Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Pathol ; 256(3): 310-320, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34825713

RESUMEN

The clinical significance of B7H3 (CD276) and its cleavage product soluble B7H3 (sB7H3) in idiopathic pulmonary fibrosis (IPF) is unknown. Mounting evidence suggests the potential utility of peripheral blood myeloid cell enumeration to predict disease outcome and indicate active lung disease. Here we hypothesized that sB7H3 is involved in regulation of circulating myeloid cells in pulmonary fibrosis. In support of this possibility, both plasma sB7H3 and B7H3+ cells were elevated in IPF patient blood samples, which correlated negatively with lung function. To analyze its function, the effects of sB7H3 on naïve or bleomycin-treated mice were examined. The results revealed that sB7H3 injection induced an influx of myeloid-derived suppressor cells (MDSCs) and Ccl2 expression in lung tissue of naïve mice, accompanied by enhanced overall inflammation. Additionally, sB7H3 caused accumulation of MDSCs in bone marrow with increased expression of inflammatory cytokines. Notably, in vitro assays revealed chemotaxis of MDSCs to sB7H3, which was dependent on TLT-2 (TREML2), a putative receptor for sB7H3. Thus, increased circulating sB7H3 and/or B7H3+ cells in IPF patient blood samples correlated with lung function decline and potential immunosuppressive status. The correlation of sB7H3 with deterioration of lung function might be due to its ability to enhance inflammation and recruitment of MDSCs into the lung and their expansion in the bone marrow, and thus potentially contribute to IPF exacerbation. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Antígenos B7/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Anciano , Animales , Antígenos B7/genética , Antígenos B7/toxicidad , Bleomicina , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiotaxis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/inmunología , Fibrosis Pulmonar Idiopática/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal
2.
Am J Respir Cell Mol Biol ; 65(5): 489-499, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34107237

RESUMEN

The Wnt/ß-catenin pathway initiates a signaling cascade that is critical in cell differentiation and the normal development of multiple organ systems. The reactivation of this pathway has been documented in experimental and human idiopathic pulmonary fibrosis, wherein Wnt/ß-catenin activation has been implicated in epithelial-cell repair. Furthermore, the canonical ligand Wnt3a is known to induce myofibroblast differentiation; however, the role of noncanonical Wnt ligands remains unclear. This study showed significantly higher levels of Wnt11 expression in cells from both patients with idiopathic pulmonary fibrosis and bleomycin-treated mice, as well as in TGFß-treated mouse lung fibroblasts. Moreover, Wnt11 induced myofibroblast differentiation as manifested by increased α-SMA (ACTA2) expression, which was similar to that induced by canonical Wnt3a/ß-catenin signaling. Further investigation revealed that Wnt11 induction of α-SMA was associated with the activation of JNK (c-Jun N-terminal kinase)/c-Jun signaling and was inhibited by a JNK inhibitor. The potential importance of this signaling pathway was supported by in vivo evidence showing significantly increased levels of Wnt11 and activated JNK in the lungs of mice with bleomycin-induced pulmonary fibrosis. Interestingly, fibroblasts did not express canonical Wnt3a, but treatment of these cells with exogenous Wnt3a induced endogenous Wnt11 and Wnt5a, resulting in repression of the Wnt3a/ß-catenin target gene Axin2. These findings suggested that the noncanonical Wnt induction of myofibroblast differentiation mediated by the JNK/c-Jun pathway might play a significant role in pulmonary fibrosis, in addition to or in synergy with canonical Wnt3a/ß-catenin signaling. Moreover, Wnt3a activation of noncanonical Wnt signaling might trigger a switch from canonical to noncanonical Wnt signaling to induce myofibroblast differentiation.


Asunto(s)
Fibrosis Pulmonar Idiopática/patología , Miofibroblastos/patología , Vía de Señalización Wnt/fisiología , Proteína Wnt3A/metabolismo , Animales , Bleomicina/toxicidad , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , Proteína Wnt3A/genética , beta Catenina/genética , beta Catenina/metabolismo
3.
J Biol Chem ; 294(22): 8861-8871, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31000627

RESUMEN

Mutations in the genes encoding telomerase reverse transcriptase (TERT) and telomerase's RNA components as well as shortened telomeres are risk factors for idiopathic pulmonary fibrosis, where repetitive injury to the alveolar epithelium is considered a key factor in pathogenesis. Given the importance of TERT in stem cells, we hypothesized that TERT plays an important role in epithelial repair and that its deficiency results in exacerbation of fibrosis by impairing this repair/regenerative process. To evaluate the role of TERT in epithelial cells, we generated type II alveolar epithelial cell (AECII)-specific TERT conditional knockout (SPC-Tert cKO) mice by crossing floxed Tert mice with inducible SPC-driven Cre mice. SPC-Tert cKO mice did not develop pulmonary fibrosis spontaneously up to 9 months of TERT deficiency. However, upon bleomycin treatment, they exhibited enhanced lung injury, inflammation, and fibrosis compared with control mice, accompanied by increased pro-fibrogenic cytokine expression but without a significant effect on AECII telomere length. Moreover, selective TERT deficiency in AECII diminished their proliferation and induced cellular senescence. These findings suggest that AECII-specific TERT deficiency enhances pulmonary fibrosis by heightening susceptibility to bleomycin-induced epithelial injury and diminishing epithelial regenerative capacity because of increased cellular senescence. We confirmed evidence for increased AECII senescence in idiopathic pulmonary fibrosis lungs, suggesting potential clinical relevance of the findings from our animal model. Our results suggest that TERT has a protective role in AECII, unlike its pro-fibrotic activity, observed previously in fibroblasts, indicating that TERT's role in pulmonary fibrosis is cell type-specific.


Asunto(s)
Senescencia Celular , Fibrosis Pulmonar/etiología , Telomerasa/genética , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina/farmacología , Proliferación Celular , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Telomerasa/deficiencia , Telomerasa/metabolismo , Telómero/metabolismo , Acortamiento del Telómero
4.
Am J Respir Crit Care Med ; 200(8): 1032-1044, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31106564

RESUMEN

Rationale: The impact of lung insult on the bone marrow (BM) and subsequent disease is unknown.Objectives: To study alterations in the BM in response to lung injury/fibrosis and examine their impact on subsequent lung insult.Methods: BM cells from control or bleomycin-treated donor mice were transplanted into naive mice, which were subsequently evaluated for bleomycin-induced pulmonary fibrosis. In addition, the effect of prior bleomycin treatment on subsequent fibrosis was examined in wild-type and B7H3-knockout mice. Samples from patients with idiopathic pulmonary fibrosis were analyzed for potential clinical relevance of the findings.Measurements and Main Results: Recipient mice transplanted with BM from bleomycin-pretreated donors showed significant exacerbation of subsequent fibrosis with increased B7H3+ cell numbers and a T-helper cell type 2-skewed phenotype. Pretreatment with a minimally fibrogenic/nonfibrogenic dose of bleomycin also caused exacerbation, but not in B7H3-deficient mice. Exacerbation was not observed if the mice received naive BM cell transplant after the initial bleomycin pretreatment. Soluble B7H3 stimulated BM Ly6Chi monocytic cell expansion in vitro and caused similar expansion in the lung in vivo. Notably, soluble B7H3 was elevated in plasma of patients with idiopathic pulmonary fibrosis and in BAL fluid in those with acute exacerbation. Finally, ST2 deficiency diminished the bleomycin-induced B7H3 and IL-13 upregulation, suggesting a role for type 2 innate lymphoid cells.Conclusions: Pulmonary fibrosis caused significant alterations in BM with expansion and activation of monocytic cells, which enhanced fibrosis when transplanted to naive recipients with potential mediation by a novel role for B7H3 in the pathophysiology of pulmonary fibrosis in both mice and humans.


Asunto(s)
Bleomicina/efectos adversos , Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fibrosis Pulmonar Idiopática/complicaciones , Fibrosis Pulmonar Idiopática/fisiopatología , Inmunidad Innata/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales
5.
J Pathol ; 245(4): 399-409, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29722022

RESUMEN

Recent evidence supports that bone marrow (BM)-derived hematopoietic progenitor cells play an important role in lung injury and fibrosis. While these cells give rise to multiple cell types, the ST2 (Il1rl1)-expressing group 2 innate lymphoid cells (ILC2s) derived from BM progenitors have been implicated in tissue repair and remodeling, including in lung fibrosis. To further investigate the precise role of BM-derived ILC2s in the pathogenesis of fibrotic lung disease, their importance in the bleomycin-induced lung fibrosis model was evaluated by analyzing the effects of selective ST2 deficiency in the BM compartment. The results showed that while ST2-sufficient control mice exhibited activation of lung IL-33/ST2 signaling, ILC2 recruitment, IL-13 induction, and fibrosis, these responses were significantly diminished in ST2-deficient-BM chimera mice, with selective loss of ST2 expression only in the BM. This diminished response to bleomycin was similar to that seen in ST2 global knockout mice, suggesting the predominant importance of ST2 from the BM compartment. In wild-type mice, ILC2 recruitment to the lung was accompanied by a concomitant decrease in ST2+ BM cells. ST2-deficient BM cells were unresponsive to IL-33-induced ILC2 maturation. Finally, lineage-negative wild-type, but not ST2-deficient BM cells from bleomycin-treated mice stimulated lung fibroblast type I collagen expression, which was associated with elevated TGFß expression in the BM cells. Taken together, these findings suggested that the BM-derived ILC2s were recruited to fibrotic lung through the IL-33/ST2 pathway, and contributed to fibroblast activation to promote lung fibrosis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Inmunidad Innata , Inmunidad Mucosa , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Pulmón/metabolismo , Células Progenitoras Linfoides/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Proteína 1 Similar al Receptor de Interleucina-1/genética , Interleucina-33/metabolismo , Pulmón/inmunología , Pulmón/patología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Comunicación Paracrina , Fenotipo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
6.
J Immunol ; 197(1): 303-12, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27206766

RESUMEN

Amphiregulin (AREG), an epidermal growth factor receptor ligand, is implicated in tissue repair and fibrosis, but its cellular source and role in regeneration versus fibrosis remain unclear. In this study, we hypothesize that AREG induced in bone marrow-derived CD11c(+) cells is essential for pulmonary fibrosis. Thus, the objectives were to evaluate the importance and role of AREG in pulmonary fibrosis, identify the cellular source of AREG induction, and analyze its regulation of fibroblast function and activation. The results showed that lung AREG expression was significantly induced in bleomycin-induced pulmonary fibrosis. AREG deficiency in knockout mice significantly diminished pulmonary fibrosis. Analysis of AREG expression in major lung cell types revealed induction in fibrotic lungs predominantly occurred in CD11c(+) cells. Moreover, depletion of bone marrow-derived CD11c(+) cells suppressed both induction of lung AREG expression and pulmonary fibrosis. Conversely, adoptive transfer of bone marrow-derived CD11c(+) cells from bleomycin-treated donor mice exacerbated pulmonary fibrosis, but not if the donor cells were made AREG deficient prior to transfer. CD11c(+) cell-conditioned media or coculture stimulated fibroblast proliferation, activation, and myofibroblast differentiation in an AREG-dependent manner. Furthermore, recombinant AREG induced telomerase reverse transcriptase, which appeared to be essential for the proliferative effect. Finally, AREG significantly enhanced fibroblast motility, which was associated with increased expression of α6 integrin. These findings suggested that induced AREG specifically in recruited bone marrow-derived CD11c(+) cells promoted bleomycin-induced pulmonary fibrosis by activation of fibroblast telomerase reverse transcriptase-dependent proliferation, motility, and indirectly, myofibroblast differentiation.


Asunto(s)
Anfirregulina/metabolismo , Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Fibrosis Pulmonar/inmunología , Factor de Transcripción STAT2/metabolismo , Anfirregulina/genética , Animales , Comunicación Autocrina , Antígeno CD11c/metabolismo , Diferenciación Celular , Células Cultivadas , Reactividad Cruzada , Femenino , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT2/genética , Receptores Toll-Like/metabolismo
7.
Am J Pathol ; 185(11): 3066-75, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26358219

RESUMEN

Notch signaling pathway is involved in the regulation of cell fate, differentiation, proliferation, and apoptosis in development and disease. Previous studies suggest the importance of Notch1 in myofibroblast differentiation in lung alveogenesis and fibrosis. However, direct in vivo evidence of Notch1-mediated myofibroblast differentiation is lacking. In this study, we examined the effects of conditional mesenchymal-specific deletion of Notch1 on pulmonary fibrosis. Crossing of mice bearing the floxed Notch1 gene with α2(I) collagen enhancer-Cre-ER(T)-bearing mice successfully generated progeny with a conditional knockout (CKO) of Notch1 in collagen I-expressing (mesenchymal) cells on treatment with tamoxifen (Notch1 CKO). Because Notch signaling is known to be activated in the bleomycin model of pulmonary fibrosis, control and Notch1 CKO mice were analyzed for their responses to bleomycin treatment. The results showed significant attenuation of pulmonary fibrosis in CKO relative to control mice, as examined by collagen deposition, myofibroblast differentiation, and histopathology. However, there were no significant differences in inflammatory or immune cell influx between bleomycin-treated CKO and control mouse lungs. Analysis of isolated lung fibroblasts confirmed absence of Notch1 expression in cells from CKO mice, which contained fewer myofibroblasts and significantly diminished collagen I expression relative to those from control mice. These findings revealed an essential role for Notch1-mediated myofibroblast differentiation in the pathogenesis of pulmonary fibrosis.


Asunto(s)
Bleomicina/efectos adversos , Colágeno/metabolismo , Fibrosis Pulmonar/patología , Receptor Notch1/deficiencia , Transducción de Señal , Animales , Apoptosis , Diferenciación Celular , Colágeno/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Pulmón/metabolismo , Pulmón/patología , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fibrosis Pulmonar/inducido químicamente , Receptor Notch1/genética , Eliminación de Secuencia
8.
Am J Pathol ; 185(10): 2768-76, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26261086

RESUMEN

Subcutaneous lipoatrophy characteristically accompanies dermal fibrosis with de novo emergence of myofibroblasts such as in systemic sclerosis or scleroderma. Recently dermal adipocytes were shown to have the capacity to differentiate to myofibroblasts in an animal model. Transforming growth factor ß can induce this phenomenon in vitro; however its in vivo significance is unclear. Because found in inflammatory zone 1 (FIZZ1) is an inducer of myofibroblast differentiation but an inhibitor of adipocyte differentiation, we investigated its potential role in adipocyte transdifferentiation to myofibroblast in dermal fibrosis. FIZZ1 caused significant and rapid suppression of the expression of fatty acid binding protein 4 and peroxisome proliferator-activated receptor-γ in adipocytes, consistent with dedifferentiation with loss of lipid and Oil Red O staining. The suppression was accompanied subsequently with stimulation of α-smooth muscle actin and type I collagen expression, indicative of myofibroblast differentiation. In vivo FIZZ1 expression was significantly elevated in the murine bleomycin-induced dermal fibrosis model, which was associated with significant reduction in adipocyte marker gene expression and subcutaneous lipoatrophy. Finally, FIZZ1 knockout mice exhibited significantly reduced bleomycin-induced dermal fibrosis with greater preservation of the subcutaneous fat than wild-type mice. These findings suggested that the FIZZ1 induction of adipocyte transdifferentiation to myofibroblast might be a key pathogenic mechanism for the accumulation of myofibroblasts in dermal fibrosis.


Asunto(s)
Diferenciación Celular/fisiología , Transdiferenciación Celular/fisiología , Fibroblastos/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lipodistrofia/metabolismo , Miofibroblastos/patología , Adipocitos/patología , Animales , Fibrosis/metabolismo , Lipodistrofia/patología , Ratones Endogámicos C57BL , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología
9.
Am J Respir Cell Mol Biol ; 52(4): 418-28, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25140582

RESUMEN

Hedgehog signaling plays important roles in cell development and differentiation. In this study, the ability of Sonic Hedgehog (SHH) to induce myofibroblast differentiation was analyzed in isolated human lung fibroblasts, and its in vivo significance was evaluated in rodent bleomycin-induced pulmonary fibrosis. The results showed that SHH could induce myofibroblast differentiation in human lung fibroblasts in a Smo- and Gli1-dependent manner. Gel shift analysis, chromatin immunoprecipitation assay, and site-directed mutagenesis revealed that a Gli1 binding consensus in the α-SMA gene promoter was important for mediating SHH-induced myofibroblast differentiation. Analysis of Hedgehog reemergence in vivo revealed that of all three Hedgehog isoforms, only SHH was significantly induced in bleomycin-injured lung along with Gli1. The induction of SHH was only noted in epithelial cells, and its expression was undetectable in lung fibroblasts or macrophages. transforming growth factor (TGF)-ß induced SHH significantly in cultured alveolar epithelial cells, whereas SHH induced TGF-ß in lung fibroblasts. Pulmonary fibrosis and α-smooth muscle actin (α-SMA) expression were significantly reduced in mice that were Smo deficient only in type I collagen-expressing cells. Thus, the reemergence of SHH in epithelial cells could result in induction of myofibroblast differentiation in a Smo-dependent manner and subsequent Gli1 activation of the α-SMA promoter.


Asunto(s)
Proteínas Hedgehog/metabolismo , Fibrosis Pulmonar/metabolismo , Actinas/biosíntesis , Actinas/genética , Animales , Secuencia de Bases , Células Cultivadas , Transición Epitelial-Mesenquimal , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Fibrosis Pulmonar/patología , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1
10.
J Pathol ; 230(2): 205-14, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23401096

RESUMEN

Stem cell factor (SCF) and its receptor c-Kit have been implicated in tissue remodelling and fibrosis. Alveolar fibroblasts from patients with diffuse interstitial fibrosis secrete more SCF. However, its precise role remains unclear. In this study the potential role of the SCF-c-Kit axis in pulmonary fibrosis was examined. Fibrosis was induced by intratracheal instillation of bleomycin (BLM), which caused increased SCF levels in plasma, bronchoalveolar lavage fluid (BALF) and lung tissue, as well as increased expression by lung fibroblasts. These changes were accompanied by increased numbers of bone marrow-derived c-Kit(+) cells in the lung, with corresponding depletion in bone marrow. Both recombinant SCF and lung extracts from BLM-treated animals induced bone-marrow cell migration, which was blocked by c-Kit inhibitor. The migrated cells promoted myofibroblast differentiation when co-cultured with fibroblasts, suggesting a paracrine pathogenic role. Interestingly, lung fibroblast cultures contained a subpopulation of cells that expressed functionally active c-Kit, which were significantly greater and more responsive to SCF induction when isolated from fibrotic lungs, including those from patients with idiopathic pulmonary fibrosis (IPF). This c-Kit(+) subpopulation was αSMA-negative and expressed lower levels of collagen I but significantly higher levels of TGFß than c-Kit-negative cells. SCF deficiency achieved by intratracheal treatment with neutralizing anti-SCF antibody or by use of Kitl(Sl)/Kitl(Sl-d) mutant mice in vivo resulted in significant reduction in pulmonary fibrosis. Taken together, the SCF-c-Kit pathway was activated in BLM-injured lung and might play a direct role in pulmonary fibrosis by the recruitment of bone marrow progenitor cells capable of promoting lung myofibroblast differentiation.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Lesión Pulmonar/metabolismo , Proteínas Proto-Oncogénicas c-kit/fisiología , Fibrosis Pulmonar/metabolismo , Transducción de Señal/fisiología , Factor de Células Madre/metabolismo , Animales , Bleomicina/toxicidad , Células de la Médula Ósea/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/química , Recuento de Células , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Proteínas Recombinantes/farmacología , Factor de Células Madre/deficiencia , Factor de Células Madre/farmacología , Extractos de Tejidos/farmacología
11.
Am J Respir Crit Care Med ; 188(8): 976-84, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24010731

RESUMEN

RATIONALE: Bone marrow (BM)-derived cells have been implicated in pulmonary fibrosis. However, their precise role in pathogenesis is incompletely understood. OBJECTIVES: To elucidate roles of BM-derived cells in bleomycin-induced pulmonary fibrosis, and clarify their potential relationship to lung hematopoietic progenitor cells (LHPCs). METHODS: GFP BM-chimera mice treated with or without bleomycin were used to assess the BM-derived cells. MEASUREMENTS AND MAIN RESULTS: GFP(+) cells in the chimera lung were found to be comprised of two distinct phenotypes: GFP(hi) and GFP(low) cells. The GFP(hi), but not GFP(low), population was significantly increased after bleomycin treatment. Flow-cytometric analysis and quantitative real-time polymerase chain reaction revealed that GFP(hi) cells exhibited phenotypic characteristics of CD11c(+) dendritic cells and macrophages. GFP(hi) cell conditioned media were chemotactic for fibroblasts obtained from fibrotic but not normal lung in vitro. Moreover, adoptive transfer of GFP(hi) cells exacerbated fibrosis in recipient mice, similar to that seen on adoptive transfer of BM-derived CD11c(+) cells from donor bleomycin-treated mice. Next, we evaluated the potential of LHPCs as the source of GFP(hi) cells. Isolation of LHPCs by flow sorting revealed enrichment in cKit(+)/Sca1(-)/Lin(-) cells, most of which were GFP(+) indicating their BM origin. The number of LHPCs increased rapidly after bleomycin treatment. Furthermore, stem cell factor induced LHPC proliferation, whereas granulocyte-macrophage-colony stimulating factor induced differentiation to GFP(hi) cells. CONCLUSIONS: BM-derived LHPCs with a novel phenotype could differentiate into GFP(hi) cells, which enhanced pulmonary fibrosis. Targeting this mobilized LHPCs might represent a novel therapeutic approach in chronic fibrotic lung diseases.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Fibrosis Pulmonar/etiología , Animales , Bleomicina/farmacología , Células Cultivadas , Quimera/fisiología , Modelos Animales de Enfermedad , Femenino , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología
12.
Methods Cell Biol ; 184: 1-15, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38555150

RESUMEN

Recently discovered heterogeneous myeloid-derived suppressor cells (MDSCs) are some of the most discussed immunosuppressive cells in contemporary immunology, especially in the tumor microenvironment, and are defined primarily by their T cell immunosuppressive function. The importance of these cells extend to other chronic pathological conditions as well, including chronic infection, inflammation, and tissue remodeling. In many of these conditions, their accumulation/expansion correlates with disease progression, poor prognosis, and reduced survival, which highlights the potential of how these cells may be used in a clinical setting as both prognostic factor and therapeutic target. In healthy individuals, these cells are usually not present in the circulation. Therefore, monitoring this cell population is of potential clinical significance, and utility in basic research. However, these cells have a complex phenotype without one single marker of sufficient specificity for their identification. Flow cytometry is a powerful tool allowing multi-parameter analysis of heterogeneous cell populations, which makes it ideally suitable for the complex phenotypic analysis essential for identification and enumeration of circulating MDSCs. This approach has the potential to provide a novel clinically useful tool for assessment of prognosis and treatment outcomes. The protocol in this chapter describes a flow cytometric analysis to identify and quantify MDSCs from human or mouse whole blood leukocytes and peripheral blood mononuclear cells, as well as a single cell suspension from solid tissue, by using multicolor fluorescence-conjugated antibodies against their surface markers.


Asunto(s)
Células Supresoras de Origen Mieloide , Animales , Ratones , Humanos , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Leucocitos Mononucleares/metabolismo , Citometría de Flujo , Linfocitos T , Fenotipo
13.
Am J Respir Cell Mol Biol ; 49(2): 260-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23526226

RESUMEN

In addition to its expression in stem cells and many cancers, telomerase activity is transiently induced in murine bleomycin (BLM)-induced pulmonary fibrosis with increased levels of telomerase transcriptase (TERT) expression, which is essential for fibrosis. To extend these observations to human chronic fibrotic lung disease, we investigated the expression of telomerase activity in lung fibroblasts from patients with interstitial lung diseases (ILDs), including idiopathic pulmonary fibrosis (IPF). The results showed that telomerase activity was induced in more than 66% of IPF lung fibroblast samples, in comparison with less than 29% from control samples, some of which were obtained from lung cancer resections. Less than 4% of the human IPF lung fibroblast samples exhibited shortened telomeres, whereas less than 6% of peripheral blood leukocyte samples from patients with IPF or hypersensitivity pneumonitis demonstrated shortened telomeres. Moreover, shortened telomeres in late-generation telomerase RNA component knockout mice did not exert a significant effect on BLM-induced pulmonary fibrosis. In contrast, TERT knockout mice exhibited deficient fibrosis that was independent of telomere length. Finally, TERT expression was up-regulated by a histone deacetylase inhibitor, while the induction of TERT in lung fibroblasts was associated with the binding of acetylated histone H3K9 to the TERT promoter region. These findings indicate that significant telomerase induction was evident in fibroblasts from fibrotic murine lungs and a majority of IPF lung samples, whereas telomere shortening was not a common finding in the human blood and lung fibroblast samples. Notably, the animal studies indicated that the pathogenesis of pulmonary fibrosis was independent of telomere length.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Telomerasa/biosíntesis , Telómero/metabolismo , Acetilación/efectos de los fármacos , Alveolitis Alérgica Extrínseca/inducido químicamente , Alveolitis Alérgica Extrínseca/genética , Alveolitis Alérgica Extrínseca/metabolismo , Alveolitis Alérgica Extrínseca/patología , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacología , Bleomicina/efectos adversos , Bleomicina/farmacología , Células Cultivadas , Enfermedad Crónica , Femenino , Fibroblastos/patología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Histonas/genética , Histonas/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Telomerasa/genética , Telómero/genética , Telómero/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
J Immunol ; 187(1): 450-61, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21602491

RESUMEN

Found in inflammatory zone (FIZZ) 2, also known as resistin-like molecule (RELM)-ß, belongs to a novel cysteine-rich secreted protein family named FIZZ/RELM. Its function is unclear, but a closely related family member, FIZZ1, has profibrotic activities. The human ortholog of rodent FIZZ1 has not been identified, but human FIZZ2 has significant sequence homology to both rodent FIZZ2 (59%) and FIZZ1 (50%). Given the greater homology to rodent FIZZ2, analyzing the role of FIZZ2 in a rodent model of bleomycin-induced pulmonary fibrosis would be of greater potential relevance to human fibrotic lung disease. The results showed that FIZZ2 was highly induced in lungs of rodents with bleomycin-induced pulmonary fibrosis and of human patients with idiopathic pulmonary fibrosis. FIZZ2 expression was induced in rodent and human lung epithelial cells by Th2 cytokines, which was mediated via STAT6 signaling. The FIZZ2 induction in murine lungs was found to be essential for pulmonary fibrosis, as FIZZ2 deficiency significantly suppressed pulmonary fibrosis and associated enhanced extracellular matrix and cytokine gene expression. In vitro analysis indicated that FIZZ2 could stimulate type I collagen and α-smooth muscle actin expression in lung fibroblasts. Furthermore, FIZZ2 was shown to have chemoattractant activity for bone marrow (BM) cells, especially BM-derived CD11c(+) dendritic cells. Notably, lung recruitment of BM-derived cells was impaired in FIZZ2 knockout mice. These findings suggest that FIZZ2 is a Th2-associated multifunctional mediator with potentially important roles in the pathogenesis of fibrotic lung diseases.


Asunto(s)
Hormonas Ectópicas/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/metabolismo , Animales , Células de la Médula Ósea/patología , Movimiento Celular/inmunología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibroblastos/patología , Hormonas Ectópicas/genética , Hormonas Ectópicas/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/patología , Fibrosis Pulmonar/patología , Ratas , Ratas Endogámicas F344 , Células Th2/inmunología , Células Th2/metabolismo , Células Th2/patología
15.
Gene ; 884: 147675, 2023 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-37541559

RESUMEN

BACKGROUND: The transcription factor CCAAT/enhancer-binding protein ß (C/EBPß) is implicated in diverse processes and diseases. Its two isoforms, namely liver-enriched activator protein (LAP) and liver-enriched inhibitor protein (LIP) are translated from the same mRNA. They share the same C-terminal DNA binding domain except LAP has an extra N-terminal activation domain. Probably due to its higher affinity for its DNA cognate sequences, LIP can inhibit LAP transcriptional activity even at substoichiometric levels. However, the regulatory mechanism of C/EBPß gene expression and the LAP: LIP ratio is unclear. METHODS: In this study, the C/EBPß promoter sequence was scanned for conserved P53 response element (P53RE), and binding of P53 to the C/EBPß promoter was tested by Electrophoretic Mobility Shift Assay (EMSA) and chromatin immunoprecipitation assay. P53 over-expression and dominant negative P53 expression plasmids were transfected into rat lung fibroblasts and tested for C/EBPß gene transcription and expression. Western blot analysis was used to test the regulation of C/EBPß LAP and LIP isoforms. Constructs containing the LAP 5'untranslated region (5'UTR) or the LIP 5'UTR region were used to test the importance of 5'UTR in the control of C/EBPß LAP and LIP translation. RESULTS: The C/EBPß promoter sequence was found to contain a conserved P53 response element (P53RE), which binds P53 as demonstrated by Electrophoresis Mobility Shift Assay and chromatin immunoprecipitation assays. P53 over-expression suppressed while dominant negative P53 stimulated C/EBPß gene transcription and expression. Western blot analysis showed that P53 differentially regulated the translation of the C/EBPß LAP and LIP isoforms through the regulation of eIF4E and eIF4E-BP1. Further studies with constructs containing the LAP 5'untranslated region (5'UTR) or the LIP 5'UTR region showed that the 5'UTR is important in differential control of C/EBPß LAP and LIP translation. CONCLUSION: Analysis of the effects of P53 on C/EBPß expression revealed a novel mechanism by which P53 could antagonize the effects of C/EBPß on its target gene expression. For the first time, P53 is shown to be a repressor of C/EBPß gene expression at both transcriptional and translational levels, with a differential effect in the magnitude of the effect on LAP vs. LIP isoforms.


Asunto(s)
Factor 4E Eucariótico de Iniciación , Proteína p53 Supresora de Tumor , Ratas , Animales , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Regiones no Traducidas 5'/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Isoformas de Proteínas/metabolismo , Expresión Génica , ADN/metabolismo , Unión Proteica
16.
Front Immunol ; 13: 901349, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36045668

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease without effective curative therapy. Recent evidence shows increased circulating myeloid-derived suppressor cells (MDSCs) in cancer, inflammation, and fibrosis, with some of these cells expressing B7H3. We sought to investigate the role of MDSCs in IPF and its potential mediation via B7H3. Here we prospectively collected peripheral blood samples from IPF patients to analyze for circulating MDSCs and B7H3 expression to assess their clinical significance and potential impact on co-cultured lung fibroblasts and T-cell activation. In parallel, we assess MDSC recruitment and potential B7H3 dependence in a mouse model of pulmonary fibrosis. Expansion of MDSCs in IPF patients correlated with disease severity. Co-culture of soluble B7H3 (sB7H3)-treated mouse monocytic MDSCs (M-MDSCs), but not granulocytic MDSCs (G-MDSCs), activated lung fibroblasts and myofibroblast differentiation. Additionally, sB7H3 significantly enhanced MDSC suppression of T-cell proliferation. Activated M-MDSCs displayed elevated TGFß and Arg1 expression relative to that in G-MDSCs. Treatment with anti-B7H3 antibodies inhibited bone marrow-derived MDSC recruitment into the bleomycin-injured lung, accompanied by reduced expression of inflammation and fibrosis markers. Selective telomerase reverse transcriptase (TERT) deficiency in myeloid cells also diminished MDSC recruitment associated with the reduced plasma level of sB7H3, lung recruitment of c-Kit+ hematopoietic progenitors, myofibroblast differentiation, and fibrosis. Lung single-cell RNA sequencing (scRNA-seq) revealed fibroblasts as a predominant potential source of sB7H3, and indeed the conditioned medium from activated mouse lung fibroblasts had a chemotactic effect on bone marrow (BM)-MDSC, which was abolished by B7H3 blocking antibody. Thus, in addition to their immunosuppressive activity, TERT and B7H3-dependent MDSC expansion/recruitment from BM could play a paracrine role to activate myofibroblast differentiation during pulmonary fibrosis with potential significance for disease progression mediated by sB7H3.


Asunto(s)
Fibrosis Pulmonar Idiopática , Células Supresoras de Origen Mieloide , Animales , Bleomicina , Inflamación , Pulmón , Ratones
17.
Blood ; 114(15): 3244-54, 2009 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-19567879

RESUMEN

Alternatively activated (M2) macrophages play critical roles in diverse chronic diseases, including parasite infections, cancer, and allergic responses. However, little is known about the acquisition and maintenance of their phenotype. We report that M2-macrophage marker genes are epigenetically regulated by reciprocal changes in histone H3 lysine-4 (H3K4) and histone H3 lysine-27 (H3K27) methylation; and the latter methylation marks are removed by the H3K27 demethylase Jumonji domain containing 3 (Jmjd3). We found that continuous interleukin-4 (IL-4) treatment leads to decreased H3K27 methylation, at the promoter of M2 marker genes, and a concomitant increase in Jmjd3 expression. Furthermore, we demonstrate that IL-4-dependent Jmjd3 expression is mediated by STAT6, a major transcription factor of IL-4-mediated signaling. After IL-4 stimulation, activated STAT6 is increased and binds to consensus sites at the Jmjd3 promoter. Increased Jmjd3 contributes to the decrease of H3K27 dimethylation and trimethylation (H3K27me2/3) marks as well as the transcriptional activation of specific M2 marker genes. The decrease in H3K27me2/3 and increase in Jmjd3 recruitment were confirmed by in vivo studies using a Schistosoma mansoni egg-challenged mouse model, a well-studied system known to support an M2 phenotype. Collectively, these data indicate that chromatin remodeling is mechanistically important in the acquisition of the M2-macrophage phenotype.


Asunto(s)
Epigénesis Genética/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Animales , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Modelos Animales de Enfermedad , Femenino , Marcadores Genéticos/genética , Marcadores Genéticos/inmunología , Histonas/genética , Histonas/inmunología , Humanos , Interleucina-4/genética , Interleucina-4/inmunología , Histona Demetilasas con Dominio de Jumonji , Activación de Macrófagos/genética , Metilación , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/inmunología , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/inmunología , Esquistosomiasis mansoni/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Activación Transcripcional/genética , Activación Transcripcional/inmunología
18.
Aging (Albany NY) ; 13(13): 16957-16973, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253690

RESUMEN

Many aging related diseases such as cancer implicate the myofibroblast in disease progression. Furthermore genesis of the myofibroblast is associated with manifestation of cellular senescence of unclear significance. In this study we investigated the role of a common regulator, namely telomerase reverse transcriptase (TERT), in order to evaluate the potential significance of this association between both processes. We analyzed the effects of TERT overexpression or deficiency on expression of CDKN2A and ACTA2 as indicators of senescence and differentiation, respectively. We assess binding of TERT or YB-1, a repressor of both genes, to their promoters. TERT repressed both CDKN2A and ACTA2 expression, and abolished stress-induced expression of both genes. Conversely, TERT deficiency enhanced their expression. Altering CDKN2A expression had no effect on ACTA2 expression. Both TERT and YB-1 were shown to bind the CDKN2A promoter but only YB-1 was shown to bind the ACTA2 promoter. TERT overexpression inhibited CDKN2A promoter activity while stimulating YB-1 expression and activation to repress ACTA2 gene. TERT repressed myofibroblast differentiation and senescence via distinct mechanisms. The latter was associated with TERT binding to the CDKN2A promoter, but not to the ACTA2 promoter, which may require interaction with co-factors such as YB-1.


Asunto(s)
Diferenciación Celular/fisiología , Senescencia Celular/fisiología , Miofibroblastos/fisiología , Telomerasa/fisiología , Actinas/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Humanos , Masculino , Regiones Promotoras Genéticas , ARN Interferente Pequeño , Telomerasa/biosíntesis , Telomerasa/genética
19.
J Clin Invest ; 117(12): 3800-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18008008

RESUMEN

In addition to its well-known expression in the germline and in cells of certain cancers, telomerase activity is induced in lung fibrosis, although its role in this process is unknown. To identify the pathogenetic importance of telomerase in lung fibrosis, we examined the effects of telomerase reverse transcriptase (TERT) deficiency in a murine model of pulmonary injury. TERT-deficient mice showed significantly reduced lung fibrosis following bleomycin (BLM) insult. This was accompanied by a significant reduction in expression of lung alpha-SMA, a marker of myofibroblast differentiation. Furthermore, lung fibroblasts isolated from BLM-treated TERT-deficient mice showed significantly decreased proliferation and increased apoptosis rates compared with cells isolated from control mice. Transplantation of WT BM into TERT-deficient mice restored BLM-induced lung telomerase activity and fibrosis to WT levels. Conversely, transplantation of BM from TERT-deficient mice into WT recipients resulted in reduced telomerase activity and fibrosis. These findings suggest that induction of telomerase in injured lungs may be caused by BM-derived cells, which appear to play an important role in pulmonary fibrosis. Moreover, TERT induction is associated with increased survival of lung fibroblasts, which favors the development of fibrosis instead of injury resolution.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Bleomicina/toxicidad , Fibroblastos/enzimología , Fibrosis Pulmonar/enzimología , Telomerasa/metabolismo , Actinas/biosíntesis , Actinas/genética , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Apoptosis/genética , Trasplante de Médula Ósea , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/genética , Fibroblastos/patología , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Noqueados , Neoplasias/enzimología , Neoplasias/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Telomerasa/genética
20.
Am J Pathol ; 174(5): 1745-55, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349363

RESUMEN

Notch1 is an evolutionarily conserved receptor that regulates cell fate, including such events as differentiation, proliferation, and apoptosis. Myofibroblast differentiation is a key feature of lung fibrosis. Found in inflammatory zone 1 (FIZZ1) has direct fibrogenic properties because of its ability to induce myofibroblast differentiation. However, the downstream signaling pathway that mediates FIZZ1 induction of myofibroblast differentiation remains unknown. The objective of this study was to investigate the involvement of Notch signaling in FIZZ1 induction of lung myofibroblast differentiation and thus explore the potential role of Notch1 in pulmonary fibrosis. The results showed that FIZZ1 increased the expression levels of activated intracellular domain of Notch1 (NIC), its ligand Jagged1, and its target gene Hes1, which were associated with elevated alpha-smooth muscle actin expression levels. Fibroblast alpha-smooth muscle actin expression is induced by the overexpression of NIC but is suppressed by the inhibition of NIC. Moreover, lung fibroblasts that were isolated from mice lacking the GDP-4-keto-6-deoxymannose3,5-epimerase-4-reductase enzyme (FX knockout) exhibited significantly reduced responsiveness to FIZZ1, which was reversed by fucose supplementation. In the absence of exogenous fucose, these FX-deficient cells exhibited defective fucosylation, which is required for Notch signaling. These knockout mice also showed impaired lung fibrosis. These findings suggest that Notch1 signaling in response to FIZZ1 may play a significant role in myofibroblast differentiation during lung fibrosis.


Asunto(s)
Diferenciación Celular , Fibroblastos/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pulmón/citología , Receptor Notch1/metabolismo , Actinas/metabolismo , Animales , Antibióticos Antineoplásicos/toxicidad , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bleomicina/toxicidad , Western Blotting , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Fucosa/administración & dosificación , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hidroliasas/fisiología , Hidroxiprolina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1 , Pulmón/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso/citología , Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Notch1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Serrate-Jagged , Factor de Transcripción HES-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA