Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 38(2): 360-72, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23376058

RESUMEN

Thymic stromal lymphopoietin (TSLP) is an epithelial cell-derived cytokine important for the initiation and development of T helper (Th2) cell-mediated allergic inflammation. In this study, we identified a positive association between interleukin-9 (IL-9) and TSLP concentration in the serum of infants with atopic dermatitis. In primary cell cultures, the addition of TSLP led to an increase in IL-9 production from human and mouse Th9 cells, and induced an increase in signal transducer and activator of transcription 5 (STAT5) activation and binding to the Il9 promoter. In vivo, use of an adoptive transfer model demonstrated that TSLP promoted IL-9-dependent, Th9 cell-induced allergic inflammation by acting directly on T cells. Moreover, transgenic expression of TSLP in the lung stimulated IL-9 production in vivo, and anti-IL-9 treatment attenuated TSLP-induced airway inflammation. Together, our results demonstrate that TSLP promotes Th9 cell differentiation and function and define a requirement for IL-9 in TSLP-induced allergic inflammation.


Asunto(s)
Citocinas/inmunología , Dermatitis Atópica/inmunología , Inflamación/inmunología , Interleucina-9/inmunología , Factor de Transcripción STAT5/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Anticuerpos Neutralizantes/farmacología , Diferenciación Celular/efectos de los fármacos , Citocinas/genética , Citocinas/farmacología , Dermatitis Atópica/genética , Dermatitis Atópica/patología , Expresión Génica/efectos de los fármacos , Humanos , Lactante , Inflamación/genética , Inflamación/patología , Interleucina-9/antagonistas & inhibidores , Interleucina-9/genética , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/inmunología , Sistema Respiratorio/patología , Factor de Transcripción STAT5/agonistas , Factor de Transcripción STAT5/genética , Transducción de Señal/efectos de los fármacos , Células Th2/efectos de los fármacos , Células Th2/patología , Linfopoyetina del Estroma Tímico
2.
Am J Respir Crit Care Med ; 204(9): 1060-1074, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34346860

RESUMEN

Rationale: Primary graft dysfunction (PGD) is a severe form of acute lung injury, leading to increased early morbidity and mortality after lung transplant. Obesity is a major health problem, and recipient obesity is one of the most significant risk factors for developing PGD. Objectives: We hypothesized that T-regulatory cells (Tregs) are able to dampen early ischemia-reperfusion events and thereby decrease the risk of PGD, whereas that action is impaired in obese recipients. Methods: We evaluated Tregs, T cells, and inflammatory markers, plus clinical data, in 79 lung transplant recipients and 41 liver or kidney transplant recipients and studied two groups of mice on a high-fat diet (HFD), which did ("inflammatory" HFD) or did not ("healthy" HFD) develop low-grade inflammation with decreased Treg function. Measurements and Main Results: We identified increased levels of IL-18 as a previously unrecognized mechanism that impairs Tregs' suppressive function in obese individuals. IL-18 decreases levels of FOXP3, the key Treg transcription factor, decreases FOXP3 di- and oligomerization, and increases the ubiquitination and proteasomal degradation of FOXP3. IL-18-treated Tregs or Tregs from obese mice fail to control PGD, whereas IL-18 inhibition ameliorates lung inflammation. The IL-18-driven impairment in Tregs' suppressive function before transplant was associated with an increased risk and severity of PGD in clinical lung transplant recipients. Conclusions: Obesity-related IL-18 induces Treg dysfunction that may contribute to the pathogenesis of PGD. Evaluation of Tregs' suppressive function together with evaluation of IL-18 levels may serve as a screening tool to identify obese individuals with an increased risk of PGD before transplant.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Interleucina-18/metabolismo , Trasplante de Pulmón/efectos adversos , Obesidad/complicaciones , Disfunción Primaria del Injerto/etiología , Daño por Reperfusión/etiología , Linfocitos T Reguladores/metabolismo , Lesión Pulmonar Aguda/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Obesos , Persona de Mediana Edad , Disfunción Primaria del Injerto/fisiopatología , Daño por Reperfusión/fisiopatología
3.
J Immunol ; 201(2): 772-781, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29884698

RESUMEN

Leukocyte-associated Ig-like receptor 1 (LAIR1) is an ITIM-bearing collagen receptor expressed by leukocytes and is implicated in immune suppression. However, using a divalent soluble LAIR1/Fc recombinant protein to block interaction of cell surface LAIR1 with matrix collagen, we found that whereas Th1 responses were enhanced as predicted, Th17 responses were strongly inhibited. Indeed, LAIR1 on both T cells and monocytes was required for optimal Th17 responses to collagen type (Col)V. For pre-existing "natural" Th17 response to ColV, the LAIR1 requirement was absolute, whereas adaptive Th17 and Th1/17 immune responses in both mice and humans were profoundly reduced in the absence of LAIR1. Furthermore, the addition of C1q, a natural LAIR1 ligand, decreased Th1 responses in a dose-dependent manner, but it had no effect on Th17 responses. In IL-17-dependent murine organ transplant models of chronic rejection, LAIR1+/+ but not LAIR1-/- littermates mounted strong fibroproliferative responses. Surface LAIR1 expression was higher on human Th17 cells as compared with Th1 cells, ruling out a receptor deficiency that could account for the differences. We conclude that LAIR1 ligation by its natural ligands favors Th17 cell development, allowing for preferential activity of these cells in collagen-rich environments. The emergence of cryptic self-antigens such as the LAIR1 ligand ColV during ischemia/reperfusion injury and early acute rejection, as well as the tendency of macrophages/monocytes to accumulate in the allograft during chronic rejection, favors Th17 over Th1 development, posing a risk to long-term graft survival.


Asunto(s)
Rechazo de Injerto/inmunología , Receptores Inmunológicos/metabolismo , Células TH1/fisiología , Células Th17/inmunología , Animales , Autoantígenos/inmunología , Células Cultivadas , Colágeno/metabolismo , Humanos , Inmunidad Celular , Inmunomodulación , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Trasplante de Órganos , Unión Proteica , Receptores Inmunológicos/genética
4.
Am J Transplant ; 19(10): 2705-2718, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31278849

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Comercio/normas , Factores de Transcripción Forkhead/fisiología , Rechazo de Injerto/prevención & control , Enfermedades Pulmonares/inmunología , Trasplante de Pulmón/efectos adversos , Microbiota , Linfocitos T Reguladores/inmunología , Aloinjertos , Animales , Linfocitos T CD4-Positivos/microbiología , Rechazo de Injerto/etiología , Rechazo de Injerto/metabolismo , Supervivencia de Injerto/inmunología , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/microbiología , Receptores de Trasplantes
6.
J Biol Chem ; 291(7): 3359-70, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26721885

RESUMEN

We have shown previously that collagen V (col(V)) autoimmunity is a consistent feature of atherosclerosis in human coronary artery disease and in the Apoe(-/-) mouse model. We have also shown sensitization of Apoe(-/-) mice with col(V) to markedly increase the atherosclerotic burden, providing evidence of a causative role for col(V) autoimmunity in atherosclerotic pathogenesis. Here we sought to determine whether induction of immune tolerance to col(V) might ameliorate atherosclerosis, providing further evidence for a causal role for col(V) autoimmunity in atherogenesis and providing insights into the potential for immunomodulatory therapeutic interventions. Mucosal inoculation successfully induced immune tolerance to col(V) with an accompanying reduction in plaque burden in Ldlr(-/-) mice on a high-cholesterol diet. The results therefore demonstrate that inoculation with col(V) can successfully ameliorate the atherosclerotic burden, suggesting novel approaches for therapeutic interventions. Surprisingly, tolerance and reduced atherosclerotic burden were both dependent on the recently described IL-35 and not on IL-10, the immunosuppressive cytokine usually studied in the context of induced tolerance and amelioration of atherosclerotic symptoms. In addition to the above, using recombinant protein fragments, we were able to localize two epitopes of the α1(V) chain involved in col(V) autoimmunity in atherosclerotic Ldlr(-/-) mice, suggesting future courses of experimentation for the characterization of such epitopes.


Asunto(s)
Aterosclerosis/prevención & control , Autoinmunidad , Colágeno Tipo V/uso terapéutico , Hipersensibilidad Tardía/prevención & control , Tolerancia Inmunológica , Interleucinas/metabolismo , Administración Intranasal , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/metabolismo , Aterosclerosis/etiología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Bovinos , Células Cultivadas , Colágeno Tipo V/administración & dosificación , Colágeno Tipo V/química , Colágeno Tipo V/genética , Dieta Occidental/efectos adversos , Mapeo Epitopo , Humanos , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/metabolismo , Hipersensibilidad Tardía/fisiopatología , Inmunidad Mucosa , Interleucinas/antagonistas & inhibidores , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bazo/inmunología , Bazo/metabolismo , Bazo/patología
7.
Am J Pathol ; 186(9): 2310-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27392970

RESUMEN

Idiopathic pulmonary fibrosis is a devastating disease, with no good diagnostic biomarker and limited treatment options. Previous studies suggest that collagen V overexpression and collagen V-mediated immune response play roles in the pathogenesis of idiopathic pulmonary fibrosis. This study aimed to identify dysregulated miRNA-related collagen V overexpression during idiopathic pulmonary fibrosis. We found that the expression levels of miR-185 and miR-186 were decreased in the lungs of idiopathic pulmonary fibrosis patients. The levels of miR-185 and miR-186 were not correlated with disease severity of idiopathic pulmonary fibrosis. The direct regulation of COL5A1 by miR-185 and miR-186 was confirmed by a luciferase reporter assay. Furthermore, mimics of miR-185 and miR-186 blocked transforming growth factor-ß-induced collagen V overexpression and alleviated transforming growth factor-ß-induced epithelial-mesenchymal transition in A549 cells and HCC827 cells. Our findings suggest that attenuated expression of miR-185 and miR-186 may be responsible for collagen V overexpression during idiopathic pulmonary fibrosis, and these miRNAs may serve as pathogenesis-related biomarkers and treatment targets.


Asunto(s)
Colágeno Tipo V/biosíntesis , Transición Epitelial-Mesenquimal/genética , Fibrosis Pulmonar Idiopática/patología , MicroARNs/metabolismo , Anciano , Western Blotting , Femenino , Regulación de la Expresión Génica/genética , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
FASEB J ; 30(6): 2336-50, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26956419

RESUMEN

Complement activation, an integral arm of innate immunity, may be the critical link to the pathogenesis of idiopathic pulmonary fibrosis (IPF). Whereas we have previously reported elevated anaphylatoxins-complement component 3a (C3a) and complement component 5a (C5a)-in IPF, which interact with TGF-ß and augment epithelial injury in vitro, their role in IPF pathogenesis remains unclear. The objective of the current study is to determine the mechanistic role of the binding of C3a/C5a to their respective receptors (C3aR and C5aR) in the progression of lung fibrosis. In normal primary human fetal lung fibroblasts, C3a and C5a induces mesenchymal activation, matrix synthesis, and the expression of their respective receptors. We investigated the role of C3aR and C5aR in lung fibrosis by using bleomycin-injured mice with fibrotic lungs, elevated local C3a and C5a, and overexpression of their receptors via pharmacologic and RNA interference interventions. Histopathologic examination revealed an arrest in disease progression and attenuated lung collagen deposition (Masson's trichrome, hydroxyproline, collagen type I α 1 chain, and collagen type I α 2 chain). Pharmacologic or RNA interference-specific interventions suppressed complement activation (C3a and C5a) and soluble terminal complement complex formation (C5b-9) locally and active TGF-ß1 systemically. C3aR/C5aR antagonists suppressed local mRNA expressions of tgfb2, tgfbr1/2, ltbp1/2, serpine1, tsp1, bmp1/4, pdgfbb, igf1, but restored the proteoglycan, dcn Clinically, compared with pathologically normal human subjects, patients with IPF presented local induction of C5aR, local and systemic induction of soluble C5b-9, and amplified expression of C3aR/C5aR in lesions. The blockade of C3aR and C5aR arrested the progression of fibrosis by attenuating local complement activation and TGF-ß/bone morphologic protein signaling as well as restoring decorin, which suggests a promising therapeutic strategy for patients with IPF.-Gu, H., Fisher, A. J., Mickler, E. A., Duerson, F., III, Cummings, O. W., Peters-Golden, M., Twigg, H. L., III, Woodruff, T. M., Wilkes, D. S., Vittal, R. Contribution of the anaphylatoxin receptors, C3aR and C5aR, to the pathogenesis of pulmonary fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis Pulmonar/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Complemento/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Línea Celular , Cadena alfa 1 del Colágeno Tipo I , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica/fisiología , Humanos , Lesión Pulmonar/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fibrosis Pulmonar/inducido químicamente , Interferencia de ARN , Receptor de Anafilatoxina C5a/genética , Receptores de Complemento/genética , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
9.
Am J Respir Cell Mol Biol ; 55(6): 889-898, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27494303

RESUMEN

Airway epithelial CD55 down-regulation occurs in several hypoxia-associated pulmonary diseases, but the mechanism is unknown. Using in vivo and in vitro assays of pharmacologic inhibition and gene silencing, the current study investigated the role of hypoxia-inducible factor (HIF)-1α in regulating airway epithelial CD55 expression. Hypoxia down-regulated CD55 expression on small-airway epithelial cells in vitro, and in murine lungs in vivo; the latter was associated with local complement activation. Treatment with pharmacologic inhibition or silencing of HIF-1α during hypoxia-recovered CD55 expression in small-airway epithelial cells. HIF-1α overexpression or blockade, in vitro or in vivo, down-regulated CD55 expression. Collectively, these data show a key role for HIF-1α in regulating the expression of CD55 on airway epithelium.


Asunto(s)
Antígenos CD55/metabolismo , Epitelio/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/metabolismo , Aminoácidos Dicarboxílicos/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL
10.
J Surg Res ; 203(1): 82-90, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27338538

RESUMEN

BACKGROUND: Lung transplantation outcomes are among the least favorable, with most recipients eventually developing bronchiolitis obliterans syndrome (BOS) and subsequent graft failure. The presence of human leukocyte antigen (HLA)-DR has been implicated in the pathogenesis of BOS and may play a role in these poor outcomes. METHODS: Lung transplant donor and recipient data were retrospectively gathered from the United Network for Organ Sharing database from January 2006 to June 2013. Donor and recipient characteristics, proportion of recipients treated for first year rejection, and 5-y rates of survival and freedom from BOS were determined according to HLA-DR1, -DR7, -DR13, and -DR15 status in both donor and recipient. Each HLA-DR allele was stratified by donor-recipient pair positivity status. RESULTS: A total of 7402 lung transplant recipients met the inclusion and exclusion criteria. There were significant but small differences in donor and recipient characteristics for each HLA-DR group. The recipients in the D(-)R(+) pairing for HLA-DR13 and those in the D(+)R(-) pairing for HLA-DR15 had significantly higher rates of receiving treatment for rejection within the first year after transplant (P = 0.024 and P = 0.001, respectively). There were no differences in 5-y survival or freedom from BOS for any of the four HLA-DR alleles studied. CONCLUSIONS: There are higher rates of patients treated for rejection within the first year who are either negative for the HLA-DR15 allele but received a donor-positive lung or positive for the HLA-DR13 allele but received a donor-negative lung for that allele. However, these differences do not appear to affect long-term outcomes.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Rechazo de Injerto/inmunología , Subtipos Serológicos HLA-DR/metabolismo , Trasplante de Pulmón , Complicaciones Posoperatorias/inmunología , Adulto , Anciano , Biomarcadores/metabolismo , Bronquiolitis Obliterante/etiología , Femenino , Humanos , Modelos Logísticos , Trasplante de Pulmón/mortalidad , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Estudios Retrospectivos , Análisis de Supervivencia
11.
Semin Immunol ; 24(2): 131-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21925897

RESUMEN

Lung transplantation is considered a definitive treatment for many lung diseases. However, rejection and other pathologic entities are major causes of morbidity and mortality for lung transplant recipients. Primary graft dysfunction (PGD) and obliterative bronchiolitis (OB) are the leading causes of early and late mortality, respectively. While the immune basis of PGD has not been clearly defined, evidence is emerging about roles for autoantibodies in this process. Similarly, the pathogenesis of OB has been linked recently to autoimmunity. This review will highlight the current understanding of autoantibodies in PGD and OB post lung transplantation.


Asunto(s)
Autoanticuerpos/biosíntesis , Rechazo de Injerto/inmunología , Trasplante de Pulmón/inmunología , Disfunción Primaria del Injerto/inmunología , Animales , Autoanticuerpos/inmunología , Bronquiolitis Obliterante/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Ratones , Ratas
12.
Am J Transplant ; 15(7): 1793-1804, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25773063

RESUMEN

Lung transplant survival is limited by obliterative bronchiolitis (OB), but the mechanisms of OB development are unknown. Previous studies in a mouse model of orthotopic lung transplantation suggested a requirement for IL-17. We have used this orthotopic mouse model to investigate the source of IL-17A and the requirement for T cells producing IL-17A. The major sources of IL-17A were CD4(+) T cells and γδ T cells. Depletion of CD4(+) T cells led to a significantly decreased frequency and number of IL-17A(+) lymphocytes and was sufficient to prevent acute rejection and OB. However, mice with STAT3-deficient T cells, which are unable to differentiate into Th17 cells, rejected lung allografts and developed OB similar to control mice. The frequency of IL-17A(+) cells was not decreased in mice with STAT3-deficient T cells due mainly to the presence of IL-17A(+) γδ T cells. Deficiency of γδ T cells also did not affect the development of airway fibrosis. Our data suggest that CD4(+) T cells are required for OB development and expansion of IL-17A responses in the lung, while Th17 and γδ T cells are not absolutely required and may compensate for each other.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Linfocitos T CD4-Positivos/inmunología , Supervivencia de Injerto/inmunología , Interleucina-17/inmunología , Trasplante de Pulmón , Células Th17/inmunología , Animales , Bronquiolitis Obliterante/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Interferón gamma/metabolismo , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT3/fisiología
13.
Am J Physiol Heart Circ Physiol ; 309(5): H719-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26116712

RESUMEN

The abdominal aortic aneurysm (AAA) is a disease process that carries significant morbidity and mortality in the absence of early identification and treatment. While current management includes surveillance and surgical treatment of low- and high-risk aneurysms, respectively, our narrow understanding of the pathophysiology of AAAs limits our ability to more effectively manage and perhaps even prevent the occurrence of this highly morbid disease. Over the past couple of decades, there has been considerable interest in exploring the role of autoimmunity as an etiological component of AAA. This review covers the current literature pertaining to this immunological process, focusing on research that highlights the local and systemic immune components found in both human patients and murine models. A better understanding of the autoimmune mechanisms in the pathogenesis of AAAs can pave the way to novel and improved treatment strategies in this patient population.


Asunto(s)
Aneurisma de la Aorta Abdominal/inmunología , Autoinmunidad , Animales , Aneurisma de la Aorta Abdominal/etiología , Humanos
14.
Eur Respir J ; 45(5): 1393-402, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25614165

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with poor prognosis. IPF appears to be heterogeneous in pathobiology with ∼40% of IPF patients found to have elevated levels of circulating antibodies to the autoantigen type V collagen (col(V)). Following a targeted, precision medicine approach, we conducted a phase 1 study to test the safety and explore potential efficacy of IW001, a col(V) oral immunotherapeutic developed to treat antibody-positive IPF patients. We divided 30 antibody-positive IPF patients into three cohorts for daily dosing over a 24-week period. All patients completed treatment without serious adverse events, acute exacerbations or IPF-related hospitalisations. A decline in lung function occurred in the lowest-dose cohort that was comparable to that reported in placebo arms of published IPF trials. In contrast, the highest-dose cohort showed a trend toward stabilisation of forced vital capacity and matrix metalloproteinase 7, and a reduction in binding of C1q to anti-col(V) antibodies. IW001 may modulate the immune response to col(V) and may represent a new therapeutic for col(V)- reactive IPF patients.


Asunto(s)
Colágeno Tipo V/uso terapéutico , Fibrosis Pulmonar Idiopática/inmunología , Fibrosis Pulmonar Idiopática/terapia , Inmunoterapia/métodos , Administración Oral , Anciano , Biomarcadores/sangre , Estudios de Cohortes , Complemento C1q/metabolismo , Femenino , Humanos , Pulmón/inmunología , Masculino , Metaloproteinasa 7 de la Matriz/sangre , Persona de Mediana Edad , Seguridad del Paciente , Pruebas de Función Respiratoria , Albúmina Sérica/metabolismo , Capacidad Vital/efectos de los fármacos
15.
FASEB J ; 28(10): 4223-34, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24958208

RESUMEN

The epithelial complement inhibitory proteins (CIPs) cluster of differentiation 46 and 55 (CD46 and CD55) regulate circulating immune complex-mediated complement activation in idiopathic pulmonary fibrosis (IPF). Our previous studies demonstrated that IL-17A mediates epithelial injury via transforming growth factor ß1 (TGF-ß1) and down-regulates CIPs. In the current study, we examined the mechanistic role of TGF-ß1 in complement activation-mediated airway epithelial injury in IPF pathogenesis. We observed lower epithelial CIP expression in IPF lungs compared to normal lungs, associated with elevated levels of complement component 3a and 5a (C3a and C5a), locally and systemically. In normal primary human small airway epithelial cells (SAECs) treated with TGF-ß1 (10 ng/ml), C3a, or C5a (100 nM), we observed loss of CIPs and increased poly(ADP-ribose) polymerase (PARP) activation [also observed with RNA interference (RNAi) of CD46/CD55]. TGF-ß1-mediated loss of CIPs and Snail induction [SNAI1; a transcriptional repressor of E-cadherin (E-CAD)] was blocked by inhibiting mitogen-activated protein kinase (p38MAPK; SB203580) and RNAi silencing of SNAI1. C3a- and C5a-mediated loss of CIPs was also blocked by p38MAPK inhibition. While C3a upregulated TGFb transcripts, both C3a and C5a down-regulated SMAD7 (negative regulator of TGF-ß), and whereas TGF-ß1 induced C3a/C5a receptor (C3aR/C5aR) expression, pharmacologic C3aR/C5aR inhibition protected against C3a-/C5a-mediated loss of CIPs. Taken together, our results suggest that epithelial injury in IPF can be collectively amplified as a result of TGF-ß1-induced loss of CIPs leading to complement activation that down-regulates CIPs and induces TGF-ß1 expression


Asunto(s)
Activación de Complemento , Fibrosis Pulmonar Idiopática/metabolismo , Mucosa Respiratoria/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Adulto , Anciano , Antígenos CD55/genética , Antígenos CD55/metabolismo , Células Cultivadas , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inmunología , Fibrosis Pulmonar Idiopática/patología , Masculino , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Persona de Mediana Edad , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Proteína smad7/genética , Proteína smad7/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Immunol ; 190(11): 5446-53, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23630356

RESUMEN

Sarcoidosis pathogenesis is characterized by peripheral anergy and an exaggerated, pulmonary CD4(+) Th1 response. In this study, we demonstrate that CD4(+) anergic responses to polyclonal TCR stimulation are present peripherally and within the lungs of sarcoid patients. Consistent with prior observations, spontaneous release of IL-2 was noted in sarcoidosis bronchoalveolar lavage CD4(+) T cells. However, in contrast to spontaneous hyperactive responses reported previously, the cells displayed anergic responses to polyclonal TCR stimulation. The anergic responses correlated with diminished expression of the Src kinase Lck, protein kinase C-θ, and NF-κB, key mediators of IL-2 transcription. Although T regulatory (Treg) cells were increased in sarcoid patients, Treg depletion from the CD4(+) T cell population of sarcoidosis patients did not rescue IL-2 and IFN-γ production, whereas restoration of the IL-2 signaling cascade, via protein kinase C-θ overexpression, did. Furthermore, sarcoidosis Treg cells displayed poor suppressive capacity indicating that T cell dysfunction was a global CD4(+) manifestation. Analyses of patients with spontaneous clinical resolution revealed that restoration of CD4(+) Th1 and Treg cell function was associated with resolution. Conversely, disease progression exhibited decreased Th1 cytokine secretion and proliferative capacity, and reduced Lck expression. These findings implicate normalized CD4(+) T cell function as a potential therapeutic target for sarcoidosis resolution.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Remisión Espontánea , Sarcoidosis Pulmonar/inmunología , Subgrupos de Linfocitos T/inmunología , Adulto , Anciano , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Linfocitos T CD4-Positivos/metabolismo , Anergia Clonal/inmunología , Femenino , Humanos , Interleucina-2/metabolismo , Masculino , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Adulto Joven
17.
J Immunol ; 191(8): 4431-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043901

RESUMEN

Obliterative bronchiolitis (OB) post-lung transplantation involves IL-17-regulated autoimmunity to type V collagen and alloimmunity, which could be enhanced by complement activation. However, the specific role of complement activation in lung allograft pathology, IL-17 production, and OB is unknown. The current study examines the role of complement activation in OB. Complement-regulatory protein (CRP) (CD55, CD46, complement receptor 1-related protein y/CD46) expression was downregulated in human and murine OB; and C3a, a marker of complement activation, was upregulated locally. IL-17 differentially suppressed complement receptor 1-related protein y expression in airway epithelial cells in vitro. Neutralizing IL-17 recovered CRP expression in murine lung allografts and decreased local C3a production. Exogenous C3a enhanced IL-17 production from alloantigen- or autoantigen (type V collagen)-reactive lymphocytes. Systemically neutralizing C5 abrogated the development of OB, reduced acute rejection severity, lowered systemic and local levels of C3a and C5a, recovered CRP expression, and diminished systemic IL-17 and IL-6 levels. These data indicated that OB induction is in part complement dependent due to IL-17-mediated downregulation of CRPs on airway epithelium. C3a and IL-17 are part of a feed-forward loop that may enhance CRP downregulation, suggesting that complement blockade could be a therapeutic strategy for OB.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Activación de Complemento , Rechazo de Injerto/inmunología , Interleucina-17/metabolismo , Trasplante de Pulmón/efectos adversos , Animales , Autoinmunidad , Líquido del Lavado Bronquioalveolar , Antígenos CD55/biosíntesis , Colágeno Tipo V/inmunología , Complemento C3a/biosíntesis , Complemento C5 , Regulación hacia Abajo , Humanos , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Interleucina-6/biosíntesis , Prueba de Cultivo Mixto de Linfocitos , Proteína Cofactora de Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptores de Complemento/biosíntesis , Receptores de Complemento 3b
18.
Am J Respir Crit Care Med ; 190(5): 560-71, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25073001

RESUMEN

RATIONALE: Effective therapeutic interventions for chronic, idiopathic lung diseases remain elusive. Normalized T-cell function is an important contributor to spontaneous resolution of pulmonary sarcoidosis. Up-regulation of inhibitor receptors, such as programmed death-1 (PD-1) and its ligand, PD-L1, are important inhibitors of T-cell function. OBJECTIVES: To determine the effects of PD-1 pathway blockade on sarcoidosis CD4(+) T-cell proliferative capacity. METHODS: Gene expression profiles of sarcoidosis and healthy control peripheral blood mononuclear cells were analyzed at baseline and follow-up. Flow cytometry was used to measure ex vivo expression of PD-1 and PD-L1 on systemic and bronchoalveolar lavage-derived cells of subjects with sarcoidosis and control subjects, as well as the effects of PD-1 pathway blockade on cellular proliferation after T-cell receptor stimulation. Immunohistochemistry analysis for PD-1/PD-L1 expression was conducted on sarcoidosis, malignant, and healthy control lung specimens. MEASUREMENTS AND MAIN RESULTS: Microarray analysis demonstrates longitudinal increase in PDCD1 gene expression in sarcoidosis peripheral blood mononuclear cells. Immunohistochemistry analysis revealed increased PD-L1 expression within sarcoidosis granulomas and lung malignancy, but this was absent in healthy lungs. Increased numbers of sarcoidosis PD-1(+) CD4(+) T cells are present systemically, compared with healthy control subjects (P < 0.0001). Lymphocytes with reduced proliferative capacity exhibited increased proliferation with PD-1 pathway blockade. Longitudinal analysis of subjects with sarcoidosis revealed reduced PD-1(+) CD4(+) T cells with spontaneous clinical resolution but not with disease progression. CONCLUSIONS: Analogous to the effects in other chronic lung diseases, these findings demonstrate that the PD-1 pathway is an important contributor to sarcoidosis CD4(+) T-cell proliferative capacity and clinical outcome. Blockade of the PD-1 pathway may be a viable therapeutic target to optimize clinical outcomes.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD4-Positivos/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Sarcoidosis Pulmonar/inmunología , Adulto , Anciano , Anticuerpos , Antígeno B7-H1/inmunología , Biomarcadores/metabolismo , Estudios de Casos y Controles , Proliferación Celular , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Masculino , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/inmunología , Remisión Espontánea , Sarcoidosis Pulmonar/metabolismo , Regulación hacia Arriba
19.
Am J Respir Cell Mol Biol ; 51(4): 467-73, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24901241

RESUMEN

In addition to its established contribution to innate immunity, recent studies have suggested novel roles for the complement system in the development of various lung diseases. Several studies have demonstrated that complement may serve as a key link between innate and adaptive immunity in a variety of pulmonary conditions. However, the specific contributions of complement to lung diseases based on innate and adaptive immunity are just beginning to emerge. Elucidating the role of complement-mediated immune regulation in these diseases will help to identify new targets for therapeutic interventions.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Enfermedades Pulmonares/inmunología , Pulmón/inmunología , Inmunidad Adaptativa , Animales , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Humanos , Inmunidad Innata , Pulmón/metabolismo , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/terapia , Transducción de Señal
20.
Am J Respir Crit Care Med ; 187(5): 527-34, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23306540

RESUMEN

RATIONALE: Primary graft dysfunction (PGD) is the main cause of early morbidity and mortality after lung transplantation. Previous studies have yielded conflicting results for PGD risk factors. OBJECTIVES: We sought to identify donor, recipient, and perioperative risk factors for PGD. METHODS: We performed a 10-center prospective cohort study enrolled between March 2002 and December 2010 (the Lung Transplant Outcomes Group). The primary outcome was International Society for Heart and Lung Transplantation grade 3 PGD at 48 or 72 hours post-transplant. The association of potential risk factors with PGD was analyzed using multivariable conditional logistic regression. MEASUREMENTS AND MAIN RESULTS: A total of 1,255 patients from 10 centers were enrolled; 211 subjects (16.8%) developed grade 3 PGD. In multivariable models, independent risk factors for PGD were any history of donor smoking (odds ratio [OR], 1.8; 95% confidence interval [CI], 1.2-2.6; P = 0.002); FiO2 during allograft reperfusion (OR, 1.1 per 10% increase in FiO2; 95% CI, 1.0-1.2; P = 0.01); single lung transplant (OR, 2; 95% CI, 1.2-3.3; P = 0.008); use of cardiopulmonary bypass (OR, 3.4; 95% CI, 2.2-5.3; P < 0.001); overweight (OR, 1.8; 95% CI, 1.2-2.7; P = 0.01) and obese (OR, 2.3; 95% CI, 1.3-3.9; P = 0.004) recipient body mass index; preoperative sarcoidosis (OR, 2.5; 95% CI, 1.1-5.6; P = 0.03) or pulmonary arterial hypertension (OR, 3.5; 95% CI, 1.6-7.7; P = 0.002); and mean pulmonary artery pressure (OR, 1.3 per 10 mm Hg increase; 95% CI, 1.1-1.5; P < 0.001). PGD was significantly associated with 90-day (relative risk, 4.8; absolute risk increase, 18%; P < 0.001) and 1-year (relative risk, 3; absolute risk increase, 23%; P < 0.001) mortality. CONCLUSIONS: We identified grade 3 PGD risk factors, several of which are potentially modifiable and should be prioritized for future research aimed at preventative strategies. Clinical trial registered with www.clinicaltrials.gov (NCT 00552357).


Asunto(s)
Trasplante de Pulmón/efectos adversos , Disfunción Primaria del Injerto/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Modelos Logísticos , Trasplante de Pulmón/mortalidad , Masculino , Persona de Mediana Edad , Análisis Multivariante , Disfunción Primaria del Injerto/mortalidad , Estudios Prospectivos , Factores de Riesgo , Estados Unidos/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA