Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Eur Respir J ; 60(6)2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36104292

RESUMO

BACKGROUND: Patients who present to an emergency department (ED) with respiratory symptoms are often conservatively triaged in favour of hospitalisation. We sought to determine if an inflammatory biomarker panel that identifies the host response better predicts hospitalisation in order to improve the precision of clinical decision making in the ED. METHODS: From April 2020 to March 2021, plasma samples of 641 patients with symptoms of respiratory illness were collected from EDs in an international multicentre study: Canada (n=310), Italy (n=131) and Brazil (n=200). Patients were followed prospectively for 28 days. Subgroup analysis was conducted on confirmed coronavirus disease 2019 (COVID-19) patients (n=245). An inflammatory profile was determined using a rapid, 50-min, biomarker panel (RALI-Dx (Rapid Acute Lung Injury Diagnostic)), which measures interleukin (IL)-6, IL-8, IL-10, soluble tumour necrosis factor receptor 1 (sTNFR1) and soluble triggering receptor expressed on myeloid cells 1 (sTREM1). RESULTS: RALI-Dx biomarkers were significantly elevated in patients who required hospitalisation across all three sites. A machine learning algorithm that was applied to predict hospitalisation using RALI-Dx biomarkers had a mean±sd area under the receiver operating characteristic curve of 76±6% (Canada), 84±4% (Italy) and 86±3% (Brazil). Model performance was 82±3% for COVID-19 patients and 87±7% for patients with a confirmed pneumonia diagnosis. CONCLUSIONS: The rapid diagnostic biomarker panel accurately identified the need for inpatient care in patients presenting with respiratory symptoms, including COVID-19. The RALI-Dx test is broadly and easily applicable across many jurisdictions, and represents an important diagnostic adjunct to advance ED decision-making protocols.


Assuntos
COVID-19 , Infecções Respiratórias , Humanos , COVID-19/diagnóstico , Curva ROC , Biomarcadores , Serviço Hospitalar de Emergência , Interleucina-6
2.
Transpl Immunol ; 69: 101467, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34547417

RESUMO

IL-17A is implicated in the pathogenesis of chronic lung allograft dysfunction, which limits survival after lung transplantation. While many cells express the IL-17 receptor A (IL-17RA) which is the main receptor for IL-17A, the cellular targets of IL-17A in development of post-transplant fibrosis are unknown. The purpose of this study was to determine whether IL-17RA expression by donor or recipient structural or bone marrow (BM) cells is required for the development of allograft fibrosis in a mouse intrapulmonary tracheal transplantation (IPTT) model. BM chimeras were generated using C57BL/6 and IL-17RA-knockout mice. After engraftment, allogeneic IPTTs were performed using the chimeric and BALB/c mice as donors or recipients. This allowed us to assess the effect of IL-17RA deficiency in recipient BM, recipient structural, donor BM, or donor structural compartments separately. Tracheal grafts, the surrounding lung, and mediastinal lymph nodes were assessed 28 days after IPTT. Only recipient BM IL-17RA deficiency resulted in attenuation of tracheal graft obliteration. In the setting of recipient BM IL-17RA deficiency, T cells and neutrophils were decreased in mediastinal lymph nodes. Additionally, recipient BM IL-17RA deficiency was associated with increased B220+PNAd+ lymphoid aggregates, consistent with tertiary lymphoid organs, in proximity to the tracheal allograft. In this IPTT model, recipient BM-derived cells appear to be the primary targets of IL-17RA signaling during fibrotic obliteration of the tracheal allograft.


Assuntos
Medula Óssea , Receptores de Interleucina-17 , Aloenxertos , Animais , Transplante de Medula Óssea , Fibrose , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
3.
Respir Res ; 22(1): 101, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33827576

RESUMO

BACKGROUND: Late onset non-infectious pulmonary complications (LONIPCs) following allogenic hematopoietic stem cell transplantation (allo-HSCT) confer a significant mortality risk. Lung transplantation (LTx) has the potential to provide survival benefit but the impact of prior allo-HSCT on post-LTx outcomes is not well studied. METHODS: This retrospective, single-centre cohort study assessed the post-LTx outcomes of adults with LONIPCs of allo-HSCT. Outcomes of LTx for LONIPCs were compared to propensity-score matched LTx controls (n = 38, non-HSCT) and recipients of re-LTx (n = 70) for chronic lung allograft dysfunction (CLAD). RESULTS: Nineteen patients underwent DLTx for LONIPCs of allo-HSCT between 2003 and 2019. Post-LTx survival was 50% at 5-years. Survival to 1-year post-LTx was similar to matched controls (p = 0.473). Survival, conditional on 1-year survival, was lower in the allo-HSCT cohort (p = 0.034). An increased risk of death due to infection was identified in the allo-HSCT cohort compared to matched controls (p = 0.003). Compared to re-LTx recipients, the allo-HSCT cohort had superior survival to 1-year post-LTx (p = 0.034) but conditional 1-year survival was similar (p = 0.145). CONCLUSION: This study identifies an increased risk of post-LTx mortality in recipients with previous allo-HSCT, associated with infection. It supports the hypothesis that allo-HSCT LTx recipients are relatively more immunosuppressed than patients undergoing LTx for other indications. Optimisation of post-LTx immunosuppressive and antimicrobial strategies to account for this finding should be considered.


Assuntos
Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Pulmão/métodos , Complicações Pós-Operatórias , Disfunção Primária do Enxerto/cirurgia , Pontuação de Propensão , Transplantados , Adolescente , Adulto , Biópsia , Broncoscopia , Doença Crônica , Feminino , Seguimentos , Humanos , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Disfunção Primária do Enxerto/diagnóstico , Disfunção Primária do Enxerto/etiologia , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Transplante Homólogo/efeitos adversos , Adulto Jovem
4.
Eur Respir J ; 54(2)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31164432

RESUMO

Human leukocyte antigen (HLA)-G is a non-classical HLA that inhibits immune responses. Its expression is modified by single nucleotide polymorphisms (SNPs), which are associated with transplant outcomes. Our aim was to investigate the association of donor and recipient HLA-G SNPs with chronic lung allograft dysfunction (CLAD) and mortality after lung transplantation.In this single-centre study, we examined 11 HLA-G SNPs in 345 consecutive recipients and 297 donors of a first bilateral lung transplant. A multivariable Cox proportional hazards model assessed associations of SNPs with death and CLAD. Transbronchial biopsies (TBBx) and bronchoalveolar lavage (BAL) samples were examined using quantitative PCR, ELISA and immunofluorescence.Over a median of 4.75 years, 142 patients (41%) developed CLAD; 170 (49%) died. Multivariable analysis revealed donor SNP +3142 (GG+CG versus CC) was associated with increased mortality (hazard ratio 1.78, 95% CI 1.12-2.84; p=0.015). In contrast, five donor SNPs, -201(CC), -716(TT), -56(CC), G*01:03(AA) and 14 bp INDEL, conferred reduced mortality risk. Specific donor-recipient SNP pairings reduced CLAD risk. Predominantly epithelial HLA-G expression was observed on TBBx without rejection. Soluble HLA-G was present in higher concentrations in the BAL samples of patients who later developed CLAD.Specific donor SNPs were associated with mortality risk after lung transplantation, while certain donor-recipient SNP pairings modulated CLAD risk. TBBx demonstrated predominantly epithelial, and therefore presumably donor-derived, HLA-G expression in keeping with these observations. This study is the first to demonstrate an effect of donor HLA-G SNPs on lung transplantation outcome.


Assuntos
Antígenos HLA-G/genética , Transplante de Pulmão/mortalidade , Polimorfismo de Nucleotídeo Único , Doadores de Tecidos , Adulto , Idoso , Alelos , Biópsia , DNA/genética , Feminino , Genótipo , Sobrevivência de Enxerto , Humanos , Estimativa de Kaplan-Meier , Leucócitos/citologia , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Risco
5.
Transpl Int ; 32(9): 965-973, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31002407

RESUMO

Chronic lung allograft dysfunction (CLAD) remains the leading cause of late death after lung transplantation. Epithelial injury is thought to be a key event in the pathogenesis of CLAD. M30 and M65 are fragments of cytokeratin-18 released specifically during epithelial cell apoptosis and total cell death, respectively. We investigated whether M30 and M65 levels in bronchoalveolar lavage (BAL) correlate with CLAD subtypes: restrictive allograft syndrome (RAS) versus bronchiolitis obliterans syndrome (BOS). BALs were obtained from 26 patients with established CLAD (10 RAS, 16 BOS) and 19 long-term CLAD-free controls. Samples with concurrent infection or acute rejection were excluded. Protein levels were measured by ELISA. Variables were compared using Kruskal-Wallis, Mann-Whitney U test and Chi-squared tests. Association of M30 and M65 levels with post-CLAD survival was assessed using a Cox PH models. M65 levels were significantly higher in RAS compared to BOS and long-term CLAD-free controls and correlated with worse post-CLAD survival. Lung epithelial cell death is enhanced in patients with RAS. Detection of BAL M65 may be used to differentiate CLAD subtypes and as a prognostic marker in patients with established CLAD. Understanding the role of epithelial cell death in CLAD pathogenesis may help identify new therapeutic targets to improve outcome.


Assuntos
Queratina-18/metabolismo , Pneumopatias/metabolismo , Transplante de Pulmão , Fragmentos de Peptídeos/metabolismo , Complicações Pós-Operatórias/metabolismo , Adulto , Biomarcadores/metabolismo , Líquido da Lavagem Broncoalveolar/química , Morte Celular , Células Epiteliais/metabolismo , Feminino , Humanos , Queratina-18/análise , Pneumopatias/mortalidade , Masculino , Pessoa de Meia-Idade , Ontário/epidemiologia , Fragmentos de Peptídeos/análise , Complicações Pós-Operatórias/mortalidade , Estudos Retrospectivos
6.
J Immunol ; 202(8): 2195-2209, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30850479

RESUMO

Regulatory T cell (Treg) therapy is a potential curative approach for a variety of immune-mediated conditions, including autoimmunity and transplantation, in which there is pathological tissue damage. In mice, IL-33R (ST2)-expressing Tregs mediate tissue repair by producing the growth factor amphiregulin, but whether similar tissue-reparative Tregs exist in humans remains unclear. We show that human Tregs in blood and multiple tissue types produced amphiregulin, but this was neither a unique feature of Tregs nor selectively upregulated in tissues. Human Tregs in blood, tonsil, synovial fluid, colon, and lung tissues did not express ST2, so ST2+ Tregs were engineered via lentiviral-mediated overexpression, and their therapeutic potential for cell therapy was examined. Engineered ST2+ Tregs exhibited TCR-independent, IL-33-stimulated amphiregulin expression and a heightened ability to induce M2-like macrophages. The finding that amphiregulin-producing Tregs have a noneffector phenotype and are progressively lost upon TCR-induced proliferation and differentiation suggests that the tissue repair capacity of human Tregs may be an innate function that operates independently from their classical suppressive function.


Assuntos
Proliferação de Células , Imunidade Inata/fisiologia , Linfócitos T Reguladores/imunologia , Adulto , Feminino , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/imunologia , Interleucina-33 , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Especificidade de Órgãos , Linfócitos T Reguladores/citologia
8.
J Thorac Cardiovasc Surg ; 156(3): 1305-1315, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29937159

RESUMO

OBJECTIVE: Our objective was to develop a rapid-onset and durable gene-delivery strategy that is applicable at the time of transplant to determine its effects on both acute rejection and chronic lung allograft fibrosis using a mouse orthotopic lung transplant model. METHODS: C57BL/6 mice received an orthotopic left lung transplant from syngeneic donors or C57BL/10 donors. By using syngeneic lung transplantation, we established a novel gene transfer protocol with lentivirus luciferase intrabronchial administration to the donor lung ex vivo before transplantation. This strategy was applied in allogeneic lung transplantation with lentivirus engineering expression of human interleukin-10 or lentivirus luciferase (control). RESULTS: Bioluminescent imaging revealed that the highest early transgene expression was achieved when lentivirus luciferase was administered both directly into the donor lung graft ex vivo before implantation and subsequently to the recipient in vivo daily on post-transplant days 1 to 4, compared with post-transplant in vivo administration only (days 0 to 4). Our previous work with adenoviral interleukin-10 gene therapy indicates that early interleukin-10 expression in the allograft is desirable. Therefore, we selected the combined protocol for human interleukin-10 encoding lentiviral vector therapy. In the allogeneic transplant setting, ex vivo and in vivo human interleukin-10 encoding lentiviral vector therapy reduced acute rejection grade (2.0 vs 3.0, P < .05) at day 28. The percentage of fibrotic obliterated airways was reduced in the human interleukin-10 encoding lentiviral vector-treated group (10.9% ± 7.7% vs 40.9% ± 9.3%, P < .05). CONCLUSIONS: Delivery of lentiviral interleukin-10 gene therapy, using a novel combined ex vivo and in vivo delivery strategy, significantly improves acute and chronic rejection in the mouse lung transplant model.


Assuntos
Técnicas de Transferência de Genes , Rejeição de Enxerto/prevenção & controle , Interleucina-10/genética , Transplante de Pulmão , Aloenxertos , Animais , Ensaio de Imunoadsorção Enzimática , Interleucina-10/fisiologia , Lentivirus , Transplante de Pulmão/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Respir Res ; 19(1): 102, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29801490

RESUMO

BACKGROUND: Bronchoalveolar lavage (BAL) has proven to be very useful to monitor the lung allograft after transplantation. In addition to allowing detection of infections, multiple BAL analytes have been proposed as potential biomarkers of lung allograft rejection or dysfunction. However, BAL collection is not well standardized and differences in BAL collection represent an important source of variation. We hypothesized that there are systematic differences between sequential BALs that are relevant to BAL analysis. METHODS: As part of 126 consecutive bronchoscopies in lung transplant recipients, two sequential BALs (BAL1 and BAL2) were performed in one location during each bronchoscopy by instilling and suctioning 50 ml of normal saline twice into separate containers. Cell concentration, viability and differentials, Surfactant Protein-D (SP-D), Club Cell Secretory Protein (CCSP), and levels of CXCL10, IL-10, CCL2, CCL5, VEGF-C, RAGE, CXCL9, CXCL1, IL-17A, IL-21, PDGF, and GCSF were compared between BAL1 and BAL2. RESULTS: Total cell concentration did not differ between BAL1 and BAL2; however, compared to BAL2, BAL1 had more dead cells, epithelial cells, neutrophils, and higher concentrations of airway epithelium-derived CCSP and inflammatory markers. BAL2 had a higher concentration of SP-D compared to BAL1. CONCLUSION: In this study performed in lung transplant recipients, we show that sequential BALs represent different lung compartments and have distinct compositions. BAL1 represents the airway compartment with more epithelial cells, neutrophils, and epithelium-derived CCSP. Conversely, BAL2 samples preferentially the distal bronchoalveolar space with greater cell viability and higher SP-D. Our findings illustrate how the method of BAL collection can influence analyte concentrations and further emphasize the need for a standardized approach in translational research involving BAL samples.


Assuntos
Pesquisa Biomédica/tendências , Líquido da Lavagem Broncoalveolar/citologia , Lavagem Broncoalveolar/tendências , Transplante de Pulmão/tendências , Pulmão/patologia , Adulto , Idoso , Broncoscopia/tendências , Estudos de Coortes , Feminino , Humanos , Pulmão/cirurgia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Fatores de Tempo
10.
PLoS One ; 8(6): e65253, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762329

RESUMO

Patients with autoimmune lymphoproliferative syndrome (ALPS) and lymphoproliferation (LPR) mice are deficient in Fas, and accumulate large numbers of αß-TCR(+), CD4(-), CD8(-) double negative (DN) T cells. The function of these DN T cells remains largely unknown. The common γ subunit of the activating Fc receptors, FcRγ, plays an important role in mediating innate immune responses. We have shown previously that a significant proportion of DN T cells express FcRγ, and that this molecule is required for TCR transgenic DN T cells to suppress allogeneic immune responses. Whether FcRγ plays a critical role in LPR DN T cell-mediated suppression of immune responses to auto and allo-antigens is not known. Here, we demonstrated that FcRγ(+), but not FcRγ(-) LPR DN T cells could suppress Fas(+) CD4(+) and CD8(+) T cell proliferation in vitro and attenuated CD4(+) T cell-mediated graft-versus host disease. Although FcRγ expression did not allow LPR DN T cells to inhibit the expansion of Fas-deficient cells within the LPR context, adoptive transfer of FcRγ(+), but not FcRγ(-), DN T cells inhibited lymphoproliferation in generalized lymphoproliferative disease (GLD) mice. Furthermore, FcRγ acted in a cell-intrinsic fashion to limit DN T cell accumulation by increasing the rate of apoptosis in proliferated cells. These results indicate that FcRγ can confer Fas-dependent regulatory properties on LPR DN T cells, and suggest that FcRγ may be a novel marker for functional DN Tregs.


Assuntos
Síndrome Linfoproliferativa Autoimune/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores de IgG/imunologia , Receptor fas/imunologia , Transferência Adotiva , Animais , Síndrome Linfoproliferativa Autoimune/genética , Síndrome Linfoproliferativa Autoimune/patologia , Antígenos CD4/genética , Antígenos CD4/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/transplante , Antígenos CD8/genética , Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Proliferação de Células , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Masculino , Camundongos , Receptores de IgG/genética , Receptor fas/deficiência , Receptor fas/genética
11.
J Autoimmun ; 42: 80-93, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23313147

RESUMO

Deficiency of Fas or its ligand leads to lymphocyte accumulation, lymphadenopathy, splenomegaly, and autoimmunity in mice and humans. Although the Fas pathway is important for limiting the number of peripheral T cells, inactivation of other pro-apoptotic molecules can also perturb T cell homeostasis independently of and/or in concert with Fas deficiency. Here, we show that combined deficiency of Fas and the Fc receptor common γ signaling chain (FcRγ) results in worsened T cell accumulation in comparison with mice lacking Fas alone, with a particularly marked increase in the number of TCRαß(+)CD4(-)CD8(-) double negative (DN) T cells. LPR FcRγ(-/-) mice exhibited reduced survival due to progressive lymphadenopathy. We further investigated the mechanisms whereby FcRγ deficiency promotes lymphoproliferative disease in Fas-mutant mice. Interestingly, there were no significant differences in T cell proliferation between LPR FcRγ(+/+) and LPR FcRγ(-/-) mice in vivo and in vitro. However, FcRγ deletion resulted in significantly decreased peripheral T cell apoptosis. Importantly, the observed increase in apoptosis was restricted to a subset of FcRγ(+) T cells. These T cells, but not those lacking FcRγ expression, exhibited increased activation of caspases 3 and 9, indicating a role for FcRγ in driving their apoptosis. FcRγ(+) DN T cells also showed enhanced sensitivity to TCR restimulation-induced cell death (RICD) despite lacking Fas, suggesting that TCR stimulation of autoreactive T cells in vivo may serve to eliminate FcRγ(+) T cells and thus exert partial control over lymphoproliferative disease. Hence, our data reveal a novel role for FcRγ in promoting peripheral T cell apoptosis in Fas-deficient mice, which may be of significant value in understanding autoimmune lymphoproliferative syndromes.


Assuntos
Síndrome Linfoproliferativa Autoimune/imunologia , Doenças Linfáticas/imunologia , Receptores de IgG/metabolismo , Linfócitos T/imunologia , Receptor fas/metabolismo , Animais , Apoptose/genética , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Processos de Crescimento Celular/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Mutação/genética , Receptores de IgG/genética , Receptor fas/genética
12.
PLoS One ; 7(10): e47732, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23077665

RESUMO

TCRαß(+) CD4(-)CD8(-)NK(-) double negative T cells (DN T cells) can act as regulatory T cells to inhibit allograft rejection and autoimmunity. Their role in graft-versus-host disease and mechanisms of suppression remain elusive. In this study, we demonstrate that DN T cells can inhibit CD4(+) T cell-mediated GVHD in a semi-allogeneic model of bone marrow transplantation. Furthermore, we present evidence of a novel autocrine IFNγ signaling pathway in Fas-deficient C57BL/6.lpr (B6.lpr) DN T cells. B6.lpr DN T cells lacking IFNγ or its receptor were impaired in their ability to suppress syngeneic CD4(+) T cells responding to alloantigen stimulation both in vitro and in vivo. Autocrine IFNγ signaling was required for sustained B6.lpr DN T cell IFNγ secretion in vivo and for upregulation of surface Fas ligand expression during TCR stimulation. Fas ligand (FasL) expression by B6.lpr DN T cells permitted lysis of activated CD4(+) T cells and was required for suppression of GVHD. Collectively, our data indicate that DN T cells can inhibit GVHD and that IFNγ plays a critical autocrine role in controlling the regulatory function of B6.lpr DN T cells.


Assuntos
Comunicação Autócrina/imunologia , Doença Enxerto-Hospedeiro/imunologia , Interferon gama , Linfócitos T , Animais , Transplante de Medula Óssea , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
13.
Transplantation ; 93(12): 1212-20, 2012 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23318304

RESUMO

BACKGROUND: Obliterative bronchiolitis after lung transplantation is associated with intrapulmonary lymphoid neogenesis. The purpose of this study was to examine the role of lymphoid neogenesis, especially its relationship with secondary lymphoid organs (SLOs) in allograft airway rejection. METHODS: A murine intrapulmonary tracheal transplant model and a conventional subcutaneous tracheal transplant model were tested using wild-type control mice and splenectomized lymphotoxin α knockout (LT) mice deficient in SLOs as recipients. RESULTS: In both subcutaneous and intrapulmonary tracheal transplant models using wild-type animals, tracheal isografts remained open without rejection, whereas allografts showed progressive luminal obliteration after transplantation. Lymphoid neogenesis containing alloreactive T cells was observed in the lungs, which received an intrapulmonary tracheal allograft. Despite a lack of SLOs, intrapulmonary allografts in splenectomized LT mice were rejected and obliterated by day 28, but the rejection of subcutaneous allografts was significantly delayed. Extensive lymphoid neogenesis was observed in the lungs of both intrapulmonary and subcutaneous allograft LT recipients. Increased proliferation of CD4 T cells and B220 B cells was observed in the lungs but not in the thymus or bone marrow. CONCLUSIONS: Intrapulmonary lymphoid neogenesis is capable of mounting alloimmune responses without SLOs. Tracheal allograft rejection occurs as efficiently as in wild-type animals when it is placed in the lungs. Tracheal allograft rejection in the subcutaneous tissue occurs in a delayed manner without SLO in association with intrapulmonary lymphoid neogenesis.


Assuntos
Bronquiolite Obliterante/patologia , Rejeição de Enxerto/patologia , Tecido Linfoide/patologia , Traqueia/transplante , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Bronquiolite Obliterante/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Doença Crônica , Modelos Animais de Doenças , Feminino , Rejeição de Enxerto/imunologia , Pulmão/imunologia , Pulmão/patologia , Tecido Linfoide/imunologia , Linfotoxina-alfa/genética , Linfotoxina-alfa/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/imunologia , Baço/patologia , Traqueia/imunologia , Traqueia/patologia , Transplante Homólogo , Transplante Isogênico
14.
Eur J Immunol ; 41(9): 2699-708, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21660936

RESUMO

TCRαß(+) CD3(+) CD4(-) CD8(-) NK1.1(-) double negative (DN) Tregs comprise 1-3% of peripheral T lymphocytes in mice and humans. It has been demonstrated that DN Tregs can suppress allo-, xeno- and auto-immune responses in an Ag-specific fashion. However, the mechanisms by which DN Tregs regulate immune responses remain elusive. Whether DN Tregs can regulate DCs has not been investigated previously. In this study, we demonstrate that DN Tregs express a high level of CTLA4 and are able to down-regulate costimulatory molecules CD80 and CD86 expressed on Ag-expressing mature DCs (mDCs). DN Tregs from CTLA4 KO mice were not able to downregulate CD80 and CD86 expression, indicating that CTLA4 is critical for DN Treg-mediated downregulation of costimulatory molecule expression on Ag-expressing mature DCs. Furthermore, DN Tregs could kill both immature and mature allogeneic DCs, as well as Ag-loaded syngeneic DCs, in an Ag-specific manner in vitro and in vivo, mainly through the Fas-FasL pathway. These data demonstrate, for the first time, that DN Tregs are potent regulators of DCs and may have the potential to be developed as a novel immune suppression treatment.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Antígeno CTLA-4/metabolismo , Células Dendríticas/metabolismo , Linfócitos T Reguladores/metabolismo , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/patologia , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Células Cultivadas , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/patologia , Regulação para Baixo , Humanos , Terapia de Imunossupressão , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
15.
Can Respir J ; 17(3): e55-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20617216

RESUMO

Pulmonary Langerhans' cell histiocytosis (PLCH) is an unusual cystic lung disease that is also characterized by extrapulmonary manifestations. The current review discusses the presenting features and relevant diagnostic testing and treatment options for PLCH in the context of a clinical case. While the focus of the present article is adult PLCH and its pulmonary manifestations, it is important for clinicians to distinguish the adult and pediatric forms of the disease, as well as to be alert for possible extrapulmonary complications. A major theme of the current series of articles on rare lung diseases has been the translation of insights gained from fundamental research to the clinic. Accordingly, the understanding of dendritic cell biology in this disease has led to important advances in the care of patients with PLCH.


Assuntos
Histiocitose de Células de Langerhans/diagnóstico , Adulto , Feminino , Histiocitose de Células de Langerhans/epidemiologia , Histiocitose de Células de Langerhans/etiologia , Histiocitose de Células de Langerhans/terapia , Humanos , Pulmão/patologia , Tomografia Computadorizada por Raios X
16.
Int Immunopharmacol ; 9(5): 534-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19539567

RESUMO

Limited data are available on the mechanisms that constrain the function of regulatory populations of T cells. Prostaglandin E2 (PGE2) is an endogenous membrane phospholipid metabolite that has important immunomodulatory effects on T cell function. Our previous microarray data indicated that E prostanoid receptor 2 (EP2), a receptor for PGE2, is expressed by regulatory alphabetaTCR(+) CD4(-) CD8(-) NK1.1(-) double negative T (DN Treg) cell clones but not by their non-regulatory natural mutants. Hence, the hypothesis that PGE2 may influence DN Treg cell proliferation and/or regulatory function was tested in this study. Our data indicate that PGE2 acts via the EP2 receptor on DN Treg cells to inhibit their proliferation, an effect reproduced by the EP2-specific agonist butaprost and abrogated by the EP2 antagonist AH6809. In contrast, PGE2 did not affect the ability of DN Treg cells to kill syngeneic CD8(+) T cells activated by allogeneic stimulation. Together, these findings suggest a role for PGE2 in limiting the expansion of DN Treg cells.


Assuntos
Dinoprostona/metabolismo , Receptores de Prostaglandina E/metabolismo , Linfócitos T Reguladores/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacologia , Animais , Antígenos CD4 , Antígenos CD8 , Proliferação de Células/efeitos dos fármacos , Células Clonais , Citotoxicidade Imunológica/efeitos dos fármacos , Dinoprostona/imunologia , Separação Imunomagnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Antagonistas de Prostaglandina/farmacologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/imunologia , Receptores de Prostaglandina E Subtipo EP2 , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Xantonas/farmacologia
17.
Am J Respir Cell Mol Biol ; 36(4): 398-408, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17099139

RESUMO

Lymphangioleiomyomatosis (LAM) is a rare progressive cystic lung disease affecting young women. The pivotal observation that LAM occurs both spontaneously and as part of the tuberous sclerosis complex (TSC) led to the hypothesis that these disorders share common genetic and pathogenetic mechanisms. In this review we describe the evolution of our understanding of the molecular and cellular basis of LAM and TSC, beginning with the discovery of the TSC1 and TSC2 genes and the demonstration of their involvement in sporadic (non-TSC) LAM. This was followed by rapid delineation of the signaling pathways in Drosophila melanogaster with confirmation in mice and humans. This knowledge served as the foundation for novel therapeutic approaches that are currently being used in human clinical trials.


Assuntos
Neoplasias Pulmonares/etiologia , Linfangioleiomiomatose/etiologia , Animais , Drosophila melanogaster/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Linfangioleiomiomatose/genética , Linfangioleiomiomatose/terapia , Camundongos , Modelos Biológicos , Miócitos de Músculo Liso/patologia , Transdução de Sinais , Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/fisiologia
18.
Can Respir J ; 13(7): 375-80, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17036091

RESUMO

The present article is the first in a series that will review selected rare lung diseases. The objective of this series is to promote a greater understanding and awareness of these unusual conditions among respirologists. Each article will begin with a case that serves as a focal point for a discussion of the pathophysiology and management of the particular condition. The first article is on lymphangioleiomyomatosis (LAM); subsequent articles will focus on pulmonary alveolar proteinosis, alpha-1-antitrypsin deficiency and primary ciliary dyskinesia. LAM is a rare, progressive and (without intervention) often fatal interstitial lung disease that predominantly affects women of childbearing age. LAM is characterized by progressive interstitial infiltration of the lung by smooth muscle cells, resulting in diffuse cystic changes of the lung parenchyma. The molecular basis of this disorder has been delineated over the past five years and LAM is now known to be a consequence of mutations in the tuberous sclerosis genes. This knowledge, combined with advances in our understanding of the signalling pathways regulated by these genes, has given rise to potential molecular therapies that hold great promise for treating this devastating disease.


Assuntos
Linfangioleiomiomatose/fisiopatologia , Comorbidade , Humanos , Transplante de Pulmão , Linfangioleiomiomatose/diagnóstico , Linfangioleiomiomatose/epidemiologia , Linfangioleiomiomatose/genética , Linfangioleiomiomatose/cirurgia , Pneumotórax/epidemiologia , Pneumotórax/terapia , Prognóstico , Testes de Função Respiratória , Espirometria , Tomografia Computadorizada por Raios X , Esclerose Tuberosa/epidemiologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA