Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Autoimmun ; 145: 103197, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38447248

RESUMEN

BACKGROUND AND OBJECTIVE: Understanding the regulation of efferocytosis by myeloid phagocytes is important in identifying novel targets in systemic lupus erythematosus (SLE). Cadherin-11 (CDH11), a cell adhesion molecule, is implicated in inflammatory arthritis and fibrosis and recently been shown to regulate macrophage phagocytosis. The extent and mechanism of this regulation is unknown. Our objective was to examine the extent to which CDH11 regulates myeloid phagocytes and contributes to autoimmunity and tissue inflammation. METHODS: We analyzed efferocytosis in macrophages and dendritic cells (DCs) from WT and Cdh11-/- mice and investigated the mechanisms in vitro. We investigated the role of CDH11 in disease development in vivo using the pristane induced lupus model. To translate the clinical relevance of CDH11 in human disease, we measured serum CDH11 levels in two independent pediatric SLE (pSLE) cohorts and healthy controls. RESULTS: Using bone marrow derived macrophages (BMDMs) and DCs (BMDCs), we found impaired efferocytosis in phagocytes from Cdh11-/- mice, mediated by downregulated efferocytosis receptor expression and RhoGTPase activation. Specifically, loss of CDH11 downregulated Mertk expression and Rac1 activation in BMDMs, and integrin αVß3 expression and Cdc42 activation in BMDCs, highlighting distinct pathways. In vivo, Cdh11-/- mice displayed defective efferocytosis and increased accumulation of apoptotic debris in pristane-induced lupus. Further, Cdh11-/- mice had enhanced systemic inflammation and autoimmune inflammation with increased anti-dsDNA autoantibodies, splenomegaly, type I interferons, and inflammatory cytokines. Paradoxically, at the tissue level, Cdh11-/- mice were protected against glomerulonephritis, indicating a dual role in murine lupus. Finally, SLE patients had increased serum CDH11 compared to controls. CONCLUSION: This study highlights a novel role of CDH11 in regulating myeloid cells and efferocytosis and its potential as a contributor to development in autoimmunity murine lupus. Despite the increase in autoimmunity, Cdh11-/- mice developed decreased tissue inflammation and damage.


Asunto(s)
Cadherinas , Células Dendríticas , Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico , Macrófagos , Ratones Noqueados , Fagocitosis , Animales , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/genética , Ratones , Cadherinas/metabolismo , Cadherinas/genética , Fagocitosis/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Humanos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inflamación/inmunología , Autoinmunidad , Femenino , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo , Fagocitos/inmunología , Fagocitos/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Niño , Terpenos
2.
J Invest Dermatol ; 143(9): 1689-1699, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36967086

RESUMEN

Unbiased informatics approaches have the potential to generate insights into uncharacterized signaling pathways in human disease. In this study, we generated longitudinal transcriptomic profiles of plaque psoriasis lesions from patients enrolled in a clinical trial of the anti-IL17A antibody ixekizumab (IXE). This dataset was then computed against a curated matrix of over 700 million data points derived from published psoriasis and signaling node perturbation transcriptomic and chromatin immunoprecipitation-sequencing datasets. We observed substantive enrichment within both psoriasis-induced and IXE-repressed gene sets of transcriptional targets of members of the MuvB complex, a master regulator of the mitotic cell cycle. These gene sets were similarly enriched for pathways involved in the regulation of the G2/M transition of the cell cycle. Moreover, transcriptional targets for MuvB nodes were strongly enriched within IXE-repressed genes whose expression levels correlated strongly with the extent and severity of the psoriatic disease. In models of human keratinocyte proliferation, genes encoding MuvB nodes were transcriptionally repressed by IXE, and depletion of MuvB nodes reduced cell proliferation. Finally, we made the expression and regulatory networks that supported this study available as a freely accessible, cloud-based hypothesis generation platform. Our study positions inhibition of MuvB signaling as an important determinant of the therapeutic impact of IXE in psoriasis.


Asunto(s)
Fármacos Dermatológicos , Psoriasis , Humanos , Fármacos Dermatológicos/farmacología , Fármacos Dermatológicos/uso terapéutico , Método Doble Ciego , Psoriasis/tratamiento farmacológico , Psoriasis/genética , Psoriasis/patología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Resultado del Tratamiento
3.
Front Immunol ; 13: 795337, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35211116

RESUMEN

Cadherin-11 (CDH11) is a cell-cell adhesion protein that has previously been reported to play an important role in the pathogenesis of pulmonary fibrosis. It is expressed on macrophages in the fibrotic lung. However, the role of CDH11 on macrophage biology has not yet been studied. We show using immunophenotypic analyses that Cdh11-/- mice have fewer recruited monocyte-derived macrophages and Ly6Chi monocytes in the lungs compared to wild-type mice in the intraperitoneal bleomycin-induced pulmonary fibrosis model. Additionally, fewer Ly6Chi monocytes were detected in the bone marrow and peripheral blood of naive Cdh11-/- mice. Given that macrophages are derived from monocytes, we investigated the precursors of the monocyte/macrophage lineage in the bone marrow. We found increased numbers of CMPs and reduced numbers of GMPs and MPs/cMoPs in Cdh11-/- mice compared to wild-type mice, suggesting decreased differentiation towards the myeloid lineage in Cdh11-/- mice. Furthermore, we show using bone marrow cells that loss of CDH11 impaired monocyte to macrophage differentiation. We also demonstrate that CDH11 deficiency repressed the M2 program and impaired the phagocytic function of bone marrow-derived macrophages. Overall, our findings demonstrate a role for CDH11 in macrophage development, M2 polarization, and phagocytic function.


Asunto(s)
Cadherinas/deficiencia , Macrófagos/metabolismo , Monocitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Antígenos Ly/metabolismo , Bleomicina/toxicidad , Cadherinas/genética , Adhesión Celular , Diferenciación Celular , Modelos Animales de Enfermedad , Macrófagos/fisiología , Masculino , Ratones , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología
4.
PLoS One ; 14(7): e0218971, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31269038

RESUMEN

BACKGROUND AND AIMS: Liver fibrosis is characterized by the excessive deposition of extracellular matrix (ECM) leading to impaired function and cirrhosis. Previous reports support a role for cadherin-11 (CDH11) in regulating the development of dermal and pulmonary fibrosis. In the current report, the extent to which CDH11 modulates the development of liver fibrosis induced by carbon tetrachloride (CCL4) was assessed. METHODS: Wild type (WT) and CDH11 deficient (CDH11-/-) mice were treated with CCl4 or vehicle control for 8 weeks to induce liver fibrosis. Liver fibrosis was assessed by histology, collagen content, and RTPCR of fibrotic mediators. RESULTS: Livers from WT mice treated with CCl4 had increased levels of CDH11 which localized to injured hepatocytes, hepatic stellate cells, and macrophages. Interestingly, CDH11-/- mice had decreased histological evidence of liver fibrosis, collagen deposition, α-smooth muscle actin (α-SMA) accumulation, and mRNA levels of fibrotic mediators such as Col1-α1, Snail, TGF-ß and IL-6. CONCLUSIONS: These data demonstrate that CDH11 is increased during liver fibrosis, is an important regulator of liver fibrosis induced by CCL4 and suggest that CDH11 may be a potential therapeutic target for liver fibrosis.


Asunto(s)
Cadherinas/genética , Matriz Extracelular/efectos de los fármacos , Cirrosis Hepática/genética , Hígado/efectos de los fármacos , Animales , Tetracloruro de Carbono/toxicidad , Células Cultivadas , Colágeno/genética , Matriz Extracelular/genética , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Factor de Crecimiento Transformador beta/genética
5.
Dis Model Mech ; 12(5)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31036697

RESUMEN

Combined pulmonary fibrosis and emphysema (CPFE) is a syndrome that predominantly affects male smokers or ex-smokers and it has a mortality rate of 55% and a median survival of 5 years. Pulmonary hypertension (PH) is a frequently fatal complication of CPFE. Despite this dismal prognosis, no curative therapies exist for patients with CPFE outside of lung transplantation and no therapies are recommended to treat PH. This highlights the need to develop novel treatment approaches for CPFE. Studies from our group have demonstrated that both adenosine and its receptor ADORA2B are elevated in chronic lung diseases. Activation of ADORA2B leads to elevated levels of hyaluronan synthases (HAS) and increased hyaluronan, a glycosaminoglycan that contributes to chronic lung injury. We hypothesize that ADORA2B and hyaluronan contribute to CPFE. Using isolated CPFE lung tissue, we characterized expression levels of ADORA2B and HAS. Next, using a unique mouse model of experimental lung injury that replicates features of CPFE, namely airspace enlargement, PH and fibrotic deposition, we investigated whether 4MU, a HAS inhibitor, was able to inhibit features of CPFE. Increased protein levels of ADORA2B and HAS3 were detected in CPFE and in our experimental model of CPFE. Treatment with 4MU was able to attenuate PH and fibrosis but not airspace enlargement. This was accompanied by a reduction of HAS3-positive macrophages. We have generated pre-clinical data demonstrating the capacity of 4MU, an FDA-approved drug, to attenuate features of CPFE in an experimental model of chronic lung injury.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Adenosina/efectos adversos , Ácido Hialurónico/efectos adversos , Fibrosis Pulmonar Idiopática/complicaciones , Fibrosis Pulmonar Idiopática/patología , Enfisema Pulmonar/complicaciones , Enfisema Pulmonar/patología , Agonistas del Receptor de Adenosina A2/farmacología , Adenosina Desaminasa/metabolismo , Animales , Línea Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Humanos , Hialuronano Sintasas/metabolismo , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Macrófagos/metabolismo , Ratones , Receptor de Adenosina A2B/metabolismo
6.
Rheumatology (Oxford) ; 57(10): 1838-1850, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029263

RESUMEN

Objective: SSc is an autoimmune disease characterized by progressive fibrosis of the skin and internal organs. IL-6 and related cytokines that signal through STAT3 have been implicated in the pathogenesis of SSc and mouse models of fibrosis. The aim of this study was to investigate the efficacy of inhibiting STAT3 in the development of fibrosis in two mouse models of skin fibrosis. Methods: Biopsy samples of skin from SSc patients and healthy control subjects were used to determine the expression pattern of phosphotyrosyl (pY705)-STAT3. C188-9, a small molecule inhibitor of STAT3, was used to treat fibrosis in the bleomycin-induced fibrosis model and Tsk-1 mice. In vitro studies were performed to determine the extent to which STAT3 regulates the fibrotic phenotype of dermal fibroblasts. Results: Increased STAT3 and pY705-STAT3 was observed in SSc skin biopsies and in both mouse models of SSc. STAT3 inhibition with C188-9 resulted in attenuated skin fibrosis, myofibroblast accumulation, pro-fibrotic gene expression and collagen deposition in both mouse models of skin fibrosis. C188-9 decreased in vitro dermal fibroblast production of fibrotic genes induced by IL-6 trans-signalling and TGF-ß. Finally, TGF-ß induced phosphotyrosylation of STAT3 in a SMAD3-dependent manner. Conclusion: STAT3 inhibition decreases dermal fibrosis in two models of SSc. STAT3 regulates dermal fibroblasts function in vitro and can be activated by TGF-ß. These data suggest that STAT3 is a potential therapeutic target for dermal fibrosis in diseases such as SSc.


Asunto(s)
Naftoles/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Enfermedades de la Piel/tratamiento farmacológico , Piel/patología , Sulfonamidas/farmacología , Factor de Crecimiento Transformador beta/fisiología , Animales , Femenino , Fibrosis , Humanos , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/fisiología , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/patología , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología
7.
PLoS One ; 12(11): e0187109, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29112946

RESUMEN

OBJECTIVE: Systemic sclerosis (SSc) is an autoimmune disease clinically manifesting as progressive fibrosis of the skin and internal organs. Cadherin-11 (CDH11) expression is increased in fibrotic skin and lung tissue. Targeting CDH11 may be an effective approach to treating fibrosis. We hypothesize that targeting CDH11 will decrease fibrosis in the tight skin-1 (Tsk-1) mouse model. METHODS: CDH11 expression was determined in the Tsk-1 mouse model using quantitative real time PCR and immunofluorescence (IF). Inhibitory anti- CDH11 monoclonal antibodies were tested in Tsk-1 mice for their ability to decrease hypodermal fibrosis. RESULTS: Expression of CDH11 was increased in fibrotic skin from Tsk-1 mice compared to pallid controls. IF staining demonstrated that CDH11 expression localized to fibroblasts within the hypodermis of fibrotic skin. Treatment with inhibitory anti-CDH11 monoclonal antibodies decreased hypodermal thickness and fibrotic mediators in Tsk-1 mice compared to control antibodies. CONCLUSIONS: These data demonstrate an important role for CDH11 in the development of skin fibrosis in Tsk-1 mice. These data add to the growing evidence for the important role of CDH11 in tissue fibrosis and fibrotic disease such as systemic sclerosis.


Asunto(s)
Cadherinas/metabolismo , Modelos Animales de Enfermedad , Esclerodermia Sistémica/patología , Enfermedades de la Piel/patología , Animales , Anticuerpos Monoclonales/inmunología , Biopsia , Cadherinas/inmunología , Femenino , Fibrosis , Humanos , Ratones , Esclerodermia Sistémica/metabolismo , Enfermedades de la Piel/metabolismo
8.
FASEB J ; 30(1): 129-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26324850

RESUMEN

Lung fibrosis is the hallmark of the interstitial lung diseases. Alveolar epithelial cell (AEC) injury is a key step that contributes to a profibrotic microenvironment. Fibroblasts and myofibroblasts subsequently accumulate and deposit excessive extracellular matrix. In addition to TGF-ß, the IL-6 family of cytokines, which signal through STAT-3, may also contribute to lung fibrosis. In the current manuscript, the extent to which STAT-3 inhibition decreases lung fibrosis is investigated. Phosphorylated STAT-3 was elevated in lung biopsies from patients with idiopathic pulmonary fibrosis and bleomycin (BLM)-induced fibrotic murine lungs. C-188-9, a small molecule STAT-3 inhibitor, decreased pulmonary fibrosis in the intraperitoneal BLM model as assessed by arterial oxygen saturation (control, 84.4 ± 1.3%; C-188-9, 94.4 ± 0.8%), histology (Ashcroft score: untreated, 5.4 ± 0.25; C-188-9, 3.3 ± 0.14), and attenuated fibrotic markers such as diminished α-smooth muscle actin, reduced collagen deposition. In addition, C-188-9 decreased the expression of epithelial injury markers, including hypoxia-inducible factor-1α (HIF-1α) and plasminogen activator inhibitor-1 (PAI-1). In vitro studies show that inhibition of STAT-3 decreased IL-6- and TGF-ß-induced expression of multiple genes, including HIF-1α and PAI-1, in AECs. Furthermore, C-188-9 decreased fibroblast-to-myofibroblast differentiation. Finally, TGF-ß stimulation of lung fibroblasts resulted in SMAD2/SMAD3-dependent phosphorylation of STAT-3. These findings demonstrate that STAT-3 contributes to the development of lung fibrosis and suggest that STAT-3 may be a therapeutic target in pulmonary fibrosis.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Bleomicina/farmacología , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis Pulmonar Idiopática/genética , Masculino , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/genética
9.
J Immunol ; 193(7): 3755-68, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172494

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with progressive fibrosis and death within 2-3 y of diagnosis. IPF incidence and prevalence rates are increasing annually with few effective treatments available. Inhibition of IL-6 results in the attenuation of pulmonary fibrosis in mice. It is unclear whether this is due to blockade of classical signaling, mediated by membrane-bound IL-6Rα, or trans signaling, mediated by soluble IL-6Rα (sIL-6Rα). Our study assessed the role of sIL-6Rα in IPF. We demonstrated elevations of sIL-6Rα in IPF patients and in mice during the onset and progression of fibrosis. We demonstrated that protease-mediated cleavage from lung macrophages was important in production of sIL-6Rα. In vivo neutralization of sIL-6Rα attenuated pulmonary fibrosis in mice as seen by reductions in myofibroblasts, fibronectin, and collagen in the lung. In vitro activation of IL-6 trans signaling enhanced fibroblast proliferation and extracellular matrix protein production, effects relevant in the progression of pulmonary fibrosis. Taken together, these findings demonstrate that the production of sIL-6Rα from macrophages in the diseased lung contributes to IL-6 trans signaling that in turn influences events crucial in pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/inmunología , Interleucina-6/inmunología , Macrófagos Alveolares/inmunología , Fibrosis Pulmonar/inmunología , Receptores de Interleucina-6/inmunología , Transducción de Señal/inmunología , Animales , Colágeno/inmunología , Modelos Animales de Enfermedad , Femenino , Fibronectinas/inmunología , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/mortalidad , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/terapia , Interleucina-6/genética , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Miofibroblastos/inmunología , Miofibroblastos/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología
10.
Arthritis Rheumatol ; 66(4): 1010-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24757152

RESUMEN

OBJECTIVE: Systemic sclerosis (SSc) is a chronic autoimmune disease clinically manifesting as progressive fibrosis of the skin and internal organs. Recent microarray studies demonstrated that cadherin 11 (Cad-11) expression is increased in the affected skin of patients with SSc. The purpose of this study was to examine our hypothesis that Cad-11 is a mediator of dermal fibrosis. METHODS: Biopsy samples of skin from SSc patients and healthy control subjects were used for real-time quantitative polymerase chain reaction analysis to assess Cad-11 expression and for immunohistochemistry to determine the expression pattern of Cad-11. To determine whether Cad-11 is a mediator of dermal fibrosis, Cad-11-deficient mice and anti-Cad-11 monoclonal antibodies (mAb) were used in the bleomycin-induced dermal fibrosis model. In vitro studies with dermal fibroblasts and bone marrow-derived macrophages were used to determine the mechanisms by which Cad-11 contributes to the development of tissue fibrosis. RESULTS: Levels of messenger RNA for Cad-11 were increased in skin biopsy samples from patients with SSc and correlated with the modified Rodnan skin thickness scores. Cad-11 expression was localized to dermal fibroblasts and macrophages in SSc skin. Cad-11-knockout mice injected with bleomycin had markedly attenuated dermal fibrosis, as quantified by measurements of skin thickness, collagen levels, myofibroblast accumulation, and profibrotic gene expression, in lesional skin as compared to the skin of wild-type mice. In addition, anti-Cad-11 mAb decreased fibrosis at various time points in the bleomycin-induced dermal fibrosis model. In vitro studies demonstrated that Cad-11 regulated the production of transforming growth factor ß (TGFß) by macrophages and the migration of fibroblasts. CONCLUSION: These data demonstrate that Cad-11 is a mediator of dermal fibrosis and TGFß production and suggest that Cad-11 may be a therapeutic target in SSc.


Asunto(s)
Cadherinas/metabolismo , Fibroblastos/metabolismo , Esclerodermia Sistémica/metabolismo , Piel/metabolismo , Adulto , Animales , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Fibroblastos/patología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Piel/patología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
Biochem J ; 456(2): 241-51, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24032673

RESUMEN

Chemokines mediate diverse functions from organogenesis to mobilizing leucocytes, and are unusual agonists for class-A GPCRs (G-protein-coupled receptors) because of their large size and multi-domain structure. The current model for receptor activation, which involves interactions between chemokine N-loop and receptor N-terminal residues (Site-I) and between chemokine N-terminal and receptor extracellular loop/transmembrane residues (Site-II), fails to describe differences in ligand/receptor selectivity and the activation of multiple signalling pathways. In the present study, we show in neutrophil-activating chemokine CXCL8 that the highly conserved GP (glycine-proline) motif located distal to both N-terminal and N-loop residues couples Site-I and Site-II interactions. GP mutants showed large differences from native-like to complete loss of function that could not be correlated with the specific mutation, receptor affinity or subtype, or a specific signalling pathway. NMR studies indicated that the GP motif does not influence Site-I interactions, but molecular dynamics simulations suggested that this motif dictates substates of the CXCL8 conformational ensemble. We conclude that the GP motif enables diverse receptor functions by controlling cross-talk between Site-I and Site-II, and further propose that the repertoire of chemokine functions is best described by a conformational ensemble model in which a network of long-range coupled indirect interactions mediate receptor activity.


Asunto(s)
Interleucina-8/química , Receptores de Interleucina-8A/química , Receptores de Interleucina-8B/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Señalización del Calcio , Línea Celular , Secuencia Conservada , Femenino , Interleucina-8/metabolismo , Ligandos , Ratones , Ratones Endogámicos BALB C , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Neutrófilos/inmunología , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/metabolismo
12.
Am J Respir Cell Mol Biol ; 49(6): 1038-47, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23855769

RESUMEN

Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death worldwide. The development of pulmonary hypertension (PH) in patients with COPD is strongly associated with increased mortality. Chronic inflammation and changes to the lung extracellular matrix (ECM) have been implicated in the pathogenesis of COPD, yet the mechanisms that lead to PH secondary to COPD remain unknown. Our experiments using human lung tissue show increased expression levels of the adenosine A2B receptor (ADORA2B) and a heightened deposition of hyaluronan (HA; a component of the ECM) in remodeled vessels of patients with PH associated with COPD. We also demonstrate that the expression of HA synthase 2 correlates with mean pulmonary arterial pressures in patients with COPD, with and without a secondary diagnosis of PH. Using an animal model of airspace enlargement and PH, we show that the blockade of ADORA2B is able to attenuate the development of a PH phenotype that correlates with reduced levels of HA deposition in the vessels and the down-regulation of genes involved in the synthesis of HA.


Asunto(s)
Ácido Hialurónico/metabolismo , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Receptor de Adenosina A2B/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Anciano , Animales , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Femenino , Humanos , Hipertensión Pulmonar/patología , Pulmón/irrigación sanguínea , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Purinas/farmacología , Pirazoles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Adenosina A2B/genética
13.
FASEB J ; 27(5): 2013-26, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23392349

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by persistent inflammation and tissue remodeling and is a leading cause of death in the United States. Increased apoptosis of pulmonary epithelial cells is thought to play a role in COPD development and progression. Identification of signaling pathways resulting in increased apoptosis in COPD can be used in the development of novel therapeutic interventions. Deoxyadenosine (dAdo) is a DNA breakdown product that amplifies lymphocyte apoptosis by being phosphorylated to deoxyadenosine triphosphate (dATP). dAdo is maintained at low levels by adenosine deaminase (ADA). This study demonstrated that mice lacking ADA developed COPD manifestations in association with elevated dAdo and dATP levels and increased apoptosis in the lung. Deoxycitidine kinase (DCK), a major enzyme for dAdo phosphorylation, was up-regulated in mouse and human airway epithelial cells in association with air-space enlargement. Hypoxia was identified as a novel regulator of DCK, and inhibition of DCK resulted in diminished dAdo-mediated apoptosis in the lungs. Our results suggest that activating the dAdo-DCK-dATP pathway directly results in increased apoptosis in the lungs of mice with air-space enlargement and suggests a novel therapeutic target for the treatment of COPD.


Asunto(s)
Apoptosis/efectos de los fármacos , Nucleótidos de Desoxiadenina/metabolismo , Desoxiadenosinas/metabolismo , Desoxicitidina Quinasa/metabolismo , Hipoxia/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , 2-Cloroadenosina/análogos & derivados , 2-Cloroadenosina/farmacología , Adenosina Desaminasa/deficiencia , Animales , Células Cultivadas , Desoxiadenosinas/farmacología , Humanos , Ratones , Regulación hacia Arriba
14.
Am J Physiol Lung Cell Mol Physiol ; 304(5): L361-70, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23316066

RESUMEN

Pulmonary endothelial cell (EC) apoptosis has been implicated in the pathogenesis of emphysema. Cigarette smoke (CS) causes lung EC apoptosis and emphysema. In this study, we show that CS exposure increased lung tissue adenosine levels in mice, an effect associated with increased lung EC apoptosis and the development of emphysema. Adenosine has a protective effect against apoptosis via adenosine receptor-mediated signaling. However, sustained elevated adenosine increases alveolar cell apoptosis in adenosine deaminase-deficient mice. We established an in vitro model of sustained adenosine exposure by incubating lung EC with adenosine in the presence of an adenosine deaminase inhibitor, deoxycoformicin. We demonstrated that sustained adenosine exposure caused lung EC apoptosis via nucleoside transporter-facilitated intracellular adenosine uptake, subsequent activation of p38 and JNK in mitochondria, and ultimately mitochondrial defects and activation of the mitochondria-mediated intrinsic pathway of apoptosis. Our results suggest that sustained elevated adenosine may contribute to CS-induced lung EC apoptosis and emphysema. Our data also reconcile the paradoxical effects of adenosine on apoptosis, demonstrating that prolonged exposure causes apoptosis via nucleoside transporter-mediated intracellular adenosine signaling, whereas acute exposure protects against apoptosis via activation of adenosine receptors. Inhibition of adenosine uptake may become a new therapeutic target in treatment of CS-induced lung diseases.


Asunto(s)
Adenosina/metabolismo , Apoptosis/efectos de los fármacos , Células Endoteliales/fisiología , Humo/efectos adversos , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Inhibidores de la Adenosina Desaminasa/farmacología , Animales , Bovinos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lesión Pulmonar , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas de Transporte de Nucleósidos/metabolismo , Pentostatina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Am J Respir Cell Mol Biol ; 47(5): 604-13, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22744860

RESUMEN

Previous studies by our group as well as others have shown that acute adenosine exposure enhances lung vascular endothelial barrier integrity and protects against increased permeability lung edema. In contrast, there is growing evidence that sustained adenosine exposure has detrimental effects on the lungs, including lung edema. It is well established that adenosine modulates lung inflammation. However, little is known concerning the effect of sustained adenosine exposure on lung endothelial cells (ECs), which are critical to the maintenance of the alveolar-capillary barrier. We show that exogenous adenosine plus adenosine deaminase inhibitor caused sustained elevation of adenosine in lung ECs. This sustained adenosine exposure decreased EC barrier function, elevated cellular reactive oxygen species levels, and activated p38, JNK, and RhoA. Inhibition of equilibrative nucleoside transporters (ENTs) prevented sustained adenosine-induced p38 and JNK activation and EC barrier dysfunction. Inhibition of p38, JNK, or RhoA also partially attenuated sustained adenosine-induced EC barrier dysfunction. These data indicate that sustained adenosine exposure causes lung EC barrier dysfunction via ENT-dependent intracellular adenosine uptake and subsequent activation of p38, JNK, and RhoA. The antioxidant N-acetylcysteine and the NADPH inhibitor partially blunted sustained adenosine-induced JNK activation but were ineffective in attenuation of p38 activation or barrier dysfunction. p38 was activated exclusively in mitochondria, whereas JNK was activated in mitochondria and cytoplasm by sustained adenosine exposure. Our data further suggest that sustained adenosine exposure may cause mitochondrial oxidative stress, leading to activation of p38, JNK, and RhoA in mitochondria and resulting in EC barrier dysfunction.


Asunto(s)
Adenosina/fisiología , Permeabilidad Capilar , Células Endoteliales/metabolismo , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Arteria Pulmonar/patología , Transducción de Señal , Adenosina/farmacología , Adenosina Desaminasa/metabolismo , Inhibidores de la Adenosina Desaminasa/farmacología , Uniones Adherentes/metabolismo , Animales , Apoptosis , Bovinos , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Mitocondrias Musculares/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Estrés Oxidativo , Pentostatina/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Fibras de Estrés/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
16.
J Biol Chem ; 286(42): 36378-84, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21903575

RESUMEN

Paclitaxel has powerful anticancer activity, but some tumors are inherently resistant to the drug, whereas others are initially sensitive but acquire resistance during treatment. To deal with this problem, it will be necessary to understand the mechanisms of drug action and resistance. Recent studies indicate that paclitaxel blocks cell division by inhibiting the detachment of microtubules from centrosomes. Here, we demonstrate that mitotic centromere-associated kinesin (MCAK), a kinesin-related protein that destabilizes microtubules, plays an important role in microtubule detachment. Depletion of MCAK altered mitotic spindle morphology, increased the frequency of lagging chromosomes, and inhibited the proliferation of WT CHO cells, confirming that it is an essential protein for cell division. In contrast, MCAK depletion rescued the proliferation of mutant paclitaxel-dependent cell lines that are unable to divide because of defective spindle function resulting from altered α-tubulin or class III ß-tubulin overexpression. In concert with the correction of mitotic defects, loss of MCAK reversed an aberrantly high frequency of microtubule detachment in the mutant cells and increased their sensitivity to paclitaxel. The results indicate that MCAK affects cell sensitivity to mitotic inhibitors by modulating the frequency of microtubule detachment, and they demonstrate that changes in a microtubule-interacting protein can reverse the effects of mutant tubulin expression.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos/fisiología , Cinesinas/metabolismo , Microtúbulos/metabolismo , Paclitaxel/farmacología , Huso Acromático/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Cinesinas/genética , Microtúbulos/genética , Mutación , Huso Acromático/genética , Tubulina (Proteína)/biosíntesis , Tubulina (Proteína)/genética
17.
PLoS One ; 6(7): e22667, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21799929

RESUMEN

BACKGROUND: Chronic lung diseases are the third leading cause of death in the United States due in part to an incomplete understanding of pathways that govern the progressive tissue remodeling that occurs in these disorders. Adenosine is elevated in the lungs of animal models and humans with chronic lung disease where it promotes air-space destruction and fibrosis. Adenosine signaling increases the production of the pro-fibrotic cytokine interleukin-6 (IL-6). Based on these observations, we hypothesized that IL-6 signaling contributes to tissue destruction and remodeling in a model of chronic lung disease where adenosine levels are elevated. METHODOLOGY/PRINCIPAL FINDINGS: We tested this hypothesis by neutralizing or genetically removing IL-6 in adenosine deaminase (ADA)-deficient mice that develop adenosine dependent pulmonary inflammation and remodeling. Results demonstrated that both pharmacologic blockade and genetic removal of IL-6 attenuated pulmonary inflammation, remodeling and fibrosis in this model. The pursuit of mechanisms involved revealed adenosine and IL-6 dependent activation of STAT-3 in airway epithelial cells. CONCLUSIONS/SIGNIFICANCE: These findings demonstrate that adenosine enhances IL-6 signaling pathways to promote aspects of chronic lung disease. This suggests that blocking IL-6 signaling during chronic stages of disease may provide benefit in halting remodeling processes such as fibrosis and air-space destruction.


Asunto(s)
Adenosina/metabolismo , Interleucina-6/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Neumonía/metabolismo , Neumonía/patología , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Femenino , Técnicas de Inactivación de Genes , Interleucina-6/deficiencia , Interleucina-6/genética , Interleucina-6/inmunología , Lesión Pulmonar/complicaciones , Lesión Pulmonar/tratamiento farmacológico , Masculino , Ratones , Neumonía/complicaciones , Neumonía/tratamiento farmacológico , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/tratamiento farmacológico , Factor de Transcripción STAT3/metabolismo
18.
J Immunol ; 186(2): 1097-106, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149612

RESUMEN

Adenosine is an extracellular signaling molecule that is generated in response to cell injury where it orchestrates tissue protection and repair. Whereas adenosine is best known for promoting anti-inflammatory activities during acute injury responses, prolonged elevations can enhance destructive tissue remodeling processes associated with chronic disease states. The generation of adenosine and the subsequent activation of the adenosine 2B receptor (A(2B)R) is an important processes in the regulation of both acute and chronic lung disease. The goal of this study was to examine the contribution of the A(2B)R in models of bleomycin-induced lung injury that exhibit varying degrees of acute and chronic injury. Intratracheal bleomycin exposure results in substantial acute lung injury followed by progressive fibrosis. In this model, genetic removal of the A(2B)R resulted in enhanced loss of barrier function and increased pulmonary inflammation, with few differences in indexes of pulmonary fibrosis. These results support an anti-inflammatory role for this receptor in this model of acute lung injury. In contrast, systemic exposure of mice to bleomycin resulted in modest acute lung injury together with progressive pulmonary fibrosis. In this model, the effects of A(2B)R removal on acute lung injury were negligible; however, there were substantial reductions in pulmonary fibrosis, supporting a profibrotic role for this receptor. A(2B)R-dependent regulation of IL-6 production was identified as a potential mechanism involved in the diminished pulmonary fibrosis seen in A(2B)R knockout mice exposed to i.p. bleomycin. These studies highlight the distinct roles of A(2B)R signaling during acute and chronic stages of lung injury.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Bleomicina/toxicidad , Receptor de Adenosina A2B/fisiología , Enfermedad Aguda , Lesión Pulmonar Aguda/patología , Animales , Enfermedad Crónica , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Intubación Intratraqueal , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA