Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Respir Cell Mol Biol ; 55(1): 58-71, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26699812

RESUMEN

M2 macrophages are implicated in the development of pulmonary fibrosis as they generate profibrotic signals. The polarization process, at least in part, is regulated by epigenetic modulation. Because Cu,Zn-superoxide dismutase-induced H2O2 can polarize macrophages to a profibrotic M2 phenotype, we hypothesized that modulation of the redox state of the cell is involved in the epigenetic modulation of the macrophage phenotype. In this study, we show that signal transducer and activator of transcription 6 (STAT6) regulates Jumonji domain containing (Jmjd) 3, a histone H3 lysine 27 demethylase, and mutation of a redox-sensitive cysteine in STAT6 attenuates jmjd3 expression. Moreover, Jmjd3 deficiency abrogates profibrotic M2 gene expression. Treatment with leflunomide, which reduces mitochondrial reactive oxygen species production and tyrosine phosphorylation, inhibits jmjd3 expression and M2 polarization, as well as development of a fibrotic phenotype. Taken together, these observations provide evidence that the redox regulation of Jmjd3 is a unique regulatory mechanism for Cu,Zn-superoxide dismutase-mediated profibrotic M2 polarization. Furthermore, leflunomide, which reduces reactive oxygen species production and tyrosine phosphorylation, may prove to be therapeutic in the treatment of asbestos-induced pulmonary fibrosis.


Asunto(s)
Polaridad Celular , Histona Demetilasas con Dominio de Jumonji/metabolismo , Macrófagos/patología , Superóxido Dismutasa-1/metabolismo , Animales , Línea Celular , Polaridad Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-4/metabolismo , Isoxazoles/farmacología , Histona Demetilasas con Dominio de Jumonji/genética , Leflunamida , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 310(1): L86-94, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26545899

RESUMEN

The calcium and calmodulin-dependent kinase II (CaMKII) translates increases in intracellular Ca(2+) into downstream signaling events. Its function in pulmonary pathologies remains largely unknown. CaMKII is a well-known mediator of apoptosis and regulator of endoplasmic reticulum (ER) Ca(2+). ER stress and apoptosis of type II pneumocytes lead to aberrant tissue repair and progressive collagen deposition in pulmonary fibrosis. Thus we hypothesized that CaMKII inhibition alleviates fibrosis in response to bleomycin by attenuating apoptosis and ER stress of type II pneumocytes. We first established that CaMKII was strongly expressed in the distal respiratory epithelium, in particular in surfactant protein-C-positive type II pneumocytes, and activated after bleomycin instillation. We generated a novel transgenic model of inducible expression of the CaMKII inhibitor peptide AC3-I limited to type II pneumocytes (Tg SPC-AC3-I). Tg SPC-AC3-I mice were protected from development of pulmonary fibrosis after bleomycin exposure compared with wild-type mice. CaMKII inhibition also provided protection from apoptosis in type II pneumocytes in vitro and in vivo. Moreover, intracellular Ca(2+) levels and ER stress were increased by bleomycin and significantly blunted with CaMKII inhibition in vitro. These data demonstrate that CaMKII inhibition prevents type II pneumocyte apoptosis and development of pulmonary fibrosis in response to bleomycin. CaMKII inhibition may therefore be a promising approach to prevent or ameliorate the progression of pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Calcio/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Células Epiteliales Alveolares/metabolismo , Animales , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Ratones Transgénicos , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología
3.
FASEB J ; 29(8): 3527-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25953850

RESUMEN

Alternative activation of alveolar macrophages is linked to fibrosis following exposure to asbestos. The scavenger receptor, macrophage receptor with collagenous structure (MARCO), provides innate immune defense against inhaled particles and pathogens; however, a receptor for asbestos has not been identified. We hypothesized that MARCO acts as an initial signaling receptor for asbestos, polarizes macrophages to a profibrotic M2 phenotype, and is required for the development of asbestos-induced fibrosis. Compared with normal subjects, alveolar macrophages isolated from patients with asbestosis express higher amounts of MARCO and have greater profibrotic polarization. Arginase 1 (40-fold) and IL-10 (265-fold) were higher in patients. In vivo, the genetic deletion of MARCO attenuated the profibrotic environment and pulmonary fibrosis in mice exposed to chrysotile. Moreover, alveolar macrophages from MARCO(-/-) mice polarize to an M1 phenotype, whereas wild-type mice have higher Ym1 (>3.0-fold) and nearly 7-fold more active TGF-ß1 in bronchoalveolar lavage (BAL) fluid (BALF). Arg(432) and Arg(434) in domain V of MARCO are required for the polarization of macrophages to a profibrotic phenotype as mutation of these residues reduced FIZZ1 expression (17-fold) compared with cells expressing MARCO. These observations demonstrate that a macrophage membrane protein regulates the fibrotic response to lung injury and suggest a novel target for therapeutic intervention.


Asunto(s)
Macrófagos/metabolismo , Macrófagos/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Receptores Depuradores/metabolismo , Animales , Arginasa/metabolismo , Asbestos Serpentinas/metabolismo , Líquido del Lavado Bronquioalveolar , Línea Celular , Humanos , Interleucina-10/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/metabolismo
4.
Am J Respir Cell Mol Biol ; 52(1): 106-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24988374

RESUMEN

Asthma is a disease of acute and chronic inflammation in which cytokines play a critical role in orchestrating the allergic inflammatory response. IL-13 and transforming growth factor (TGF)-ß promote fibrotic airway remodeling, a major contributor to disease severity. Improved understanding is needed, because current therapies are inadequate for suppressing development of airway fibrosis. IL-13 is known to stimulate respiratory epithelial cells to produce TGF-ß, but the mechanism through which this occurs is unknown. Here, we tested the hypothesis that reactive oxygen species (ROS) are a critical signaling intermediary between IL-13 or allergen stimulation and TGF-ß-dependent airway remodeling. We used cultured human bronchial epithelial cells and an in vivo mouse model of allergic asthma to map a pathway where allergens enhanced mitochondrial ROS, which is an essential upstream signal for TGF-ß activation and enhanced collagen production and deposition in airway fibroblasts. We show that mitochondria in airway epithelium are an essential source of ROS that activate TGF-ß expression and activity. TGF-ß from airway epithelium stimulates collagen expression in fibroblasts, contributing to an early fibrotic response to allergen exposure in cultured human airway cells and in ovalbumin-challenged mice. Treatment with the mitochondrial-targeted antioxidant, (2-(2,2,6,6-Tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mitoTEMPO), significantly attenuated mitochondrial ROS, TGF-ß, and collagen deposition in OVA-challenged mice and in cultured human epithelial cells. Our findings suggest that mitochondria are a critical source of ROS for promoting TGF-ß activity that contributes to airway remodeling in allergic asthma. Mitochondrial-targeted antioxidants may be a novel approach for future asthma therapies.


Asunto(s)
Antioxidantes/farmacología , Asma/tratamiento farmacológico , Asma/metabolismo , Colágeno/biosíntesis , Mitocondrias/metabolismo , Compuestos Organofosforados/farmacología , Piperidinas/farmacología , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Asma/inducido químicamente , Asma/genética , Asma/patología , Células Cultivadas , Colágeno/genética , Modelos Animales de Enfermedad , Humanos , Interleucina-13/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/genética
5.
J Biol Chem ; 289(52): 36204-19, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25378391

RESUMEN

Protein kinase B (Akt) is a key effector of multiple cellular processes, including cell survival. Akt, a serine/threonine kinase, is known to increase cell survival by regulation of the intrinsic pathway for apoptosis. In this study, we found that Akt modulated the mevalonate pathway, which is also linked to cell survival, by increasing Rho GTPase activation. Akt modulated the pathway by phosphorylating mevalonate diphosphate decarboxylase (MDD) at Ser(96). This phosphorylation in macrophages increased activation of Rac1, which enhanced macrophage survival because mutation of MDD (MDDS96A) induced apoptosis. Akt-mediated activation in macrophages was specific for Rac1 because Akt did not increase activity of other Rho GTP-binding proteins. The relationship between Akt and Rac1 was biologically relevant because Akt(+/-) mice had significantly less active Rac1 in alveolar macrophages, and macrophages from Akt(+/-) mice had an increase in active caspase-9 and -3. More importantly, Akt(+/-) mice were significantly protected from the development of pulmonary fibrosis, suggesting that macrophage survival is associated with the fibrotic phenotype. These observations for the first time suggest that Akt plays a critical role in the development and progression of pulmonary fibrosis by enhancing macrophage survival via modulation of the mevalonate pathway.


Asunto(s)
Macrófagos Alveolares/fisiología , Ácido Mevalónico/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Fibrosis Pulmonar/enzimología , Secuencia de Aminoácidos , Animales , Vías Biosintéticas , Polaridad Celular , Supervivencia Celular , Células Cultivadas , Activación Enzimática , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Neuropéptidos/metabolismo , Estrés Oxidativo , Fosforilación , Procesamiento Proteico-Postraduccional , Fibrosis Pulmonar/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína de Unión al GTP rac1/metabolismo
6.
J Biol Chem ; 288(28): 20745-57, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23720777

RESUMEN

Macrophages not only initiate and accentuate inflammation after tissue injury, but they are also involved in resolution and repair. This difference in macrophage activity is the result of a differentiation process to either M1 or M2 phenotypes. M1 macrophages are pro-inflammatory and have microbicidal and tumoricidal activity, whereas the M2 macrophages are involved in tumor progression and tissue remodeling and can be profibrotic in certain conditions. Because mitochondrial Cu,Zn-superoxide dismutase (Cu,Zn-SOD)-mediated H2O2 is crucial for development of pulmonary fibrosis, we hypothesized that Cu,Zn-SOD modulated the macrophage phenotype. In this study, we demonstrate that Cu,Zn-SOD polarized macrophages to an M2 phenotype, and Cu,Zn-SOD-mediated H2O2 levels modulated M2 gene expression at the transcriptional level by redox regulation of a critical cysteine in STAT6. Furthermore, overexpression of Cu,Zn-SOD in mice resulted in a profibrotic environment and accelerated the development of pulmonary fibrosis, whereas polarization of macrophages to the M1 phenotype attenuated pulmonary fibrosis. Taken together, these observations provide a novel mechanism of Cu,Zn-SOD-mediated and Th2-independent M2 polarization and provide a potential therapeutic target for attenuating the accelerated development of pulmonary fibrosis.


Asunto(s)
Activación de Macrófagos/genética , Macrófagos/metabolismo , Fibrosis Pulmonar/metabolismo , Superóxido Dismutasa/metabolismo , Adolescente , Adulto , Animales , Línea Celular , Polaridad Celular/genética , Citocinas/genética , Citocinas/metabolismo , Progresión de la Enfermedad , Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Immunoblotting , Macrófagos/clasificación , Macrófagos/enzimología , Macrófagos Alveolares/clasificación , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/genética , Células Th2/metabolismo , Adulto Joven
7.
J Biol Chem ; 287(5): 3301-12, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22157762

RESUMEN

The generation of reactive oxygen species, particularly H(2)O(2), from alveolar macrophages is causally related to the development of pulmonary fibrosis. Rac1, a small GTPase, is known to increase mitochondrial H(2)O(2) generation in macrophages; however, the mechanism by which this occurs is not known. This study shows that Rac1 is localized in the mitochondria of alveolar macrophages from asbestosis patients, and mitochondrial import requires the C-terminal cysteine of Rac1 (Cys-189), which is post-translationally modified by geranylgeranylation. Furthermore, H(2)O(2) generation mediated by mitochondrial Rac1 requires electron transfer from cytochrome c to a cysteine residue on Rac1 (Cys-178). Asbestos-exposed mice harboring a conditional deletion of Rac1 in macrophages demonstrated decreased oxidative stress and were significantly protected from developing pulmonary fibrosis. These observations demonstrate that mitochondrial import and direct electron transfer from cytochrome c to Rac1 modulates mitochondrial H(2)O(2) production in alveolar macrophages pulmonary fibrosis.


Asunto(s)
Citocromos c/metabolismo , Macrófagos Alveolares/enzimología , Proteínas Mitocondriales/metabolismo , Neuropéptidos/metabolismo , Fibrosis Pulmonar/enzimología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Adolescente , Adulto , Anciano , Animales , Amianto/toxicidad , Carcinógenos/toxicidad , Citocromos c/genética , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/genética , Electrones , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Neuropéptidos/genética , Prenilación de Proteína/efectos de los fármacos , Prenilación de Proteína/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética
8.
Biochem J ; 445(2): 229-36, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22519702

RESUMEN

Rac1, a small GTPase, regulates macrophage MMP (matrix metalloproteinase)-9 in an ERK (extracellular-signal-regulated kinase)- and SP (specificity protein)-1-dependent manner. SP-1 contains a PEST (Pro-Glu-Ser-Thr) domain that may modulate protein stability. We hypothesize that Thr578, Ser586 and/or Ser587 in the PEST domain are required for SP-1 stability and MMP-9 expression secondary to activation of ERK, a serine/threonine kinase. We determined the effects of Rac1 and ERK on MMP-9 expression driven by SP-1WT (wild-type) and the SP-1 mutants T578A, S586A and S587A. Expression of WT and mutant SP-1 increased MMP9 promoter activity in alveolar macrophages. However, constitutively active Rac1 suppressed MMP9 promoter activity in cells expressing SP-1WT, SP-1T578A and SP-1S587A, but not SP-1S586A. Furthermore, constitutive ERK activation, which was inhibited by Rac1, significantly increased MMP9 transcription in cells expressing SP-1WT, but not SP-1S586A. As Rac1 activation and ERK inactivation increased degradation of SP-1WT and not SP-1S586A, the results of the present study suggest that SP-1 stability mediated at Ser586 regulates MMP9 transcription. Ex vivo, alveolar macrophages obtained from patients with asbestosis had less MMP-9 expression that was associated with decreased SP-1 expression and ERK activation. These observations demonstrate that Ser586 in the PEST domain of SP-1 is important for MMP9 gene expression in alveolar macrophages and highlight the importance of these proteins in pulmonary fibrosis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Macrófagos Alveolares/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Serina/metabolismo , Factor de Transcripción Sp1/metabolismo , Adolescente , Adulto , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación/genética , Neuropéptidos/fisiología , Estructura Terciaria de Proteína , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina/química , Serina/genética , Transducción de Señal , Factor de Transcripción Sp1/genética , Adulto Joven , Proteínas de Unión al GTP rac/fisiología , Proteína de Unión al GTP rac1
9.
bioRxiv ; 2023 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-37398026

RESUMEN

Lymphangioleiomyomatosis (LAM) is a debilitating, progressive lung disease with few therapeutic options, largely due to a paucity of mechanistic knowledge of disease pathogenesis. Lymphatic endothelial cells (LECs) are known to envelope and invade clusters of LAM-cells, comprising of smooth muscle α-actin and/or HMB-45 positive "smooth muscle-like cells" however the role of LECs in LAM pathogenesis is still unknown. To address this critical knowledge gap, we investigated wether LECs interact with LAM-cells to augment their metastatic behaviour of LAM-cells. We performed in situ spatialomics and identified a core of transcriptomically related cells within the LAM nodules. Pathway analysis highlights wound and pulmonary healing, VEGF signaling, extracellular matrix/actin cytoskeletal regulating and the HOTAIR regulatory pathway enriched in the LAM Core cells. We developed an organoid co-culture model combining primary LAM-cells with LECs and applied this to evaluate invasion, migration, and the impact of Sorafenib, a multi-kinase inhibitor. LAM-LEC organoids had significantly higher extracellular matrix invasion, decreased solidity and a greater perimeter, reflecting increased invasion compared to non-LAM control smooth muscle cells. Sorafenib significantly inhibited this invasion in both LAM spheroids and LAM-LEC organoids compared to their respective controls. We identified TGFß1ι1, a molecular adapter coordinating protein-protein interactions at the focal adhesion complex and known to regulate VEGF, TGFß and Wnt signalling, as a Sorafenib-regulated kinase in LAM-cells. In conclusion we have developed a novel 3D co-culture LAM model and have demonstrated the effectiveness of Sorafenib to inhibit LAM-cell invasion, identifying new avenues for therapeutic intervention.

10.
J Biol Chem ; 286(17): 15597-607, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21393238

RESUMEN

The release of H(2)O(2) from alveolar macrophages has been linked to the development of pulmonary fibrosis, but little is known about its source or mechanism of production. We found that alveolar macrophages from asbestosis patients spontaneously produce high levels of H(2)O(2) and have high expression of Cu,Zn-superoxide dismutase (SOD). Because Cu,Zn-SOD is found in the mitochondrial intermembrane space (IMS), we hypothesized that mitochondrial Cu,Zn-SOD-mediated H(2)O(2) generation contributed to pulmonary fibrosis. Asbestos-induced translocation of Cu,Zn-SOD to the IMS was unique to macrophages and dependent on functional mitochondrial respiration and the presence of at least one of the conserved cysteines required for disulfide bond formation. These conserved cysteine residues were also necessary for enzyme activation and H(2)O(2) generation. Cu,Zn-SOD-mediated H(2)O(2) generation was inhibited by knockdown of the iron-sulfur protein, Rieske, in complex III. The role of Cu,Zn-SOD was biologically relevant in that Cu,Zn-SOD(-/-) mice generated significantly less H(2)O(2) and had less oxidant stress in bronchoalveolar lavage fluid and lung parenchyma. Furthermore, Cu,Zn-SOD(-/-) mice did not develop pulmonary fibrosis, and knockdown of Cu,Zn-SOD in monocytes attenuated collagen I deposition by lung fibroblasts. Our findings demonstrate a novel mechanism for the pathogenesis of pulmonary fibrosis where the antioxidant enzyme Cu,Zn-SOD translocates to the mitochondrial IMS to increase H(2)O(2) generation in alveolar macrophages.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Fibrosis Pulmonar/etiología , Superóxido Dismutasa/metabolismo , Adolescente , Adulto , Animales , Amianto/farmacología , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Mitocondriales , Estrés Oxidativo , Transporte de Proteínas , Regulación hacia Arriba , Adulto Joven
11.
J Leukoc Biol ; 112(3): 457-473, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35075692

RESUMEN

Inflammatory agents, microbial products, or stromal factors pre-activate or prime neutrophils to respond to activating stimuli in a rapid and aggressive manner. Primed neutrophils exhibit enhanced chemotaxis, phagocytosis, and respiratory burst when stimulated by secondary activating stimuli. We previously reported that Triggering Receptor Expressed on Myeloid cells-1 (TREM-1) mediates neutrophil effector functions such as increased superoxide generation, transepithelial migration, and chemotaxis. However, it is unclear whether TREM-1 is required for the process of priming itself or for primed responses to subsequent stimulation. To investigate this, we utilized in vitro and in vivo differentiated neutrophils that were primed with TNF-α and then stimulated with the particulate agonist, opsonized zymosan (OpZ). Bone marrow progenitors isolated from WT and Trem-1-/- mice were transduced with estrogen regulated Homeobox8 (ER-Hoxb8) fusion transcription factor and differentiated in vitro into neutrophils following estrogen depletion. The resulting neutrophils expressed high levels of TREM-1 and resembled mature in vivo differentiated neutrophils. The effects of priming on phagocytosis and oxidative burst were determined. Phagocytosis did not require TREM-1 and was not altered by priming. In contrast, priming significantly enhanced OpZ-induced oxygen consumption and superoxide production in WT but not Trem-1-/- neutrophils indicating that TREM-1 is required for primed oxidative burst. TREM-1-dependent effects were not mediated during the process of priming itself as priming enhanced degranulation, ICAM-1 shedding, and IL-1ß release to the same extent in WT and Trem-1-/- neutrophils. Thus, TREM-1 plays a critical role in primed phagocytic respiratory burst and mediates its effects following priming.


Asunto(s)
Estallido Respiratorio , Superóxidos , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Animales , Ratones , Neutrófilos/metabolismo , Zimosan/administración & dosificación
12.
J Biol Chem ; 285(32): 25062-73, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20529870

RESUMEN

Aberrant matrix deposition is a hallmark of pulmonary fibrosis and is characterized by an imbalance between matrix deposition and degradation. We have previously shown that mice harboring a conditional deletion of the GTP-binding protein, Rac1, in macrophages are protected from asbestos-induced pulmonary fibrosis. To investigate the contribution of aberrant matrix degradation, we addressed the role of Rac1 in regulating expression of macrophage-specific MMP-9 (matrix metalloproteinase-9). We found that MMP-9 gene transcription was significantly greater in Rac1 null macrophages. Deletion and mutational analysis of the MMP-9 promoter revealed that both SP-1 and AP-1 are essential for MMP-9 transcription. Overexpression of constitutive active Rac1 (V12) revealed that H(2)O(2) was derived from the mitochondria. Rac1-induced H(2)O(2) generation down-regulated MMP-9 gene transcription, whereas catalase overexpression in WT cells enhanced MMP-9 expression. SP-1 interacted directly with both c-Jun and c-Fos, and H(2)O(2) decreased this binding, suggesting that SP-1 and AP-1 function cooperatively to regulate MMP-9 transcription. Rac1-mediated H(2)O(2) inhibited the ERK MAPK, which was essential for activation of SP-1 and AP-1. ERK activation and MMP-9 expression were recovered by overexpressing catalase or transfecting siRNA for the mitochondrial iron-sulfur protein, Rieske. These observations were recapitulated in vivo. MMP-9 mRNA was higher in alveolar macrophages isolated from Rac1 null mice and wild type mice given catalase. Rac1 regulates MMP-9 transcription via mitochondrial H(2)O(2) generation, providing a potential mechanism by which Rac1 null mice fail to develop pulmonary fibrosis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Peróxido de Hidrógeno/química , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción AP-1/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L846-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19684199

RESUMEN

The release of reactive oxygen species (ROS) and cytokines by alveolar macrophages has been demonstrated in asbestos-induced pulmonary fibrosis, but the mechanism linking alveolar macrophages to the pathogenesis is not known. The GTPase Rac1 is a second messenger that plays an important role in host defense. In this study, we demonstrate that Rac1 null mice are protected from asbestos-induced pulmonary fibrosis, as determined by histological and biochemical analysis. We hypothesized that Rac1 induced pulmonary fibrosis via generation of ROS. Asbestos increased TNF-alpha and ROS in a Rac1-dependent manner. TNF-alpha was elevated only 1 day after exposure, whereas ROS generation progressively increased in bronchoalveolar lavage cells obtained from wild-type (WT) mice. To determine whether ROS generation contributed to pulmonary fibrosis, we overexpressed catalase in WT monocytes and observed a decrease in ROS generation in vitro. More importantly, administration of catalase to WT mice attenuated the development of fibrosis in vivo. For the first time, these results demonstrate that Rac1 plays a crucial role in asbestos-induced pulmonary fibrosis. Moreover, it suggests that a simple intervention may be useful to prevent progression of the disease.


Asunto(s)
Catalasa/farmacología , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Amianto , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Línea Celular , Movimiento Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-1beta/metabolismo , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Ratones , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína de Unión al GTP rac1/deficiencia
14.
J Leukoc Biol ; 105(6): 1195-1207, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30667543

RESUMEN

Neutrophil migration across tissue barriers to the site of injury involves integration of complex danger signals and is critical for host survival. Numerous studies demonstrate that these environmental signals fundamentally alter the responses of extravasated or "primed" neutrophils. Triggering receptor expressed on myeloid cells 1 (TREM-1) plays a central role in modulating inflammatory signaling and neutrophil migration into the alveolar airspace. Using a genetic approach, we examined the role of TREM-1 in extravasated neutrophil function. Neutrophil migration in response to chemoattractants is dependent upon multiple factors, including reactive oxygen species (ROS) generated either extracellularly by epithelial cells or intracellularly by NADPH oxidase (NOX). We, therefore, questioned whether ROS were responsible for TREM-1-mediated regulation of migration. Thioglycollate-elicited peritoneal neutrophils isolated from wild-type (WT) and TREM-1-deficient mice were stimulated with soluble and particulate agonists. Using electron paramagnetic resonance spectroscopy, we demonstrated that NOX2-dependent superoxide production is impaired in TREM-1-deficient neutrophils. Consistent with these findings, we confirmed with Clark electrode that TREM-1-deficient neutrophils consume less oxygen. Next, we demonstrated that TREM-1 deficient neutrophils have impaired directional migration to fMLP and zymosan-activated serum as compared to WT neutrophils and that deletion or inhibition of NOX2 in WT but not TREM-1-deficient neutrophils significantly impaired direction sensing. Finally, TREM-1 deficiency resulted in decreased protein kinase B (AKT) activation. Thus, TREM-1 regulates neutrophil migratory properties, in part, by promoting AKT activation and NOX2-dependent superoxide production. These findings provide the first mechanistic evidence as to how TREM-1 regulates neutrophil migration.


Asunto(s)
Quimiotaxis/inmunología , NADPH Oxidasa 2/inmunología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Superóxidos/inmunología , Receptor Activador Expresado en Células Mieloides 1/inmunología , Animales , Quimiotaxis/genética , Activación Enzimática/genética , Activación Enzimática/inmunología , Ratones , Ratones Noqueados , NADPH Oxidasa 2/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/genética , Receptor Activador Expresado en Células Mieloides 1/genética
15.
Nat Commun ; 10(1): 116, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30631065

RESUMEN

The development of multiple organ dysfunction syndrome (MODS) following infection or tissue injury is associated with increased patient morbidity and mortality. Extensive cellular injury results in the release of nuclear proteins, of which histones are the most abundant, into the circulation. Circulating histones are implicated as essential mediators of MODS. Available anti-histone therapies have failed in clinical trials due to off-target effects such as bleeding and toxicity. Here, we describe a therapeutic strategy for MODS based on the neutralization of histones by chemically stabilized nucleic acid bio-drugs (aptamers). Systematic evolution of ligands by exponential enrichment technology identified aptamers that selectively bind those histones responsible for MODS and do not bind to serum proteins. We demonstrate the efficacy of histone-specific aptamers in human cells and in a murine model of MODS. These aptamers could have a significant therapeutic benefit in the treatment of multiple diverse clinical conditions associated with MODS.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Insuficiencia Multiorgánica/metabolismo , Proteínas Nucleares/metabolismo , ARN/metabolismo , Animales , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Histonas/antagonistas & inhibidores , Histonas/genética , Histonas/metabolismo , Humanos , Ratones Endogámicos BALB C , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/prevención & control , Proteínas Nucleares/genética , Unión Proteica , ARN/antagonistas & inhibidores , ARN/genética
16.
PLoS One ; 12(10): e0186311, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29059213

RESUMEN

The multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a serine/threonine kinase important in transducing intracellular Ca2+ signals. While in vitro data regarding the role of CaMKII in the regulation of endothelial nitric oxide synthase (eNOS) are contradictory, its role in endothelial function in vivo remains unknown. Using two novel transgenic models to express CaMKII inhibitor peptides selectively in endothelium, we examined the effect of CaMKII on eNOS activation, NO production, vasomotor tone and blood pressure. Under baseline conditions, CaMKII activation was low in the aortic wall. Consistently, systolic and diastolic blood pressure, heart rate and plasma NO levels were unaltered by endothelial CaMKII inhibition. Moreover, endothelial CaMKII inhibition had no significant effect on NO-dependent vasodilation. These results were confirmed in studies of aortic rings transduced with adenovirus expressing a CaMKII inhibitor peptide. In cultured endothelial cells, bradykinin treatment produced the anticipated rapid influx of Ca2+ and transient CaMKII and eNOS activation, whereas CaMKII inhibition blocked eNOS phosphorylation on Ser-1179 and dephosphorylation at Thr-497. Ca2+/CaM binding to eNOS and resultant NO production in vitro were decreased under CaMKII inhibition. Our results demonstrate that CaMKII plays an important role in transient bradykinin-driven eNOS activation in vitro, but does not regulate NO production, vasorelaxation or blood pressure in vivo under baseline conditions.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Animales , Línea Celular , Humanos , Fosforilación
17.
Biochem J ; 377(Pt 3): 545-52, 2004 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-14604434

RESUMEN

The effect of fatty acids on LXR (liver X receptors)-mediated enhancement of ABCA1 (ATP-binding cassette transporter A1) expression and cholesterol efflux was investigated in human intestinal cells CaCo-2. LXR activation by T0901317 increased basolateral cholesterol efflux to lipoprotein particles isolated at a density of 1.21 g/ml or higher. Oleic and arachidonic acids attenuated the amount of cholesterol isolated from these particles. Stearic, linoleic and docosahexaenoic acids also decreased cholesterol efflux from basolateral membranes, with the polyunsaturated fatty acids being the most potent. Although oleic, arachidonic and docosahexaenoic acids modestly decreased ABCA1 mRNA levels in response to LXR activation, stearic and linoleic acids did not. Except for oleic acid, all fatty acids substantially attenuated an increase in ABCA1 mass secondary to LXR activation. Inhibiting acyl-CoA:cholesterol acyltransferase activity prevented the decrease in cholesterol efflux caused by oleic acid. Thus, in response to LXR activation, all fatty acids decreased the efflux of cholesterol from the basolateral membrane of CaCo-2 cells. Although modest suppression of ABCA1 gene expression by oleic, arachidonic and docosahexaenoic acids cannot be completely excluded as a mechanism, the predominant effect of fatty acids on ABCA1 expression and cholesterol efflux is at a post-transcriptional level.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/biosíntesis , Células CACO-2/química , Colesterol/metabolismo , Ácidos Grasos/farmacología , Lipoproteínas HDL/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Ácidos Araquidónicos/farmacología , Células CACO-2/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/efectos de los fármacos , HDL-Colesterol/química , HDL-Colesterol/metabolismo , Proteínas de Unión al ADN , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Receptores X del Hígado , Ácido Oléico/farmacología , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/fisiología
18.
Free Radic Biol Med ; 86: 47-56, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25958207

RESUMEN

Fibrotic remodeling in lung injury is a major cause of morbidity. The mechanism that mediates the ongoing fibrosis is unclear, and there is no available treatment to abate the aberrant repair. Reactive oxygen species (ROS) have a critical role in inducing fibrosis by modulating extracellular matrix deposition. Specifically, mitochondrial hydrogen peroxide (H2O2) production by alveolar macrophages is directly linked to pulmonary fibrosis as inhibition of mitochondrial H2O2 attenuates the fibrotic response in mice. Prior studies indicate that the small GTP-binding protein, Rac1, directly mediates H2O2 generation in the mitochondrial intermembrane space. Geranylgeranylation of the C-terminal cysteine residue (Cys(189)) is required for Rac1 activation and mitochondrial import. We hypothesized that impairment of geranylgeranylation would limit mitochondrial oxidative stress and, thus, abrogate progression of pulmonary fibrosis. By targeting the isoprenoid pathway with a novel agent, digeranyl bisphosphonate (DGBP), which impairs geranylgeranylation, we demonstrate that Rac1 mitochondrial import, mitochondrial oxidative stress, and progression of the fibrotic response to lung injury are significantly attenuated. These observations reveal that targeting the isoprenoid pathway to alter Rac1 geranylgeranylation halts the progression of pulmonary fibrosis after lung injury.


Asunto(s)
Difosfonatos/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Terpenos/farmacología , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Activación Enzimática , Humanos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones Endogámicos C57BL , Persona de Mediana Edad , Terapia Molecular Dirigida , Neuropéptidos/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Terpenos/metabolismo , Adulto Joven , Proteína de Unión al GTP rac1/metabolismo
19.
J Biol Chem ; 280(51): 41793-804, 2005 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-16221687

RESUMEN

Fatty acid biosynthesis is transcriptionally regulated by liver X receptor (LXR) and its gene target, sterol regulatory element binding protein-1c (SREBP-1c). LXR activation is induced by oxysterol end products of the mevalonate pathway and is inhibited by the upstream non-sterol isoprenoid, geranylgeranyl pyrophosphate (GGPP). Whether isoprenoids play a role in regulating the transcription of genes involved in fatty acid biosynthesis is unknown. In CaCo-2 colon epithelial cells, depletion of mevalonate and its derivatives, including oxysterol ligands for LXR, increased fatty acid synthesis. Addition of mevalonate or its isoprenoid derivative, farnesyl pyrophosphate (FPP), prevented this increase. The effects of FPP were likely due to itself or its degradation products, because none of its downstream derivatives, GGPP, ubiquinone, or cholesterol, were effective. Moreover, the effects of FPP could not be accounted for by protein prenylation, because inhibition of farnesylation did not alter fatty acid synthesis in mevalonate-depleted cells incubated with the isoprenoid. Neither was fatty acid synthesis in these cells altered by inhibition of beta-oxidation. Mevalonate depletion increased fatty acid synthase (FAS) mRNA by transcriptional mechanisms, without increasing gene expression of other enzymes involved in fatty acid biosynthesis or of SREBP-1c. The abundance of mature SREBP-2 but not SREBP-1 was increased following mevalonate depletion. FPP prevented the increase in FAS mRNA in mevalonate-depleted cells without altering SREBP-2 activation. Thus, FPP regulates fatty acid synthesis by a mechanism that is likely independent of the SREBP pathway.


Asunto(s)
Ácidos Grasos/biosíntesis , Fosfatos de Poliisoprenilo/farmacología , Secuencia de Bases , Western Blotting , Células CACO-2 , Cartilla de ADN , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Humanos , Ácido Mevalónico/metabolismo , Oxidación-Reducción , Ácido Palmítico/metabolismo , ARN Mensajero/genética , Sesquiterpenos
20.
Biochem J ; 368(Pt 3): 855-64, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12213084

RESUMEN

Regulation of sterol regulatory element-binding proteins (SREBPs) by fatty acid flux was investigated in CaCo-2 cells. Cells were incubated with 1 mM taurocholate with or without 250 microM 18:0, 18:1, 18:2, 20:4, 20:5 or 22:6 fatty acids. Fatty acid synthase (FAS) and acetyl-CoA carboxylase mRNA levels and gene and protein expression of SREBPs were estimated. 18:2, 20:4, 20:5 and 22:6 fatty acids decreased the amount of mature SREBP-1 and mRNA levels of SREBP-1c, SREBP-1a, FAS and acetyl-CoA carboxylase. SREBP-2 gene or mature protein expression was not altered. Liver X receptor (LXR) activation by T0901317 increased gene expression of SREBP-1c, SREBP-1a, FAS and acetyl-CoA carboxylase without altering SREBP-2. 20:5, but not 18:1, prevented the full expression of SREBP-1c mRNA by T0901317. T0901317 increased SREBP-1 mass without altering the mass of mature SREBP-2. Although only 18:2, 20:4, 20:5 and 22:6 suppressed SREBP-1, acetyl-CoA carboxylase and FAS expression, all fatty acids decreased the rate of fatty acid synthesis. T0901317 increased endogenous fatty acid synthesis yet did not increase secretion of triacylglycerol-rich lipoproteins. In CaCo-2 cells, polyunsaturated fatty acids decrease gene and protein expression of SREBP-1 and FAS mRNA, probably through interference with LXR activity. Since all fatty acids decreased fatty acid synthesis, mechanisms other than changes in SREBP-1c expression must be entertained. Increased endogenous fatty acid synthesis does not promote triacylglycerol-rich lipoprotein secretion.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Ácido Graso Sintasas/biosíntesis , Ácidos Grasos Insaturados/metabolismo , Expresión Génica , Factores de Transcripción , Triglicéridos/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Anticolesterolemiantes/farmacología , Apolipoproteínas B/metabolismo , Células CACO-2 , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Eicosapentaenoico/farmacología , Ácidos Grasos/metabolismo , Humanos , Hidrocarburos Fluorados , Immunoblotting , Plásmidos/metabolismo , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasas/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Sulfonamidas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA