Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Respir Res ; 25(1): 299, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39113018

RESUMEN

BACKGROUND: Although recent studies provide mechanistic understanding to the pathogenesis of radiation induced lung injury (RILI), rare therapeutics show definitive promise for treating this disease. Type II alveolar epithelial cells (AECII) injury in various manner results in an inflammation response to initiate RILI. RESULTS: Here, we reported that radiation (IR) up-regulated the TNKS1BP1, causing progressive accumulation of the cellular senescence by up-regulating EEF2 in AECII and lung tissue of RILI mice. Senescent AECII induced Senescence-Associated Secretory Phenotype (SASP), consequently activating fibroblasts and macrophages to promote RILI development. In response to IR, elevated TNKS1BP1 interacted with and decreased CNOT4 to suppress EEF2 degradation. Ectopic expression of EEF2 accelerated AECII senescence. Using a model system of TNKS1BP1 knockout (KO) mice, we demonstrated that TNKS1BP1 KO prevents IR-induced lung tissue senescence and RILI. CONCLUSIONS: Notably, this study suggested that a regulatory mechanism of the TNKS1BP1/CNOT4/EEF2 axis in AECII senescence may be a potential strategy for RILI.


Asunto(s)
Células Epiteliales Alveolares , Senescencia Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Animales , Humanos , Masculino , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Células Epiteliales Alveolares/patología , Células Cultivadas , Senescencia Celular/efectos de la radiación , Senescencia Celular/fisiología , Quinasa del Factor 2 de Elongación/metabolismo , Quinasa del Factor 2 de Elongación/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/genética , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo
2.
Int J Radiat Oncol Biol Phys ; 120(2): 579-592, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38621606

RESUMEN

PURPOSE: Proton minibeam radiation therapy (pMBRT) is an innovative radiation therapy approach that highly modulates the spatial dimension of the dose delivery using narrow, parallel, and submillimetric proton beamlets. pMBRT has proven its remarkable healthy tissue preservation in the brain and skin. This study assesses the potential advantages of pMBRT for thoracic irradiations compared with conventional radiation therapy in terms of normal tissue toxicity. The challenge here was the influence of respiratory motion on the typical peak and valley dose patterns of pMBRT and its potential biologic effect. METHODS AND MATERIALS: The whole thorax of naïve C57BL/6 mice received one fraction of high dose (18 Gy) pMBRT or conventional proton therapy (CPT) without any respiratory control. The development of radiation-induced pulmonary fibrosis was longitudinally monitored using cone beam computed tomography. Anatomopathologic analysis was carried out at 9 months postirradiation and focused on the reaction of the lungs' parenchyma and the response of cell types involved in the development of radiation-induced fibrosis and lung regeneration as alveolar type II epithelial cells, club cells, and macrophages. RESULTS: pMBRT has milder effects on survival, skin reactions, and lung fibrosis compared with CPT. The pMBRT-induced lung changes were more regional and less severe, with evidence of potential reactive proliferation of alveolar type II epithelial cells and less extensive depletion of club cells and macrophage invasion than the more damaging effects observed in CPT. CONCLUSIONS: pMBRT appears suitable to treat moving targets, holding a significant ability to preserve healthy lung tissue, even without respiratory control or precise targeting.


Asunto(s)
Pulmón , Ratones Endogámicos C57BL , Terapia de Protones , Fibrosis Pulmonar , Terapia de Protones/efectos adversos , Terapia de Protones/métodos , Animales , Ratones , Pulmón/efectos de la radiación , Fibrosis Pulmonar/etiología , Tratamientos Conservadores del Órgano/métodos , Tórax/efectos de la radiación , Piel/efectos de la radiación , Órganos en Riesgo/efectos de la radiación , Respiración , Tomografía Computarizada de Haz Cónico , Neumonitis por Radiación/etiología , Dosificación Radioterapéutica , Células Epiteliales Alveolares/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Femenino
3.
Mol Med Rep ; 24(5)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34558633

RESUMEN

Radiation­induced lung tissue injury is an important reason for the limited application of radiotherapy on thoracic malignancies. Previously, we reported that administration of Jiawei­Maxing­Shigan decoction (JMSD) attenuated the radiation­induced epithelial­mesenchymal transition (EMT) in alveolar epithelial cells (AECs) via TGF­ß/Smad signaling. The present study aimed to examine the role of protein phosphatase Mg2+/Mn2+­dependent 1A (PPM1A) in the anti­EMT activity of JMSD on AECs. The components in the aqueous extract of JMSD were identified by high­performance liquid chromatography coupled with electrospray mass spectrometry. Primary rat type II AECs were treated with radiation (60Co γ­ray at 8 Gy) and JMSD­medicated serum. PPM1A was overexpressed and knocked down in the AECs via lentivirus transduction and the effects of JMSD administration on the key proteins related to TGF­ß1/Smad signaling were measured by western blotting. It was found that radiation decreased the PPM1A expression in the AECs and JMSD­medicated serum upregulated the PPM1A expressions in the radiation­induced AECs. PPM1A overexpression increased the E­cadherin level but decreased the phosphorylated (p­)Smad2/3, vimentin and α­smooth muscle actin (α­SMA) levels in the AECs. By contrast, the PPM1A knockdown decreased the E­cadherin level and increased the p­Smad2/3, vimentin and α­SMA levels in the AECs and these effects could be blocked by SB431542 (TGF­ß1/Smad signaling inhibitor). JMSD administration increased the E­cadherin level and decreased the p­Smad2/3, vimentin and α­SMA levels in the AECs; however, these effects could be blocked by siPPM1A­2. In conclusion, PPM1A is a key target of JMSD administration for the attenuation of the radiation­induced EMT in primary type II AECs via the TGF­ß1/Smad pathway.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de la radiación , Proteína Fosfatasa 2C/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Cromatografía Líquida de Alta Presión , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Proteína Fosfatasa 2C/genética , Ratas , Proteínas Smad/genética , Proteínas Smad/metabolismo , Espectrometría de Masa por Ionización de Electrospray
4.
Int J Radiat Oncol Biol Phys ; 110(2): 526-538, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33385497

RESUMEN

PURPOSE: Type II pneumocyte (alveolar epithelial cells type II [AECII]) senescence has been implicated in the progression of lung fibrosis. The capacity of senescent cells to modulate pulmonary macrophages to drive fibrosis is unexplored. Insulin-like growth factor-1 receptor (IGF-1R) signaling has been implicated as a regulator of senescence and aging. METHODS AND MATERIALS: Mice with an AECII-specific deletion of IGF-1R received thoracic irradiation (n ≥ 5 per condition), and the effect of IGF-1R deficiency on radiation-induced AECII senescence and macrophage polarization to an alternatively activated phenotype (M2) was investigated. IGF-1R signaling, macrophage polarization, and senescence were evaluated in surgically resected human lung (n = 63). RESULTS: IGF-1R deficient mice demonstrated reduced AECII senescence (senescent AECII/field; intact: 7.25% ± 3.5% [mean ± SD], deficient: 2.75% ± 2.8%, P = .0001), reduced accumulation of M2 macrophages (intact: 24.7 ± 2.2 cells/field, deficient: 15.5 ± 1.2 cells/field, P = .0086), and fibrosis (hydroxyproline content; intact: 71.9 ± 21.7 µg/lung, deficient: 31.7 ± 7.9, P = .0485) after irradiation. Senescent AECII enhanced M2 polarization in a paracrine fashion (relative Arg1 mRNA, 0 Gy: 1.0 ± 0.4, 17.5 Gy: 7.34 ± 0.5, P < .0001). Evaluation of surgical samples from patients treated with chemoradiation demonstrated increased expression of IGF-1 (unirradiated: 10.2% ± 4.9% area, irradiated: 15.1% ± 11.5%, P = .0377), p21 (unirradiated: 0.013 ± 0.02 histoscore, irradiated: 0.084 ± 0.09 histoscore, P = .0002), IL-13 (unirradiated: 13.7% ± 2.8% area, irradiated: 21.7% ± 3.8%, P < .0001), and M2 macrophages in fibrotic regions relative to nonfibrotic regions (unirradiated: 11.4 ± 12.2 CD163 + cells/core, irradiated: 43.1 ± 40.9 cells/core, P = .0011), consistent with findings from animal models of lung fibrosis. CONCLUSIONS: This study demonstrates that senescent AECII are necessary for the progression of pulmonary fibrosis and serve as a targetable, chronic stimuli for macrophage activation in fibrotic lung.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Polaridad Celular , Senescencia Celular/fisiología , Macrófagos Alveolares/fisiología , Fibrosis Pulmonar/etiología , Receptor IGF Tipo 1/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Senescencia Celular/efectos de la radiación , Quimioradioterapia , Eliminación de Gen , Humanos , Hidroxiprolina/análisis , Pulmón/metabolismo , Pulmón/efectos de la radiación , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Activación de Macrófagos , Macrófagos Alveolares/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/fisiopatología , Traumatismos Experimentales por Radiación/prevención & control , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/genética
5.
J Radiat Res ; 62(1): 119-132, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33302295

RESUMEN

The contribution of type II alveolar epithelial stem cells (AEC II) to radiation-induced lung fibrosis (RILF) is largely unknown. Cell differentiation phenotypes are determined by the balance between Lin28 and lethal-7 microRNA (let-7 miRNA). Lin28 is activated by ß-catenin. The aim of this study was to track AEC II phenotypes at different phases of injury following thoracic irradiation and examine the expression of ß-catenin, Lin28 and let-7 to identify their role in AEC II differentiation. Results showed that coexpression of prosurfactant protein C (proSP-C, an AEC II biomarker) and HOPX (homeobox only protein X, an AEC I biomarker) or vimentin (a differentiation marker) was detected in AEC II post-irradiation. The protein expression levels of HOPX and proSP-C were significantly downregulated, but vimentin was significantly upregulated following irradiation. The expression of E-cadherin, which prevents ß-catenin from translocating to the nucleus, was downregulated, and the expression of ß-catenin and Lin28 was upregulated after irradiation (P < 0.05 to P < 0.001). Four let-7 miRNA members (a, b, c and d) were upregulated in irradiated lungs (P < 0.05 to P < 0.001), but let-7d was significantly downregulated at 5 and 6 months (P < 0.001). The ratios of Lin28 to four let-7 members were low during the early phase of injury and were slightly higher after 2 months. Intriguingly, the Lin28/let-7d ratio was strikingly increased after 4 months. We concluded that ß-catenin contributed to RILF by promoting Lin28 expression, which increased the number of AEC II and the transcription of profibrotic molecules. In this study, the downregulation of let-7d miRNA by Lin28 resulted in the inability of AEC II to differentiate into type I alveolar epithelial cells (AEC I).


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Redes Reguladoras de Genes , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Células Madre/metabolismo , Tórax/efectos de la radiación , beta Catenina/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Biomarcadores/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Redes Reguladoras de Genes/efectos de la radiación , Proteínas de Homeodominio/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , MicroARNs/genética , Fenotipo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Células Madre/efectos de la radiación , Vimentina/metabolismo , Rayos X
6.
J Cell Mol Med ; 24(18): 11018-11023, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32700471

RESUMEN

Radiotherapy is one of the most important treatments for chest tumours. Although there are plenty of strategies to prevent damage to normal lung tissues, it cannot be avoided with the emergence of radiation-induced lung injury. The purpose of this study was to investigate the potential radioprotective effects of glucosamine, which exerted anti-inflammatory activity in joint inflammation. In this study, we found glucosamine relieved inflammatory response and structural damages in lung tissues after radiation via HE staining. Then, we detected the level of epithelial-mesenchymal transition marker in vitro and in vivo, which we could clearly observe that glucosamine treatment inhibited epithelial-mesenchymal transition. Besides, we found glucosamine could inhibit apoptosis and promote proliferation of normal lung epithelial cells in vitro caused by radiation. In conclusion, our data showed that glucosamine alleviated radiation-induced lung injury via inhibiting epithelial-mesenchymal transition, which indicated glucosamine could be a novel potential radioprotector for radiation-induced lung injury.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Antiinflamatorios/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucosamina/uso terapéutico , Pulmón/efectos de la radiación , Fibrosis Pulmonar/prevención & control , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Neumonitis por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Células Epiteliales Alveolares/efectos de la radiación , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Femenino , Rayos gamma/efectos adversos , Glucosamina/farmacología , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/etiología , Neumonitis por Radiación/etiología , Protectores contra Radiación/farmacología , Ratas
7.
Radiat Res ; 192(4): 367-379, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31373871

RESUMEN

Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive complication of therapeutic irradiation of the thorax. It has been suggested that senescence of type II pneumocytes (AECIIs), an alveolar stem cell, plays a role in the development of RIPF through loss of replicative reserve and via senescent AECII-driven release of proinflammatory and profibrotic cytokines. Within this context, we hypothesized that arachidonate 12-lipoxygenase (12-LOX) is a critical mediator of AECII senescence and RIPF. Treatment of wild-type AECIIs with 12S-hydroxyeicosateraenoic acid (12S-HETE), a downstream product of 12-LOX, was sufficient to induce senescence in a NADPH oxidase 4 (NOX4)-dependent manner. Mice deficient in 12-LOX exhibited reduced AECII senescence, pulmonary collagen accumulation and accumulation of alternatively activated (M2) macrophages after thoracic irradiation (5 × 6 Gy) compared to wild-type mice. Conditioned media from irradiated or 12S-HETE-treated primary pneumocytes contained elevated levels of IL-4 and IL-13 compared to untreated pneumocytes. Primary macrophages treated with conditioned media from irradiated AECII demonstrated preferential M2 type polarization when AECIIs were derived from wild-type mice compared to 12-LOX-deficient mice. Together, these data identified 12-LOX as a critical component of RIPF and a therapeutic target for radiation-induced lung injury.


Asunto(s)
Células Epiteliales Alveolares/patología , Araquidonato 12-Lipooxigenasa/metabolismo , Senescencia Celular/efectos de la radiación , Macrófagos/efectos de la radiación , Neumonitis por Radiación/enzimología , Células Epiteliales Alveolares/efectos de la radiación , Animales , Araquidonato 12-Lipooxigenasa/genética , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Interleucina-13/biosíntesis , Interleucina-4/biosíntesis , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neumonitis por Radiación/genética , Neumonitis por Radiación/inmunología , Neumonitis por Radiación/patología
8.
J Cell Biochem ; 120(8): 12500-12507, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30834595

RESUMEN

Radiation-induced lung injury (RILI) frequently occurs in patients with thoracic malignancies. In response to radiation, alveolar epithelial cells (AEC) undergo epithelial-mesenchymal transition (EMT) and contribute to the pathogenesis of RILI. Insulin-like growth factor binding protein 7 (IGFBP7) is reported as a downstream mediator of transforming growth factor-ß1 (TGF-ß1) pathway, which plays a crucial role in radiation-induced EMT. In the present study, the levels of IGFBP7 and TGF-ß1 were simultaneously increased in experimental RILI models and radiation-treated AEC (human pulmonary alveolar epithelial cells [HPAEpic]). The expression of IGFBP7 in radiation-treated HPAEpic cells was obviously inhibited by the specific inhibitor of TGF-ß receptor antagonist SB431542 and TGF-ß1 neutralizing antibody, and time-dependently enhanced by TGF-ß1 treatment. Moreover, IGFBP7 knockdown significantly attenuated the effects of radiation on morphology change, cell migration, expression of EMT-related markers (E-cadherin, α-SMA, and Vimentin), and phosphorylation of extracellular-signal-regulated kinase (ERK). The effects of IGFBP7 overexpression on the expression of EMT-related markers were partially reversed by the ERK inhibitor PD98059. In conclusion, IGFBP7, was enhanced by TGF-ß1, may be involved in radiation-induced EMT of AEC via the ERK signaling pathway, thus contributing to the pathogenesis of RILI.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Sistema de Señalización de MAP Quinasas , Células Epiteliales Alveolares/fisiología , Células Epiteliales Alveolares/efectos de la radiación , Animales , Línea Celular , Movimiento Celular , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Ratas , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
9.
Biomed Pharmacother ; 95: 1219-1224, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28931214

RESUMEN

BACKGROUND: Radiation therapy is commonly used to treat thoracic malignancies. However, it may lead to severe lung pneumonitis and ultimately fibrosis. Irradiation has been reported to increase epithelial-mesenchymal transition (EMT) of type II alveolar epithelial cells (AEC), which play an important role in pulmonary fibrosis. The transforming growth factor-ß (TGF-ß) and ERK/glycogen synthase kinase 3ß (GSK3ß) pathways are critically involved in radiation-induced EMT. In the present study, we investigated whether baicalin was a novel therapeutic candidate for radiation-induced EMT in type II AEC. METHODS: Primary type II AEC were isolated and treated with 60Co γ-rays and a series doses of baicalin (2µM, 10µM and 50µM). The ultrastructure and morphology changes were observed by transmission electron microscopy and optical microscopy, respectively. Protein expression was determined by western blotting analysis. Immunofluorescence staining was performed to detect the nuclear translocation of Snail. RESULTS: After irradiation, type II AEC displayed a mesenchymal-like morphology accompanied by a decrease in E-cadherin expression, an increase in the expression of Vimentin and α-SMA. Nuclear translocation of Snail, the activation of TGF-ß/Smad pathway, and the inactivation of GSK3ß were prominent in radiation-treated cells. Baicalin significantly attenuated the effects of radiation on type II AEC. CONCLUSIONS: Baicalin may a useful radioprotective agent through suppressing the EMT of type II AEC.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Transición Epitelial-Mesenquimal , Flavonoides/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/efectos de la radiación , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Rayos gamma , Transporte de Proteínas/efectos de los fármacos , Ratas Sprague-Dawley , Proteínas Smad/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo
10.
Lasers Med Sci ; 32(8): 1825-1834, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28712048

RESUMEN

Lung fibrosis (LF) is a chronic and progressive lung disease characterized by pulmonary parenchyma progressive lesion, inflammatory infiltration, and interstitial fibrosis. It is developed by excessive collagen deposition and other cellular matrix components, resulting in severe changes in the alveolar architecture. Considering the absence of effective treatment, the aim of this study was to investigate the effect of photobiomodulation therapy (PBMT) on the development of PF. For this purpose, we used C57BL6 mice subjected to induction of LF by bleomycin administration (1.5 U/kg) by orotracheal route and, after 14 days of the induction, mice were treated with PBMT applied to the thorax 1×/day for 8 days (wavelength 660 ± 20 nm, power 100 mW, radiant exposure 5 J/cm2, irradiance 33.3 mW/cm2, spot size 2.8cm2, total energy 15 J, time of irradiation: 150 s) and inflammatory and fibrotic parameters were evaluated with or without PBMT. Our results showed that PBMT significantly reduced the number of inflammatory cells in the alveolar space, collagen production, interstitial thickening, and static and dynamic pulmonary elastance. In addition, we observed reduced levels of IL-6 e CXCL1/KC released by pneumocytes in culture as well as reduced level of CXCL1/KC released by fibroblasts in culture. We can conclude that the PBMT improves both inflammatory and fibrotic parameters showing a promising therapy which is economical and has no side effects.


Asunto(s)
Inflamación/patología , Terapia por Luz de Baja Intensidad/métodos , Fibrosis Pulmonar/radioterapia , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Bleomicina , Lavado Broncoalveolar , Quimiocina CXCL1/metabolismo , Colágeno/biosíntesis , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Inflamación/complicaciones , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Pulmón/patología , Pulmón/efectos de la radiación , Masculino , Ratones Endogámicos C57BL
11.
Med Sci Monit ; 23: 3446-3450, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28710886

RESUMEN

The lung is one of several moderately radiosensitive organs. Radiation-induced lung injury (RILI), including acute radiation pneumonitis and chronic radiation-induced pulmonary fibrosis, occurs most often in radiotherapy of lung cancer, esophageal cancer, and other thoracic cancers. Clinical symptoms of RILI include dry cough, shortness of breath, chest pain, fever, and even severe respiratory failure and death. The occurrence of RILI is a complex process that includes a variety of cellular and molecular interactions which ultimately leads to large fibroblast accumulation, proliferation, and differentiation, resulting in excessive extracellular matrix deposits, causing pulmonary fibrosis. The progress that has been made in recent years in the understanding of cellular and molecular mechanisms of RILI is summarized in this review.


Asunto(s)
Pulmón/efectos de la radiación , Traumatismos por Radiación/tratamiento farmacológico , Neumonitis por Radiación/metabolismo , Anomalías Inducidas por Radiación/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Citocinas/efectos de la radiación , Células Endoteliales/efectos de la radiación , Fibroblastos/efectos de la radiación , Humanos , Pulmón/metabolismo , Pulmón/fisiología , Lesión Pulmonar/metabolismo , Neoplasias Pulmonares/complicaciones , Macrófagos/efectos de la radiación , Fibrosis Pulmonar/etiología
12.
Int J Mol Sci ; 17(6)2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27322243

RESUMEN

Spaceflight occasionally requires multiple extravehicular activities (EVA) that potentially subject astronauts to repeated changes in ambient oxygen superimposed on those of space radiation exposure. We thus developed a novel in vitro model system to test lung cell damage following repeated exposure to radiation and hyperoxia. Non-tumorigenic murine alveolar type II epithelial cells (C10) were exposed to >95% O2 for 8 h only (O2), 0.25 Gy ionizing γ-radiation (IR) only, or a double-hit combination of both challenges (O2 + IR) followed by 16 h of normoxia (ambient air containing 21% O2 and 5% CO2) (1 cycle = 24 h, 2 cycles = 48 h). Cell survival, DNA damage, apoptosis, and indicators of oxidative stress were evaluated after 1 and 2 cycles of exposure. We observed a significant (p < 0.05) decrease in cell survival across all challenge conditions along with an increase in DNA damage, determined by Comet analysis and H2AX phosphorylation, and apoptosis, determined by Annexin-V staining, relative to cells unexposed to hyperoxia or radiation. DNA damage (GADD45α and cleaved-PARP), apoptotic (cleaved caspase-3 and BAX), and antioxidant (HO-1 and Nqo1) proteins were increased following radiation and hyperoxia exposure after 1 and 2 cycles of exposure. Importantly, exposure to combination challenge O2 + IR exacerbated cell death and DNA damage compared to individual exposures O2 or IR alone. Additionally levels of cell cycle proteins phospho-p53 and p21 were significantly increased, while levels of CDK1 and Cyclin B1 were decreased at both time points for all exposure groups. Similarly, proteins involved in cell cycle arrest was more profoundly changed with the combination challenges as compared to each stressor alone. These results correlate with a significant 4- to 6-fold increase in the ratio of cells in G2/G1 after 2 cycles of exposure to hyperoxic conditions. We have characterized a novel in vitro model of double-hit, low-level radiation and hyperoxia exposure that leads to oxidative lung cell injury, DNA damage, apoptosis, and cell cycle arrest.


Asunto(s)
Daño del ADN , Hiperoxia , Modelos Biológicos , Estrés Oxidativo , Radiación Ionizante , Vuelo Espacial , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Antioxidantes/metabolismo , Apoptosis/genética , Apoptosis/efectos de la radiación , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Regulación Enzimológica de la Expresión Génica , Histonas/metabolismo , Humanos , Ratones , Oxidación-Reducción , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
13.
Mol Med Rep ; 13(6): 4666-70, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27082939

RESUMEN

Radiation-induced lung injury (RILI) involves pneumonitis and fibrosis, and results in pulmonary dysfunction. Moreover, RILI can be a fatal complication of thoracic radiotherapy. The present study investigated the protective effect of geranylgeranlyacetone (GGA), an inducer of heat shock protein (HSP)70, on RILI using a C57BL/6 mouse model of RILI developing 6 months subsequent to exposure to 12.5 Gy thoracic radiation. GGA was administered 5 times orally prior and subsequent to radiation exposure, and the results were assessed by histological analysis and western blotting. The results show that late RILI was alleviated by GGA treatment, possibly through the suppression of epithelial­to­mesenchymal transition (EMT) marker expression. Based on histological examination, orally administered GGA during the acute phase of radiation injury not only significantly inhibited pro­surfactant protein C (pro­SPC) and vimentin expression, but also preserved E­cadherin expression 6 months after irradiation­induced injury of the lungs. GGA induced HSP70 and inhibited EMT marker expression in L132 human lung epithelial cells following IR. These data suggest that the prevention of EMT signaling is a key cytoprotective effect in the context of RILI. Thus, HSP70­inducing drugs, such as GGA, could be beneficial for protection against RILI.


Asunto(s)
Diterpenos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neumonitis por Radiación/metabolismo , Neumonitis por Radiación/patología , Transducción de Señal/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/efectos de la radiación , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Traumatismos por Radiación/tratamiento farmacológico , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Neumonitis por Radiación/tratamiento farmacológico
14.
Int J Radiat Oncol Biol Phys ; 94(5): 1163-72, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26883561

RESUMEN

PURPOSE: To determine whether the delivery of recombinant truncated plasminogen activator inhibitor-1 (PAI-1) protein (rPAI-1(23)) would protect from the development of radiation-induced lung injury. METHODS AND MATERIALS: C57Bl/6 mice received intraperitoneal injections of rPAI-1(23) (5.4 µg/kg/d) or vehicle for 18 weeks, beginning 2 days before irradiation (IR) (5 daily fractions of 6 Gy). Cohorts of mice were followed for survival (n=8 per treatment) and tissue collection (n=3 per treatment and time point). Fibrosis in lung was assessed with Masson-Trichrome staining and measurement of hydroxyproline content. Senescence was assessed with staining for ß-galactosidase activity in lung and primary pneumocytes. RESULTS: Hydroxyproline content in irradiated lung was significantly reduced in mice that received rPAI-1(23) compared with mice that received vehicle (IR+vehicle: 84.97 µg/lung; IR+rPAI-1(23): 56.2 µg/lung, P=.001). C57Bl/6 mice exposed to IR+vehicle had dense foci of subpleural fibrosis at 19 weeks, whereas the lungs of mice exposed to IR+rPAI-1(23) were largely devoid of fibrotic foci. Cellular senescence was significantly decreased by rPAI-1(23) treatment in primary pneumocyte cultures and in lung at multiple time points after IR. CONCLUSIONS: These studies identify that rPAI-1(23) is capable of preventing radiation-induced fibrosis in murine lungs. These antifibrotic effects are associated with increased fibrin metabolism, enhanced matrix metalloproteinase-3 expression, and reduced senescence in type 2 pneumocytes. Thus, rPAI-1(23) is a novel therapeutic option for radiation-induced fibrosis.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/uso terapéutico , Fibrosis Pulmonar/prevención & control , Neumonitis por Radiación/complicaciones , Proteínas Recombinantes/uso terapéutico , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Proliferación Celular , Senescencia Celular/efectos de la radiación , Colágeno/metabolismo , Citocinas/metabolismo , Femenino , Fibrina/metabolismo , Hidroxiprolina/análisis , Hidroxiprolina/metabolismo , Pulmón/metabolismo , Pulmón/efectos de la radiación , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Inhibidor 1 de Activador Plasminogénico/metabolismo , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Neumonitis por Radiación/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Asian Pac J Cancer Prev ; 16(7): 2613-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25854336

RESUMEN

Radiation induced lung injury has long been considered a treatment limiting factor for patients requiring thoracic radiation. This radiation induced lung injury happens early as well as late. Radiation induced lung injury can occur in two phases viz. early (<6 months) when it is called radiation pneumonitis and late (>6 months) when it is called radiation induced lung fibrosis. There are multiple factors that can be patient, disease or treatment related that predict the incidence and severity of radiation pneumonitis. Radiation induced damage to the type I pneumocytes is the triggering factor to initiate such reactions. Over the years, radiation therapy has witnessed a paradigm shift in radiation planning and delivery and successfully reduced the incidence of lung injury. Radiation pneumonitis is usually a diagnosis of exclusion. Steroids, ACE inhibitors and pentoxyphylline constitute the cornerstone of therapy. Radiation induced lung fibrosis is another challenging aspect. The pathophysiology of radiation fibrosis includes continuing inflammation and microvascular changes due to pro-angiogenic and pro- fibrogenic stimuli resembling those in adult bronchiectasis. General supportive management, mobilization of airway secretions, anti-inflammatory therapy and management of acute exacerbations remains the treatment option. Radiation induced lung injury is an inevitable accompaniment of thoracic radiation.


Asunto(s)
Antiinflamatorios/uso terapéutico , Neumonitis por Radiación/tratamiento farmacológico , Neumonitis por Radiación/fisiopatología , Protectores contra Radiación/uso terapéutico , Radiografía Torácica/efectos adversos , Anciano , Células Epiteliales Alveolares/efectos de la radiación , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antioxidantes/uso terapéutico , Humanos , Inflamación , Pulmón/efectos de la radiación , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/fisiopatología , Pentoxifilina/uso terapéutico , Prednisolona/uso terapéutico , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/fisiopatología
16.
Lung ; 193(3): 409-19, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25749666

RESUMEN

INTRODUCTION: Stereotactic ablative radiotherapy is a newly emerging radiotherapy treatment method that, compared with conventionally fractionated radiation therapy (CFRT), allows an ablative dose of radiation to be delivered to a confined area around a tumor. The aim of the present study was to investigate the changes of various cytokines that may be involved in ablative radiation-induced lung injury in vitro and in vivo. METHODS: In the in vivo study, ablative-dose radiation was delivered to a small volume of the left lung of C3H/HeJCr mice using a small-animal irradiator. The levels of 24 cytokines in the peripheral blood were tested at several time points after irradiation. For the in vitro study, three mouse cell types (type II pneumocytes, alveolar macrophages, and fibroblasts) known to play important roles in radiation-induced pneumonitis and lung fibrosis were analyzed using a co-culture system. RESULTS: In the in vivo study, we found obvious patterns of serum cytokine changes depending on the volume of tissue irradiated (2-mm vs. 3.5-mm collimator). Only the levels of 3 cytokines increased with the 2-mm collimator at the acute phase (1-2 weeks after irradiation), while the majority of cytokines were elevated with the 3.5-mm collimator. In the in vitro co-culture system, after the cells were given an ablative dose of irradiation, the levels of five cytokines (GM-CSF, G-CSF, IL-6, MCP-1, and KC) increased significantly in a dose-dependent manner. CONCLUSIONS: The cytokine levels in our radiation-induced lung injury model showed specific changes, both in vivo and in vitro. These results imply that biological studies related to ablative-dose small-volume irradiation should be investigated using the corresponding experimental models rather than on those simulating large-volume CFRT.


Asunto(s)
Técnicas de Ablación , Citocinas/sangre , Pulmón/efectos de la radiación , Neumonitis por Radiación/sangre , Radiocirugia , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Animales , Técnicas de Cocultivo , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Regulación de la Expresión Génica , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/efectos de la radiación , Ratones , Ratones Endogámicos C3H , Células 3T3 NIH , Dosis de Radiación , Neumonitis por Radiación/etiología , Neumonitis por Radiación/genética , Neumonitis por Radiación/inmunología , Factores de Tiempo
17.
Technol Cancer Res Treat ; 14(1): 81-98, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24325134

RESUMEN

The goal was to characterize differences in cell response after exposure to active beam scanning (ABS) protons compared to a passive delivery system. Human lung epithelial (HLE) cells were evaluated at various locations along the proton depth dose profile. The dose delivered at the Bragg peak position was essentially identical (∼4 Gy) with the two techniques, but depth dose data showed that ABS resulted in lower doses at entry and more rapid drop-off after the peak. Average dose rates for the passive and ABS beams were 1.1 Gy/min and 5.1 Gy/min, respectively; instantaneous dose rates were 19.2 Gy/min and 2,300 Gy/min (to a 0.5 × 0.5 mm(2) voxel). Analysis of DNA synthesis was based on (3)H-TdR incorporation. Quantitative real-time polymerase chain reaction (RT-PCR) was done to determine expression of genes related to p53 signaling and DNA damage; a total of 152 genes were assessed. Spectral karyotyping and analyses of the Golgi apparatus and cytokines produced by the HLE cells were also performed. At or near the Bragg peak position, ABS protons resulted in a greater decrease in DNA synthesis compared to passively delivered protons. Genes with >2-fold change (P < 0.05 vs. 0 Gy) after passive proton irradiation at one or more locations within the Bragg curve were BTG2, CDKN1A, IFNB1 and SIAH1. In contrast, many more genes had >2-fold difference with ABS protons: BRCA1, BRCA2, CDC25A, CDC25C, CCNB2, CDK1, DMC1, DNMT1, E2F1, EXO1, FEN1, GADD45A, GTSE1, IL-6, JUN, KRAS, MDM4, PRC1, PTTG1, RAD51, RPA1, TNF, WT1, XRCC2, XRCC3 and XRCC6BP1. Spectral karyotyping revealed numerous differences in chromosomal abnormalities between the two delivery systems, especially at or near the Bragg peak. Percentage of cells staining for the Golgi apparatus was low after exposure to passive and active proton beams. Studies such as this are needed to ensure patient safety and make modifications in ABS delivery, if necessary.


Asunto(s)
Células Epiteliales Alveolares/efectos de la radiación , Protones/efectos adversos , Células Epiteliales Alveolares/metabolismo , Aberraciones Cromosómicas/efectos de la radiación , Citocinas/genética , Citocinas/metabolismo , Daño del ADN/genética , Replicación del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de la radiación , Aparato de Golgi/metabolismo , Aparato de Golgi/efectos de la radiación , Humanos , Cariotipo , Persona de Mediana Edad , Terapia de Protones , Dosis de Radiación , Radiación Ionizante , Efectividad Biológica Relativa , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
18.
J Natl Cancer Inst ; 105(19): 1474-84, 2013 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-24052614

RESUMEN

BACKGROUND: Radiation is a commonly delivered therapeutic modality for cancer. The causes underlying the chronic, progressive nature of radiation injury in the lung are poorly understood. METHODS: C57Bl/6NCr mice were exposed to thoracic irradiation (n = 3 per dose and time point for tissue collection). Microarray analysis of gene expression from irradiated murine lung was performed using one-way analysis of variance with post hoc Scheffe analysis. Senescence and type II airway epithelial cell (AECII) count were assayed in irradiated murine lung tissue (n = 3 per condition). Irradiated mice were treated with diphenyleneiodonium (DPI), an inhibitor of NADPH oxidase (NOX), and fibrosis was assessed by collagen assays. All statistical tests were two-tailed. RESULTS: Gene expression in lung tissue from mice irradiated to 17.5 Gy clustered with that of aged unirradiated mice. Only fibrogenic exposures led to AECII senescence (0 Gy: 0.66% ± 0.67%; 5 Gy: 4.5% ± 1.19%; 17.5 Gy: 18.7% ± 3.05; P = .007) and depletion (0 Gy: 2.89 per alveolus ± 0.26; 5 Gy: 2.41 ± 0.19; 17.5 Gy: 1.6 ± 0.14; P < .001) at 30 weeks. Treatment of irradiated mice with DPI for 16 weeks markedly reduced collagen accumulation (5×6 Gy: 57.26 µg/lung ± 9.91; 5×6 Gy ± DPI: 36.54µg/lung ± 4.39; P = .03) and AECII senescence (5×6 Gy: 37.61% ± 4.82%; 5×6 Gy ± DPI: 12.38% ± 2.78; P < .001). CONCLUSIONS: These studies identify senescence as an important process in AECII in vivo and indicate that NOX is a critical mediator of radiation-induced AECII senescence and pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/efectos de la radiación , Senescencia Celular , Colágeno/metabolismo , NADPH Oxidasas/metabolismo , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/patología , Animales , Senescencia Celular/efectos de la radiación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/efectos de la radiación , Pulmón/patología , Pulmón/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/antagonistas & inhibidores , Compuestos Onio , Factores de Tiempo , Análisis de Matrices Tisulares
20.
In Vivo ; 27(3): 291-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23606683

RESUMEN

BACKGROUND/AIM: Thoracic irradiation results in an acute inflammatory response, latent period, and late fibrosis. Little is known about the mechanisms involved in triggering late radiation fibrosis. MATERIALS AND METHODS: Thoracic irradiated fibrosis prone C57BL/6NTac mice were followed for detectable mRNA transcripts in isolated lung cells and micro-RNA in whole-tissues, and the effect of administration of water-soluble oxetanyl sulfoxide MMS350 was studied. Marrow stromal cell motility in medium from fibrotic-phase explanted pulmonary endothelial and alveolar type-II cells was measured. RESULTS: RNA and micro-RNA expression in lung correlated with fibrosis. MMS350 reduced pro-fibrotic gene expression in both endothelial and alveolar type-II cells in irradiated mice. Conditioned medium from irradiated cells did not alter cell motility in vitro. CONCLUSION: These studies should facilitate identification of potential new drug targets for ameliorating irradiation-induced pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/efectos de la radiación , Éteres Cíclicos/metabolismo , Alveolos Pulmonares/efectos de la radiación , Fibrosis Pulmonar/etiología , Neumonitis por Radiación , Radioterapia/efectos adversos , Sulfóxidos/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Neumonitis por Radiación/genética , Neumonitis por Radiación/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/efectos de la radiación , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA