Survie du greffon/immunologie , Antigènes HLA/immunologie , Test d'histocompatibilité , Humains , Alloanticorps/sang , Rein/immunologie , Rein/anatomopathologie , Transplantation rénale/effets indésirables , Mâle , Adulte d'âge moyen , Études rétrospectives , Facteurs temps , Donneurs de tissus , Résultat thérapeutique
18.
Am J Transplant ; 19(1): 247-258, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30378739

RÉSUMÉ

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.


Sujet(s)
Bronchiolite oblitérante/étiologie , Rejet du greffon/étiologie , Maladies pulmonaires/chirurgie , Transplantation pulmonaire/effets indésirables , Transplantation pulmonaire/méthodes , Allogreffes/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Fibrose , Inflammation , Isoantigènes , Poumon/immunologie , Poumon/anatomopathologie , Maladies pulmonaires/immunologie , Mâle , Souris , Souris de lignée C57BL , Phénotype , Complications postopératoires/anatomopathologie , Donneurs de tissus , Transplantation homologue/effets indésirables
19.
Am J Transplant ; 18(12): 2849-2856, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30133953

RÉSUMÉ

The Banff classification of renal allograft pathology defines specific morphologic lesions that are used in the diagnosis of active (glomerulitis, peritubular capillaritis, endarteritis) and chronic (transplant glomerulopathy, peritubular capillary basement membrane multilayering, transplant arteriopathy) antibody-mediated rejection (ABMR). However, none of these individual lesions are specific for ABMR, and for this reason Banff requires 1 or more additional findings, including C4d deposition in peritubular capillaries, presence of circulating donor-specific antibodies (DSAs), and/or expression in the tissue of transcripts strongly associated with ABMR, for a definitive diagnosis of ABMR to be made. In addition, while animal studies examining serial biopsies have established the progression of morphologic lesions of active to chronic ABMR as well as intermediate forms (chronic active ABMR) exhibiting features of both, clear documentation that lesions of chronic ABMR require the earlier presence of corresponding active and intermediate lesions is less well established in human renal allografts. This review examines temporal relationships between key morphologic lesions of active and chronic ABMR in biopsies of human grafts, likely intermediate forms, and findings for and possibly against direct and potentially interruptible progression from active to chronic lesions.


Sujet(s)
Rejet du greffon/étiologie , Rejet du greffon/anatomopathologie , Alloanticorps/effets indésirables , Transplantation rénale/effets indésirables , Donneurs de tissus , Humains , Pronostic
20.
Am J Transplant ; 18(12): 3045-3059, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30019840

RÉSUMÉ

Pirfenidone may attenuate the decline of pulmonary function in restrictive allograft syndrome (RAS) after lung transplantation. We retrospectively assessed all lung transplant recipients with RAS who were treated with pirfenidone for at least 3 months (n = 11) in our lung transplant center and report on their long-term outcomes following initiation of pirfenidone. Main outcome parameters included evolution of pulmonary function and overall survival. Pirfenidone appears to attenuate the decline in forced vital capacity and forced expiratory volume in 1 second. Notably, 3 patients were bridged to redo-transplantation with pirfenidone for 11 (5-12) months and are currently alive, while 3 other patients demonstrate long-term stabilization of pulmonary function after 26.6 (range 18.4-46.6) months of treatment. Median overall 3-year survival after RAS diagnosis was 54.5%. Subjective intolerance, mainly anorexia and nausea, necessitating pirfenidone dose de-escalation in 55% of patients, as well as calcineurin dose increase requirements with about 20% are important complications during pirfenidone treatment after lung transplantation. Our findings provide further evidence that pirfenidone appears to be safe and may attenuate the rate of decline in lung function in patients with RAS, but the actual clinical benefit cannot be assessed in the context of this study design and requires further investigation in a larger randomized trial.


Sujet(s)
Rejet du greffon/prévention et contrôle , Survie du greffon/effets des médicaments et des substances chimiques , Maladies pulmonaires/chirurgie , Transplantation pulmonaire/effets indésirables , Dysfonction primaire du greffon/prévention et contrôle , Fibrose pulmonaire/prévention et contrôle , Pyridones/usage thérapeutique , Allogreffes , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Femelle , Études de suivi , Rejet du greffon/étiologie , Humains , Mâle , Adulte d'âge moyen , Complications postopératoires/étiologie , Complications postopératoires/prévention et contrôle , Dysfonction primaire du greffon/étiologie , Pronostic , Fibrose pulmonaire/étiologie , Études rétrospectives , Facteurs de risque , Syndrome
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...