Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 496
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(1): 67-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25419628

RESUMEN

Immune responses are tightly regulated to ensure efficient pathogen clearance while avoiding tissue damage. Here we report that Setdb2 was the only protein lysine methyltransferase induced during infection with influenza virus. Setdb2 expression depended on signaling via type I interferons, and Setdb2 repressed expression of the gene encoding the neutrophil attractant CXCL1 and other genes that are targets of the transcription factor NF-κB. This coincided with occupancy by Setdb2 at the Cxcl1 promoter, which in the absence of Setdb2 displayed diminished trimethylation of histone H3 Lys9 (H3K9me3). Mice with a hypomorphic gene-trap construct of Setdb2 exhibited increased infiltration of neutrophils during sterile lung inflammation and were less sensitive to bacterial superinfection after infection with influenza virus. This suggested that a Setdb2-mediated regulatory crosstalk between the type I interferons and NF-κB pathways represents an important mechanism for virus-induced susceptibility to bacterial superinfection.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/inmunología , FN-kappa B/inmunología , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Neumonía/inmunología , Sobreinfección/inmunología , Animales , Quimiocina CXCL1/inmunología , Susceptibilidad a Enfermedades , Femenino , Interferón Tipo I/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/virología , Neumonía/enzimología , Neumonía/virología , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Organismos Libres de Patógenos Específicos , Sobreinfección/enzimología , Sobreinfección/microbiología
2.
Proc Natl Acad Sci U S A ; 119(24): e2201707119, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35671428

RESUMEN

A number of inflammatory lung diseases, including chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and pneumonia, are modulated by WNT/ß-catenin signaling. However, the underlying molecular mechanisms remain unclear. Here, starting with a forward genetic screen in mouse, we identify the WNT coreceptor Related to receptor tyrosine kinase (RYK) acting in mesenchymal tissues as a cell survival and antiinflammatory modulator. Ryk mutant mice exhibit lung hypoplasia and inflammation as well as alveolar simplification due to defective secondary septation, and deletion of Ryk specifically in mesenchymal cells also leads to these phenotypes. By analyzing the transcriptome of wild-type and mutant lungs, we observed the up-regulation of proapoptotic and inflammatory genes whose expression can be repressed by WNT/RYK signaling in vitro. Moreover, mesenchymal Ryk deletion at postnatal and adult stages can also lead to lung inflammation, thus indicating a continued role for WNT/RYK signaling in homeostasis. Our results indicate that RYK signaling through ß-catenin and Nuclear Factor kappa B (NF-κB) is part of a safeguard mechanism against mesenchymal cell death, excessive inflammatory cytokine production, and inflammatory cell recruitment and accumulation. Notably, RYK expression is down-regulated in the stromal cells of pneumonitis patient lungs. Altogether, our data reveal that RYK signaling plays critical roles as an antiinflammatory modulator during lung development and homeostasis and provide an animal model to further investigate the etiology of, and therapeutic approaches to, inflammatory lung diseases.


Asunto(s)
Neumonía , Proteínas Tirosina Quinasas Receptoras , Vía de Señalización Wnt , beta Catenina , Animales , Humanos , Pulmón/enzimología , Pulmón/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , FN-kappa B/metabolismo , Neumonía/enzimología , Neumonía/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células del Estroma/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791530

RESUMEN

Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.


Asunto(s)
Neutrófilos , Neumonía , Humanos , Neutrófilos/metabolismo , Neutrófilos/enzimología , Neutrófilos/inmunología , Animales , Neumonía/metabolismo , Neumonía/enzimología , Neumonía/patología , Serina Proteasas/metabolismo , Péptido Hidrolasas/metabolismo
4.
IUBMB Life ; 74(2): 131-142, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34590785

RESUMEN

Chronic respiratory diseases (CRDs), including pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), lung cancer, and asthma, are significant global health problems due to their prevalence and rising incidence. The roles of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) in controlling tyrosine phosphorylation of targeting proteins modulate multiple physiological cellular responses and contribute to the pathogenesis of CRDs. Src homology-2 domain-containing PTP2 (SHP2) plays a pivotal role in modulating downstream growth factor receptor signaling and cytoplasmic PTKs, including MAPK/ERK, PI3K/AKT, and JAK/STAT pathways, to regulate cell survival and proliferation. In addition, SHP2 mutation and activation are commonly implicated in tumorigenesis. However, little is known about SHP2 in chronic pulmonary inflammation and fibrosis. This review discusses the potential involvement of SHP2 deregulation in chronic pulmonary inflammation and fibrosis, as well as the therapeutic effects of targeting SHP2 in CRDs.


Asunto(s)
Neumonía , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Enfermedades Respiratorias , Fibrosis , Humanos , Neumonía/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Enfermedades Respiratorias/epidemiología , Transducción de Señal
5.
Am J Physiol Regul Integr Comp Physiol ; 322(2): R126-R135, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34984926

RESUMEN

Many lung diseases are caused by an excessive inflammatory response, and inflammatory lung diseases are often modeled using lipopolysaccharide (LPS) in mice. Cyclooxygenase-2 (COX-2) encoded by the Ptgs2 gene is induced in response to inflammatory stimuli including LPS. The objective of this study was to test the hypothesis that mice deficient in COX-2 (Ptgs2-/-) will be protected from LPS-induced lung injury. Wild-type (WT; CD1 mice) and Ptgs2-/- mice (on a CD1 background) were treated with LPS or vehicle for 24 h. LPS treatment resulted in histological evidence of lung injury, which was attenuated in the Ptgs2-/- mice. LPS treatment increased the mRNA levels for tumor necrosis factor-α, interleukin-10, and monocyte chemoattractant protein-1 in the lungs of WT mice, and the LPS-induced increases in these levels were attenuated in the Ptgs2-/- mice. The protein levels of active caspase-3 and caspase-9 were lower in the LPS-treated lungs of Ptgs2-/- mice than in LPS-treated WT mice, as were the number of terminal deoxynucleotide transferase dUTP nick end labeling-positive cells in lung sections. LPS exposure resulted in a greater lung wet-to-dry weight ratio (W/D) in WT mice, suggestive of pulmonary edema, while in LPS-treated Ptgs2-/- mice, the W/D was not different from controls and less than in LPS-treated WT mice. These results demonstrate that COX-2 is involved in the inflammatory response to LPS and suggest that COX-2 not only acts as a downstream participant in the inflammatory response, but also acts as a regulator of the inflammatory response likely through a feed-forward mechanism following LPS stimulation.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Apoptosis , Ciclooxigenasa 2/deficiencia , Pulmón/enzimología , Neumonía/prevención & control , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclooxigenasa 2/genética , Modelos Animales de Enfermedad , Femenino , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolisacáridos , Pulmón/patología , Masculino , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
FASEB J ; 35(7): e21519, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34137477

RESUMEN

Globally, COPD remains a major cause of disability and death. In the United States alone, it is estimated that approximately 14 million people suffer from the disease. Given the high disease burden and requirement for chronic, long-term medical care associated with COPD, it is essential that new disease modifying agents are developed to complement the symptomatic therapeutics currently available. In the present report, we have identified a potentially novel therapeutic agent through the use of a high throughput screen based on the knowledge that cigarette smoke induces the proteolytic enzyme MMP1 leading to destruction of the lung in COPD. A construct utilizing the cigarette responsive promoter element of MMP-1 was conjugated to a luciferase reporter and utilized in an in vitro assay to screen the NIH Molecular Libraries Small Molecule Repository to identify putative targets that suppressed luciferase expression in response to cigarette smoke extract (CSE). Selective serotonin reuptake inhibitors potently inhibited luciferase expression and were further validated. SSRI treatment suppressed MMP-1 production in small airway epithelial cells exposed to (CSE) in vitro as well as in smoke exposed rabbits. In addition, SSRI treatment inhibited inflammatory cytokine production while rescuing cigarette smoke induced downregulation in vivo of the anti-inflammatory lipid transporter ABCA1, previously shown by our laboratory to be lung protective. Importantly, SSRI treatment prevented lung destruction in smoke exposed rabbits as measured by morphometry. These studies support further investigation into SSRIs as a novel therapeutic for COPD may be warranted.


Asunto(s)
Fumar Cigarrillos/efectos adversos , Células Epiteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , Metaloproteinasa 1 de la Matriz/química , Neumonía/tratamiento farmacológico , Enfisema Pulmonar/tratamiento farmacológico , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Pulmón/metabolismo , Pulmón/patología , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/enzimología , Enfisema Pulmonar/patología , Conejos , Serotonina/metabolismo
7.
Am J Respir Cell Mol Biol ; 65(6): 646-657, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34251297

RESUMEN

Compromised endothelial-cell (EC) barrier function is a hallmark of inflammatory diseases. mTOR inhibitors, widely applied as clinical therapies, cause pneumonitis through mechanisms that are not yet fully understood. This study aimed to elucidate the EC mechanisms underlying the pathogenesis of pneumonitis caused by mTOR inhibition (mTORi). Mice with EC-specific deletion of mTOR complex components (Mtor, Rptor or Rictor) were administered LPS to induce pulmonary injury. Cultured ECs were treated with pharmacologic inhibitors, siRNA, or overexpression plasmids. EC barrier function was evaluated in vivo with Evans blue assay and in vitro by measurement of transendothelial electrical resistance and albumin flux. mTORi increased basal and TNFα-induced EC permeability, which was caused by myosin light chain (MLC) phosphorylation-dependent cell contraction. Inactivation of mTOR kinase activity by mTORi triggered PKCδ/p38/NF-κB signaling that significantly upregulated TNFα-induced MLCK (MLC kinase) expression, whereas Raptor promoted the phosphorylation of PKCα/MYPT1 independently of its interaction with mTOR, leading to suppression of MLCP (MLC phosphatase) activity. EC-specific deficiency in mTOR, Raptor or Rictor aggravated lung inflammation in LPS-treated mice. These findings reveal that mTORi induces PKC-dependent endothelial MLC phosphorylation, contraction, and hyperpermeability that promote pneumonitis.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidores mTOR/efectos adversos , Neumonía/enzimología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Humanos , Lipopolisacáridos/toxicidad , Inhibidores mTOR/farmacología , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Permeabilidad , Fosforilación/efectos de los fármacos , Neumonía/inducido químicamente , Serina-Treonina Quinasas TOR/metabolismo
8.
Am J Respir Cell Mol Biol ; 64(5): 629-640, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33662226

RESUMEN

Deficiency of ASM (acid sphingomyelinase) causes the lysosomal storage Niemann-Pick disease (NPD). Patients with NPD type B may develop progressive interstitial lung disease with frequent respiratory infections. Although several investigations using the ASM-deficient (ASMKO) mouse NPD model revealed inflammation and foamy macrophages, there is little insight into the pathogenesis of NPD-associated lung disease. Using ASMKO mice, we report that ASM deficiency is associated with a complex inflammatory phenotype characterized by marked accumulation of monocyte-derived CD11b+ macrophages and expansion of airspace/alveolar CD11c+ CD11b- macrophages, both with increased size, granularity, and foaminess. Both the alternative and classical pathways were activated, with decreased in situ phagocytosis of opsonized (Fc-coated) targets, preserved clearance of apoptotic cells (efferocytosis), secretion of Th2 cytokines, increased CD11c+/CD11b+ cells, and more than a twofold increase in lung and plasma proinflammatory cytokines. Macrophages, neutrophils, eosinophils, and noninflammatory lung cells of ASMKO lungs also exhibited marked accumulation of chitinase-like protein Ym1/2, which formed large eosinophilic polygonal Charcot-Leyden-like crystals. In addition to providing insight into novel features of lung inflammation that may be associated with NPD, our report provides a novel connection between ASM and the development of crystal-associated lung inflammation with alterations in macrophage biology.


Asunto(s)
Glicoproteínas/inmunología , Lisofosfolipasa/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Enfermedad de Niemann-Pick Tipo A/inmunología , Enfermedad de Niemann-Pick Tipo B/inmunología , Neumonía/inmunología , Esfingomielina Fosfodiesterasa/inmunología , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Tamaño de la Célula , Quitinasas/genética , Quitinasas/inmunología , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Eosinófilos/patología , Femenino , Expresión Génica , Glicoproteínas/genética , Humanos , Lectinas/genética , Lectinas/inmunología , Pulmón/inmunología , Pulmón/patología , Lisofosfolipasa/genética , Macrófagos/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Enfermedad de Niemann-Pick Tipo A/enzimología , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo B/enzimología , Enfermedad de Niemann-Pick Tipo B/genética , Enfermedad de Niemann-Pick Tipo B/patología , Fagocitosis , Neumonía/enzimología , Neumonía/genética , Neumonía/patología , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Balance Th1 - Th2/genética , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología
9.
Toxicol Appl Pharmacol ; 432: 115757, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34673086

RESUMEN

An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1ß, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-ß1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-ß1/SMAD pathways.


Asunto(s)
Proteína HMGB1/metabolismo , Pulmón/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Neumonía/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/enzimología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Arsenitos , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Pulmón/ultraestructura , Masculino , Fosforilación , Neumonía/inducido químicamente , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas Wistar , Transducción de Señal , Compuestos de Sodio
10.
Respir Res ; 22(1): 267, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34666758

RESUMEN

BACKGROUND: The non-cancerous functions of Akt in the airway are understudied. In some tissues, Akt phosphorylates and activates endothelial nitric oxide synthase (eNOS) to produce nitric oxide (NO) that has anti-inflammatory effects. NO production has antibacterial and antiviral effects in the airway, and increasing NO may be a useful anti-pathogen strategy. Akt also stimulates the nuclear factor erythroid 2-related factor 2 (Nrf-2) transcription factor, which transcribes antioxidant genes. Therefore, we hypothesized that activation of the Akt/eNOS pathway, which also activates Nrf-2, may have protective effects in human airway cells against injury. METHODS: To directly test the effects of Akt signaling in the airway, we treated A549 and 16HBE cells as well as primary bronchial, nasal, and type II alveolar epithelial cells with small molecule Akt activator SC79. We examined the effects of SC79 on eNOS activation, NO production, Nrf-2 target levels, and interleukin-8 (IL-8) transcription during exposure to TNF-α or Pseudomonas flagellin (TLR5 agonist). Additionally, air-liquid interface bronchial cultures were treated with cadmium, an oxidative stressor that causes airway barrier breakdown. RESULTS: SC79 induced a ~ twofold induction of p-eNOS and Nrf-2 protein levels blocked by PI3K inhibitor LY294002. Live cell imaging revealed SC79 increased acute NO production. Quantitative RT-PCR showed a ~ twofold increase in Nrf-2 target gene transcription. TNF-α or flagellin-induced IL-8 levels were also significantly reduced with SC79 treatment. Moreover, the transepithelial electrical resistance decrease observed with cadmium was ameliorated by SC79, likely by an acute increase in tight junction protein ZO-1 levels. CONCLUSIONS: Together, the data presented here demonstrate SC79 activation of Akt induces potentially anti-pathogenic NO production, antioxidant gene transcription, reduces IL-8 transcription, and may protect against oxidative barrier dysfunction in a wide range of airway epithelial cells.


Asunto(s)
Acetatos/farmacología , Antiinflamatorios/farmacología , Benzopiranos/farmacología , Activadores de Enzimas/farmacología , Células Epiteliales/efectos de los fármacos , Interleucina-8/metabolismo , Pulmón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/metabolismo , Neumonía/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células A549 , Impedancia Eléctrica , Activación Enzimática , Células Epiteliales/enzimología , Células Epiteliales/inmunología , Humanos , Interleucina-8/genética , Pulmón/enzimología , Pulmón/inmunología , Factor 2 Relacionado con NF-E2/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Neumonía/enzimología , Neumonía/genética , Neumonía/inmunología , Transducción de Señal , Transcripción Genética , Proteína de la Zonula Occludens-1/metabolismo
11.
J Biochem Mol Toxicol ; 35(12): e22923, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34590760

RESUMEN

Amiodarone (AMD), an antiarrhythmic drug, is used cautiously due to its lung toxicity that is characterized by alveolar inflammation followed by fatal fibrosis. AMD induces lung inflammation via increasing the alveolar macrophages and disturbing the balance of T-helper-1 (Th1) and Th2 cells cytokines. In this study, the role of the mitogen-activated protein kinases (MAPKs)/activator protein-1 (AP-1) pathway in AMD-induced lung inflammation was evaluated. Also, the anti-inflammatory and antifibrotic effects of losartan and/or vitamin D were investigated following 7, 14, and 28 days of AMD administration. AMD resulted in lung injury, inflammatory infiltration, and increased pulmonary levels of inflammatory cytokines starting from Week 1 of exposure. A significant increase in serum levels of interleukin-4 along with a significant reduction of interferon-gamma, in addition to strong expression of CD68, were reported after 14 and 28 days of AMD administration reflecting Th1/Th2 cytokines imbalance and the accumulation of alveolar macrophages, respectively. The phosphorylation of MAPKs (ERK1/2, JNK, p38) and AP-1 was significantly enhanced starting from Week 1 of exposure. Marked expression of transforming growth factor beta-1 and massive deposition of collagen were detected after 28 days reflecting late fibrosis. All these abnormalities were significantly mitigated by vitamin D and its combination with losartan. Losartan alone has less prominent anti-inflammatory effects particularly after 28 days; however, it efficiently prevented late fibrosis. This study concludes that MAPKs/AP-1 pathway is involved in AMD-induced lung inflammation and that vitamin D and/or losartan could be used as a prophylactic agent to prevent AMD-induced lung toxicity.


Asunto(s)
Amiodarona/toxicidad , Antiarrítmicos/toxicidad , Losartán/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neumonía/inducido químicamente , Neumonía/prevención & control , Factor de Transcripción AP-1/metabolismo , Animales , Antiarrítmicos/farmacología , Interferón gamma/sangre , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Neumonía/enzimología , Ratas , Ratas Wistar , Vitamina D/farmacología
12.
Int J Mol Sci ; 22(11)2021 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-34073678

RESUMEN

The heme molecule serves as an essential prosthetic group for oxygen transport and storage proteins, as well for cellular metabolic enzyme activities, including those involved in mitochondrial respiration, xenobiotic metabolism, and antioxidant responses. Dysfunction in both heme synthesis and degradation pathways can promote human disease. Heme is a pro-oxidant via iron catalysis that can induce cytotoxicity and injury to the vascular endothelium. Additionally, heme can modulate inflammatory and immune system functions. Thus, the synthesis, utilization and turnover of heme are by necessity tightly regulated. The microsomal heme oxygenase (HO) system degrades heme to carbon monoxide (CO), iron, and biliverdin-IXα, that latter which is converted to bilirubin-IXα by biliverdin reductase. Heme degradation by heme oxygenase-1 (HO-1) is linked to cytoprotection via heme removal, as well as by activity-dependent end-product generation (i.e., bile pigments and CO), and other potential mechanisms. Therapeutic strategies targeting the heme/HO-1 pathway, including therapeutic modulation of heme levels, elevation (or inhibition) of HO-1 protein and activity, and application of CO donor compounds or gas show potential in inflammatory conditions including sepsis and pulmonary diseases.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hemo/metabolismo , Inflamación/metabolismo , Neumonía/metabolismo , Animales , Humanos , Inflamación/enzimología , Inflamación/etiología , Neumonía/enzimología , Neumonía/etiología , Sepsis/etiología , Sepsis/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo
13.
Pediatr Res ; 87(3): 494-500, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31578032

RESUMEN

BACKGROUND: Intrauterine inflammation affects fetal lung development. BTB and CNC homology 1 (Bach1) is a transcriptional repressor of heme oxygenase-1 (HO-1) and interleukin-6 (IL-6) genes. We investigated the role of Bach1 in the development of fetal mouse lungs exposed to lipopolysaccharide (LPS) using a whole fetal lung tissue culture system. METHODS: We isolated and cultured embryonic day 12.5 fetal mouse lungs from pregnant Bach1 knockout (-/-) and wild-type (WT) mice. Airway branching morphogenesis was assessed by microscopically counting peripheral lung buds after incubation with/without LPS. Expression levels of genes related to inflammation and oxidative stress were evaluated using quantitative PCR. Zinc protoporphyrin, HO-1-specific inhibitor, was used. RESULTS: Branching morphogenesis was observed in Bach1-/- and WT fetal mice lungs without LPS exposure; after exposure to LPS, the number of peripheral lung buds was suppressed in Bach1-/- group only. Basal messenger RNA (mRNA) and protein expression of HO-1 was significantly higher in Bach1-/- group than in WT group; IL-6 and monocyte chemoattractant protein-1 mRNA expression was significantly increased after LPS exposure in both groups. Zinc protoporphyrin mitigated the LPS-induced suppression of branching morphogenesis in Bach1-/- mice. CONCLUSION: The ablation of Bach1 suppresses airway branching morphogenesis after LPS exposure by increased basal expression levels of HO-1.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Hemo-Oxigenasa 1/metabolismo , Pulmón/enzimología , Proteínas de la Membrana/metabolismo , Neumonía/enzimología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Hemo-Oxigenasa 1/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/embriología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Técnicas de Cultivo de Órganos , Neumonía/embriología , Embarazo , Transducción de Señal , Regulación hacia Arriba
14.
Med Sci Monit ; 26: e924482, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32844782

RESUMEN

BACKGROUND Acute pneumonia is a severe inflammatory disease of the respiratory system. Drugs used to treat acute pneumonia often have strong side effects. Recent studies have shown that tetrahydropyrimidines, ZL-5015 has anti-inflammatory and antitumor effects. However, whether ZL-5015 can relieve symptoms of acute pneumonia is unclear. MATERIAL AND METHODS In this study, we used lipo-polysaccharide (LPS) to stimulate SD rats to simulate conditions of acute pneumonia. Diverse doses of ZL-5015 were used for treatment of these rats. After the rates were sacrificed, serum, lung tissue, and bronchoalveolar lavage fluid were collected for the next study. Hematoxylin-eosin (H&E) staining then was used to detect pathologic changes in lung tissues. Enzyme-linked immunosorbent assay was performed to assess levels of inflammatory factors in serum. Commercial kits were used to assess levels of reactive oxygen species (ROS) in bronchoalveolar lavage fluid. RESULTS Treatment of ZL-5015 relieved stenosis of the alveolar space and pulmonary edema. Furthermore, levels of inflammatory factors (TNF-alpha, IL-1ß and IL-18) in the lung tissues and serum were downregulated after treatment with ZL-5015. Production of ROS also was suppressed after application of ZL-5015. Moreover, inhibition of expression of NRF-2 and HO-1 was relieved after treatment with ZL-5015. The therapeutic effect of ZL-5015 showed a dose-response relationship. CONCLUSIONS ZL-5015 alleviated LPS-induced inflammatory injury and oxidative damage by activating the NRF-2/HO-1 pathway.


Asunto(s)
Ésteres/farmacología , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Neumonía/prevención & control , Pirimidinas/farmacología , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Neumonía/enzimología , Neumonía/metabolismo , Ratas
15.
J Stroke Cerebrovasc Dis ; 29(9): 104942, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32807413

RESUMEN

BACKGROUND AND OBJECTIVES: Studies implicate the lung in moderating systemic immune activation via effects on circulating leukocytes. In this study, we investigated whether targeted expression of the antioxidant extracellular superoxide dismutase (SOD3) within the lung would influence post-ischemic peripheral neutrophil activation and CNS reperfusion injury. METHODS: Adult, male mice expressing human SOD3 within type II pneumocytes were subjected to 15 min of transient global cerebral ischemia. Three days post-reperfusion, lung and brain tissue was collected and analyzed by immunohistochemistry for inflammation and injury markers. In vitro motility and neurotoxicity assays were conducted to ascertain the direct effects of hSOD3 on PMN activation. Results were compared against C57BL/6 age and sex-matched controls. RESULTS: Relative to wild-type controls, hSOD3 heterozygous mice exhibited a reduction in lung inflammation, blood-brain barrier damage, and post-ischemic neuronal injury within the hippocampus and cortex. PMNs harvested from hSOD3 mice were also resistant to LPS priming, slower-moving, and less toxic to primary neuronal cultures. CONCLUSIONS: Constitutive, focal expression of hSOD3 is neuroprotective in a model of global cerebral ischemia-reperfusion injury. The underlying mechanism of SOD3-dependent protection is attributable in part to effects on the activation state and toxic potential of circulating neutrophils. These results implicate lung-brain coupling as a determinant of cerebral ischemia-reperfusion injury and highlight post-stroke lung inflammation as a potential therapeutic target in acute ischemic cerebrovascular injuries.


Asunto(s)
Células Epiteliales Alveolares/enzimología , Isquemia Encefálica/enzimología , Encéfalo/metabolismo , Neuronas/metabolismo , Activación Neutrófila , Neutrófilos/metabolismo , Neumonía/prevención & control , Daño por Reperfusión/prevención & control , Superóxido Dismutasa/metabolismo , Células Epiteliales Alveolares/patología , Animales , Encéfalo/patología , Isquemia Encefálica/genética , Isquemia Encefálica/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología , Neutrófilos/inmunología , Neumonía/enzimología , Neumonía/genética , Neumonía/inmunología , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Transducción de Señal , Superóxido Dismutasa/genética
16.
Genes Dev ; 25(23): 2480-8, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22156208

RESUMEN

Macrophages, a key cellular component of inflammation, become functionally polarized in a signal- and context-specific manner. Th2 cytokines such as interleukin 4 (IL-4) polarize macrophages to a state of alternative activation that limits inflammation and promotes wound healing. Alternative activation is mediated by a transcriptional program that is influenced by epigenomic modifications, including histone acetylation. Here we report that macrophages lacking histone deacetylase 3 (HDAC3) display a polarization phenotype similar to IL-4-induced alternative activation and, furthermore, are hyperresponsive to IL-4 stimulation. Throughout the macrophage genome, HDAC3 deacetylates histone tails at regulatory regions, leading to repression of many IL-4-regulated genes characteristic of alternative activation. Following exposure to Schistosoma mansoni eggs, a model of Th2 cytokine-mediated disease that is limited by alternative activation, pulmonary inflammation was ameliorated in mice lacking HDAC3 in macrophages. Thus, HDAC3 functions in alternative activation as a brake whose release could be of benefit in the treatment of multiple inflammatory diseases.


Asunto(s)
Epigénesis Genética , Histona Desacetilasas/genética , Activación de Macrófagos/genética , Macrófagos/metabolismo , Animales , Histona Desacetilasas/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos , Neumonía/enzimología , Neumonía/inmunología , Neumonía/parasitología , Schistosoma mansoni , Células Th2/inmunología , Células Th2/metabolismo
17.
Biochem Biophys Res Commun ; 505(2): 498-504, 2018 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-30268495

RESUMEN

BACKGROUND: Insulin resistance (IR), a common co-morbidity of chronic obstructive pulmonary disease (COPD), aggravates airway inflammation in COPD patients, but its mechanism is unclear. Sfrp5, a novel anti-inflammatory adipocytokine, inhibits macrophage-mediated inflammation of adipose tissue and abrogates IR. However, few studies have been conducted on the regulatory role of Sfrp5 in lung inflammation. METHODS: In the present study, 30 SD rats were divided into two groups: the normal food (NF) group and the high-fat diet (HFD) group. Oral glucose tolerance test (OGTT) and insulin release test were performed to assess whether a successful IR rat model was established. The expression of Sfrp5 and key downstream moleculars of Wnt5a/JNKl signaling was detected. Lung tissue pathomorphology and macrophage activation were observed. In addition, we counted the number of inflammatory cells and measured inflammatory cytokines in bronchoalveolar lavage fluid (BALF). In vitro, rat lung macrophages were isolated and treated with Wnt5a, Sfrp5, and/or JNK inhibitor SP600125. JNK activity and inflammatory cytokines expression were examined. RESULTS: We found that in a rat model of IR, Sfrp5 expression of lung tissue was downregulated, while the Wnt5a/JNKl pathway was activated and the lung inflammatory response was enhanced. Meanwhile, Sfrp5 significantly suppressed Wnt5a/JNKl-induced macrophage activation. CONCLUSIONS: Collectively, IR reduces Sfrp5 expression of lung tissue and activates the Wnt5a/JNK1 pathway, promoting macrophage activation and contributing to the lung's inflammatory response. In contrast, Sfrp5 suppresses the inflammatory response by inhibiting the Wnt5a/JNKl pathway, which could be a target of treatment of COPD.


Asunto(s)
Adipoquinas/metabolismo , Resistencia a la Insulina , Macrófagos/metabolismo , Neumonía/metabolismo , Transducción de Señal , Animales , Regulación hacia Abajo , Activación de Macrófagos , Masculino , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Neumonía/enzimología , Neumonía/patología , Ratas Sprague-Dawley , Proteína Wnt-5a/metabolismo
18.
Am J Pathol ; 187(6): 1288-1300, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28399390

RESUMEN

Chronic obstructive pulmonary disease (COPD) comprises chronic bronchitis and emphysema, and is a leading cause of morbidity and mortality. Because tissue destruction is the prominent characteristic of emphysema, extracellular proteinases, particularly those with elastolytic ability, are often considered to be key drivers in this disease. Several human and mouse studies have implicated roles for matrix metalloproteinases (MMPs), particularly macrophage-derived proteinases, in COPD pathogenesis. MMP-28 is expressed by the pulmonary epithelium and macrophage, and we have found that it regulates macrophage recruitment and polarization. We hypothesized that MMP-28 has contributory roles in emphysema via alteration of macrophage numbers and activation. Because of the established association of emphysema pathogenesis to macrophage influx, we evaluated the inflammatory changes and lung histology of Mmp28-/- mice exposed to 3 and 6 months of cigarette smoke. At earlier time points, we found altered macrophage polarization in the smoke-exposed Mmp28-/- lung consistent with other published findings that MMP-28 regulates macrophage activation. At both 3 and 6 months, Mmp28-/- mice had blunted inflammatory responses more closely resembling nonsmoked mice, with a reduction in neutrophil recruitment and CXCL1 chemokine expression. By 6 months, Mmp28-/- mice were protected from emphysema. These results highlight a previously unrecognized role for MMP-28 in promoting chronic lung inflammation and tissue remodeling induced by cigarette smoke and highlight another potential target to modulate COPD.


Asunto(s)
Metaloproteinasas de la Matriz Secretadas/fisiología , Enfisema Pulmonar/enzimología , Animales , Líquido del Lavado Bronquioalveolar/citología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Enzimológica de la Expresión Génica/fisiología , Pulmón/enzimología , Macrófagos Alveolares/enzimología , Masculino , Metaloproteinasas de la Matriz Secretadas/deficiencia , Metaloproteinasas de la Matriz Secretadas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/fisiología , Neumonía/enzimología , Neumonía/etiología , Neumonía/genética , Neumonía/patología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Enfisema Pulmonar/etiología , Enfisema Pulmonar/genética , Enfisema Pulmonar/patología , Contaminación por Humo de Tabaco/efectos adversos
19.
J Surg Res ; 224: 200-206, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29506841

RESUMEN

BACKGROUND: Pulmonary complications after esophagectomy are often fatal. The prediction of postoperative pulmonary complications remains a challenge. Accumulating evidence demonstrates a physiological and pathological role for angiotensin-converting enzyme 2 (ACE2) in the respiratory system. The purpose of this study was to evaluate the predictive value of ACE2 levels for the development of postoperative pneumonia. METHODS: To evaluate the association between serum ACE2 levels and pneumonia after esophagectomy, we retrospectively reviewed the medical records of 80 patients who underwent thoracoscopic esophagectomy for esophageal cancer from 2009 to 2014. RESULTS: Nineteen patients (23.8%) developed pneumonia after esophagectomy. Patients with pneumonia had significantly higher levels of ACE2 from the preoperative day to postoperative day (POD) 3, white blood cell count (POD7), and C-reactive protein (POD3, POD5, and POD7) than patients without pneumonia. Patients with postoperative pneumonia had higher serum ACE2 levels on POD3 than patients without pneumonia. CONCLUSIONS: The elevation of ACE2 levels on POD3 may predict the incidence of pneumonia.


Asunto(s)
Neoplasias Esofágicas/cirugía , Esofagectomía/efectos adversos , Peptidil-Dipeptidasa A/sangre , Neumonía/enzimología , Complicaciones Posoperatorias/enzimología , Anciano , Enzima Convertidora de Angiotensina 2 , Proteína C-Reactiva/análisis , Neoplasias Esofágicas/enzimología , Femenino , Humanos , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
20.
Am J Respir Cell Mol Biol ; 56(3): 362-371, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27805412

RESUMEN

Loss of extracellular superoxide dismutase 3 (SOD3) contributes to inflammatory and fibrotic lung diseases. The human SOD3 R213G polymorphism decreases matrix binding, redistributing SOD3 from the lung to extracellular fluids, and protects against LPS-induced alveolar inflammation. We used R213G mice expressing a naturally occurring single-nucleotide polymorphism, rs1799895, within the heparin-binding domain of SOD3, which results in an amino acid substitution at position 213 to test the hypothesis that the redistribution of SOD3 into the extracellular fluids would impart protection against bleomycin-induced lung fibrosis and secondary pulmonary hypertension (PH). In R213G mice, SOD3 content and activity was increased in extracellular fluids and decreased in lung at baseline, with greater increases in bronchoalveolar lavage fluid (BALF) SOD3 compared with wild-type mice 3 days after bleomycin. R213G mice developed less fibrosis based on pulmonary mechanics, fibrosis scoring, collagen quantification, and gene expression at 21 days, and less PH by right ventricular systolic pressure and pulmonary arteriole medial wall thickening at 28 days. In wild-type mice, macrophages, lymphocytes, neutrophils, proinflammatory cytokines, and protein increased in BALF on Day 7 and/or 21. In R213G mice, total BALF cell counts increased on Day 7 but resolved by 21 days. At 1 or 3 days, BALF pro- and antiinflammatory cytokines and BALF protein were higher in R213G mice, resolving by 21 days. We conclude that the redistribution of SOD3 as a result of the R213G single-nucleotide polymorphism protects mice from bleomycin-induced fibrosis and secondary PH by improved resolution of alveolar inflammation.


Asunto(s)
Neumonía/complicaciones , Neumonía/genética , Polimorfismo de Nucleótido Simple/genética , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/genética , Superóxido Dismutasa/genética , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina , Líquido del Lavado Bronquioalveolar , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Ratones Endogámicos C57BL , Modelos Biológicos , Neumonía/sangre , Neumonía/enzimología , Neumonía/fisiopatología , Fibrosis Pulmonar/sangre , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/fisiopatología , Superóxido Dismutasa/sangre , Remodelación Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA