Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Transl Med ; 15(1): 123, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28569199

RESUMEN

BACKGROUND: Shikonin, a natural naphthoquinone pigment purified from Lithospermum erythrorhizon, induces necroptosis in various cancer types, but the mechanisms underlying the anticancer activity of shikonin in lung cancer are not fully understood. This study was designed to clarify whether shikonin causes necroptosis in non-small cell lung cancer (NSCLC) cells and to investigate the mechanism of action. METHODS: Multiplex and caspase 8 assays were used to analyze effect of shikonin on A549 cells. Cytometry with annexin V/PI staining and MTT assays were used to analyze the mode of cell death. Western blotting was used to determine the effect of shikonin-induced necroptosis and autophagy. Xenograft and orthotopic models with A549 cells were used to evaluate the anti-tumor effect of shikonin in vivo. RESULTS: Most of the cell death induced by shikonin could be rescued by the specific necroptosis inhibitor necrostatin-1, but not by the general caspase inhibitor Z-VAD-FMK. Tumor growth was significantly lower in animals treated with shikonin than in the control group. Shikonin also increased RIP1 protein expression in tumor tissues. Autophagy inhibitors, including methyladenine (3-MA), ATG5 siRNA, and bafilomycin A, enhanced shikonin-induced necroptosis, whereas RIP1 siRNA had no effect on the apoptotic potential of shikonin. CONCLUSIONS: Our data indicated that shikonin treatment induced necroptosis and autophagy in NSCLC cells. In addition, the inhibition of shikonin-induced autophagy enhanced necroptosis, suggesting that shikonin could be a novel therapeutic strategy against NSCLC.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Naftoquinonas/farmacología , Necrosis , Células A549 , Animales , Caspasa 8/metabolismo , Línea Celular Tumoral , Silenciador del Gen , Humanos , Imidazoles/farmacología , Indoles/farmacología , Lithospermum , Macrólidos/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , ARN Interferente Pequeño/metabolismo , Microtomografía por Rayos X
2.
Tohoku J Exp Med ; 242(2): 137-142, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28626164

RESUMEN

Malignancy and tuberculosis are common causes of lymphocytic exudative pleural effusion. However, it is occasionally difficult to differentiate malignant pleural effusion from tuberculous pleural effusion. Vascular endothelial growth factor (VEGF) is a critical cytokine in the pathogenesis of malignant pleural effusion. Endocan is a dermatan sulfate proteoglycan that is secreted by endothelial cells. Importantly, endocan mediates the vascular growth-promoting action of VEGF. The aim of this study was to evaluate the diagnostic significance of VEGF and endocan in pleural effusion. We thus measured the levels of VEGF and endocan in the pleural effusion and serum samples of patients with lung cancer (n = 59) and those with tuberculosis (n = 32) by enzyme-linked immunosorbent assay. Lung cancer included 40 cases of adenocarcinoma, 13 of squamous cell carcinoma, and 6 of small cell carcinoma. Pleural effusion VEGF levels were significantly higher in the malignant group than in the tuberculosis group (2,091.47 ± 1,624.80 pg/mL vs. 1,291.05 ± 1,100.53 pg/mL, P < 0.05), whereas pleural effusion endocan levels were similar between the two groups (1.22 ± 0.74 ng/mL vs. 0.87 ± 0.53 ng/mL). The areas under the curve of VEGF and endocan were 0.73 and 0.52, respectively. Notably, the VEGF levels were similar in malignant pleural effusion, irrespective of the histological type of lung cancer. Moreover, no significant difference was found in the serum VEGF and endocan levels between patients with lung cancer and those with tuberculosis. In conclusion, high VEGF levels in pleural effusion are suggestive of malignant pleural effusion.


Asunto(s)
Derrame Pleural Maligno/diagnóstico , Tuberculosis Pleural/diagnóstico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/sangre , Derrame Pleural Maligno/sangre , Proteoglicanos/sangre , Curva ROC , Tuberculosis Pleural/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Adulto Joven
3.
Exp Cell Res ; 323(2): 288-96, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24631288

RESUMEN

Although non-small cell lung cancer (NSCLC) tumors with activating mutations in the epidermal growth factor receptor (EGFR) are highly responsive to EGFR tyrosine kinase inhibitors (TKIs) including gefitinib and erlotinib, development of acquired resistance is almost inevitable. Statins show antitumor activity, but it is unknown whether they can reverse EGFR-TKIs resistance in NSCLC with the T790M mutation of EGFR. This study investigated overcoming resistance to EGFR-TKI using simvastatin. We demonstrated that addition of simvastatin to gefitinib enhanced caspase-dependent apoptosis in T790M mutant NSCLC cells. Simvastatin also strongly inhibited AKT activation, leading to suppression of ß-catenin activity and the expression of its targets, survivin and cyclin D1. Both insulin treatment and AKT overexpression markedly increased p-ß-catenin and survivin levels, even in the presence of gefitinib and simvastatin. However, inhibition of AKT by siRNA or LY294002 treatment decreased p-ß-catenin and survivin levels. To determine the role of survivin in simvastatin-induced apoptosis of gefitinib-resistant NSCLC, we showed that the proportion of apoptotic cells following treatment with survivin siRNA and the gefitinib-simvastatin combination was greater than the theoretical additive effects, whereas survivin up-regulation could confer protection against gefitinib and simvastatin-induced apoptosis. Similar results were obtained in erlotinib and simvastatin-treated HCC827/ER cells. These findings suggest that survivin is a key molecule that renders T790M mutant NSCLC cells resistant to apoptosis induced by EGFR-TKIs and simvastatin. Overall, these data indicate that simvastatin may overcome EGFR-TKI resistance in T790M mutant NSCLCs via an AKT/ß-catenin signaling-dependent down-regulation of survivin and apoptosis induction.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Simvastatina/farmacología , Apoptosis/efectos de los fármacos , Cateninas/genética , Cateninas/metabolismo , Línea Celular Tumoral , Receptores ErbB/genética , Gefitinib , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mutación Missense , Survivin
4.
Tohoku J Exp Med ; 235(4): 327-33, 2015 04.
Artículo en Inglés | MEDLINE | ID: mdl-25854270

RESUMEN

Residual pleural thickening (RPT) is the most frequent complication associated with pleural tuberculosis, and may occur even after successful anti-tuberculosis medications. Matrix metalloproteinases (MMPs) are zinc-dependent proteinases capable of degrading all components of the extracellular matrix. The proteolytic action of MMPs may be involved in the pathogenesis of tuberculosis. MMP-9, secreted by monocytes and lymphocyte, may lead to long-term fibrosis. The aim of the present study was to determine whether MMP-2 and/or MMP-9 and their specific inhibitors, tissue inhibitors of metalloproteinase 1 (TIMP-1) and TIMP-2, could be used to predict RPT. This retrospective study enrolled 52 patients diagnosed with pleural tuberculosis. Levels of MMP-2, MMP-9, TIMP-1, and TIM-2 were determined in the pleural fluid by ELISA. The RPT was measured on chest X-ray at the completion of treatment and the final follow-up. The average periods of anti-tuberculosis medication and the follow-up after completion of treatment were 6.7 and 7.6 months, respectively. MMP-2 or MMP-9 levels had no significant correlation to RPT. The patients with RPT > 2 mm at the completion of anti-tuberculosis medication and the final follow-up had higher TIMP-1 levels (p = 0.00 and p = 0.001, respectively). However, patients with RPT > 2 mm at the completion of anti-tuberculosis medication had lower TIMP-2 levels (p = 0.005). In a logistic regression model, elevated TIMP-1 levels at the completion of anti-tuberculosis medications were associated with RPT. In conclusion, higher TIMP-1 levels are responsible for the development of RPT and may be helpful for predicting RPT in pleural tuberculosis.


Asunto(s)
Pleura/enzimología , Pleura/patología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Tuberculosis Pleural/enzimología , Tuberculosis Pleural/patología , Adulto , Antituberculosos/uso terapéutico , Femenino , Estudios de Seguimiento , Humanos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Análisis Multivariante , Pleura/diagnóstico por imagen , Radiografía , Tuberculosis Pleural/diagnóstico por imagen , Tuberculosis Pleural/tratamiento farmacológico
5.
Cytokine ; 58(3): 336-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22480951

RESUMEN

BACKGROUND: YKL-40 (a chitinase-like protein) is an inflammatory biomarker that is associated with lung injury pathogenesis. We aimed to identify the diagnostic values of YKL-40 in pleural effusions and to evaluate circulating YKL-40 levels during multiple etiological pulmonary/pleural diseases and the role of YKL-40 as a monitoring marker of inflammatory pulmonary disease. METHODS: Pleural YKL-40 (n=197), YKL-39 (the most homologous chitinase-like protein to human YKL-40), and conventional pleural marker levels were measured in patients with pulmonary/pleural disease. Additionally, serum YKL-40 and YKL-39 levels were analyzed in both patients and controls (n=432) and serially monitored in patients with asthma (n=27) or pneumonia (n=22). RESULTS: Pleural YKL-40 levels were higher than those in the serum and highest in tuberculous pleural effusions (TPEs; 1181 ng/mL), followed by parapneumonic, malignant, and cardiogenic effusions (560 ng/mL). The diagnostic accuracy of pleural YKL-40 (0.78) for discriminating between tuberculous and malignant effusion was comparable to or greater than those of YKL-39, total protein, C-reactive protein and CYFRA 21-1, and lower than those of adenosine deaminase (p<0.05) and carcinoembriogenic antigen (p=0.05). Serum YKL-40 levels were higher in the pneumonia group than in the cancer, asthma, or control groups. Following treatment, serum YKL-40 levels were more greatly reduced in pneumonia patients than in asthma patients. Serum YKL-39 levels did not differ between patients and controls. CONCLUSIONS: Pleural YKL-40 levels are elevated in TPEs and have fairly good diagnostic efficacy for detecting TPEs. However, adenosine deaminase is more efficient for detecting TPEs than pleural YKL-40. Serum YKL-40 levels are highest during pneumonia compared to common pulmonary/pleural diseases and are more useful for monitoring pneumonia than asthma.


Asunto(s)
Adipoquinas/metabolismo , Lectinas/metabolismo , Enfermedades Pulmonares/metabolismo , Enfermedades Pleurales/metabolismo , Derrame Pleural/metabolismo , Adipoquinas/sangre , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Proteína 1 Similar a Quitinasa-3 , Femenino , Humanos , Lectinas/sangre , Enfermedades Pulmonares/sangre , Masculino , Persona de Mediana Edad , Enfermedades Pleurales/sangre , Estudios Prospectivos
6.
J Infect Chemother ; 18(6): 951-4, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22398882

RESUMEN

Although tuberculosis can affect various organs and tissues, the lung is the site most commonly involved. Extrapulmonary tuberculosis (EPTB) involves relatively inaccessible and variable sites and is consequently often overlooked by clinicians. The ear is a notably very rare site of EPTB, and the diagnosis is difficult because of the variable and confusing signs and symptoms. To our knowledge, this is the first case in which tuberculous otitis media and endobronchial tuberculosis coexisted.


Asunto(s)
Mycobacterium tuberculosis/aislamiento & purificación , Otitis Media/diagnóstico , Tuberculosis Pulmonar/diagnóstico , Tuberculosis/diagnóstico , Antituberculosos , Biopsia , Bronquios/microbiología , Broncoscopía , Oído Medio/microbiología , Oído Medio/patología , Femenino , Humanos , Persona de Mediana Edad , Otitis Media/microbiología , Radiografía Torácica , Hueso Temporal/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Tuberculosis/microbiología , Tuberculosis Pulmonar/microbiología
7.
Invest New Drugs ; 29(5): 945-52, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20464445

RESUMEN

Statins, HMG-CoA reductase inhibitors have been studied for their antiproliferative and proapototic effects. Recently, statin-induced apoptosis has been associated with down-regulation of survivin expression in cancer cells. However, the mechanism of deregulated survivin by simvastatin on lung cancer is still unclear. Herein, we demonstrated that simvastatin induced caspase-dependent apoptosis in A549 lung cancer cells. Simvastatin also resulted in a decrease in the expression of phosphorylated Akt. In addition, simvastatin effectively down-regulated survivin mRNA and protein, but not cIAP-1 and cIAP-2. The combination of simvastatin and 10 µM LY294002 (non-toxic dose) augmented apoptosis significantly, as evidenced by cleavage of PARP. The immunoreactive band of survivin was markedly decreased in cells treated with 50 µM LY294002 (toxic dose) as well as by the combination of simvastatin and 10 µM LY294002. Moreover, survivin down-regulation by RNA interference induced apoptosis accompanied by an increase in hypodiploid DNA content. Taken together, these data suggest that the anti-cancer effect of simvastatin via induction of apoptosis is related to Akt signaling dependent down-regulation of survivin in lung cancer A549 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/genética , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Simvastatina/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Morfolinas/farmacología , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Survivin
8.
Lung Cancer ; 60(1): 47-56, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18006113

RESUMEN

Heme oxygenase-1 (HO-1) is highly expressed in various tumor tissues and plays an important role in tumor cell growth through anti-oxidative and anti-apoptotic effects. Herein, we demonstrate that A549 cells express high levels of HO-1, Nrf2, and NF-kappaB compared to other lung cancer cell lines, including H23, H157, and H460. Ectopic expression of HO-1 small interfering RNA (siRNA) increased both apoptosis and degradation of procaspase-3. Transfection studies with siRNA specific for Nrf2 and NF-kappaB revealed that HO-1 expression in A549 cells is mediated by transcriptional activation of Nrf2, but not NF-kappaB. A549 cells are less susceptible to cisplatin cytotoxicity than other lung cancer cell lines, concomitant with increases in HO-1 expression and MAPK phosphorylation in a time-dependent fashion. Furthermore, inhibition of HO-1 by siRNA and a specific HO-1 inhibitor ZnPP augments cisplatin cytotoxicity toward A549 cells. Pharmacologic suppression of HO-1 activity resulted in a marked increase in the ROS generation in cisplatin-treated cells. In addition, pharmacologic inhibitors of MAPK suppressed the induction of HO-1 and Nrf2 expression by cisplatin. These findings suggest that HO-1 may modulate the chemosensitivity of lung cancer A549 cells to cisplatin through the MAPK-Nrf2 pathway.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Hemo-Oxigenasa 1/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Apoptosis , Línea Celular Tumoral , Hemo-Oxigenasa 1/análisis , Hemo-Oxigenasa 1/genética , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor 2 Relacionado con NF-E2/análisis , Factor 2 Relacionado con NF-E2/fisiología , FN-kappa B/análisis , Fosforilación , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Activación Transcripcional
9.
Oncol Rep ; 20(2): 379-84, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18636201

RESUMEN

Arsenic trioxide (As2O3) has been introduced to the treatment of acute promyelocytic leukemia (APL), and has also been shown to induce apoptosis in a variety of solid tumor cell lines, including non-small cell lung cancer. However, the prohibitively high concentration required for the induction of apoptotic cell death in many solid tumor cells is unacceptable for clinical utilization due to the excessive toxicity associated with this dose. Sulindac is known to enhance the cellular responsiveness of tumors toward chemotherapeutic drugs. Herein, we demonstrated that combination treatment with As2O3 and sulindac resulted in a synergistic augmentation of cytotoxicity in H157 lung cancer cells, which was revealed by apoptotic induction as demonstrated by an increase in the sub-G0/G1 fraction. In addition, combination treatment with As2O3 and sulindac increased reactive oxygen species (ROS) and oxidative stress, as evidenced by the heme oxygenase-1 (HO-1) expression and mitogen-activated protein kinase (MAPK) phosphorylation. MAPK inhibitors blocked the induction of HO-1 by combination treatment. Inhibitors of p38 and JNK partially inhibited the augmented cell death whereas the ERK inhibitor showed poor inhibition. Combination treatment with As2O3 and sulindac induced oxidative DNA damage in a time-dependent fashion, which was evaluated by H2AX phosphorylation along with HO-1 induction.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Pulmonares/patología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/administración & dosificación , Western Blotting , Daño del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Fase G1/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Histonas/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxidos/administración & dosificación , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Sulindac/administración & dosificación , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Respir Med Case Rep ; 25: 58-60, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30003022

RESUMEN

We report first case of pulmonary siderosis appearing as a consolidation upon radiological examination and being misdiagnosed as pneumonia. A 59-year-old man visited our hospital with a cough and sputum that had persisted for more than a month. He had undergone chest computed tomography (CT) after abnormal findings on chest X-ray at other hospitals. Based on the chest CT results, he was diagnosed with pneumonia. He was then administered antibiotics for 3 weeks, but there was no improvement. We identified the patient's occupational history first, and then performed bronchoalveolar lavage and chest CT-guided transthoracic lung biopsy. The obtained specimen showed alveolar, macrophage-containing, Prussian blue-positive iron particles. Based on the results, we diagnose/d the patient with pulmonary siderosis. We advised him to discontinue his job. He is currently undergoing observation, and has not shown any special symptoms.

11.
Korean J Intern Med ; 33(2): 340-346, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28192888

RESUMEN

BACKGROUND/AIMS: Brain and bone metastases are common in patients with lung cancer. The development of metastasis is associated with poor survival in lung cancer patients. Although tumor morphologic features on radiographs are routinely assessed for differentiation between benign and malignant lung nodules, they are not used to predict metastasis. We assessed morphologic features of pulmonary adenocarcinomas with brain/bone metastasis on computed tomography (CT) to identify related factors for metastasis. METHODS: We performed a retrospective analysis of initial chest CT findings (size, type of contour, percentage of necrosis, enhancement, presence or absence of calcification, and air cavity) from 2009 to 2010 of patients with brain or bone metastasis and compared the findings with those of patients without metastases. RESULTS: In total, 128 patients were included (78 men, 52 women; mean age 69 years; range, 36 to 87). Nineteen patients had brain metastases and 32 had bone metastases. Morphologic features associated with brain metastasis included size ≥ 50 mm (odds ratio [OR], 3.37; 95% confidence interval [CI], 1.24 to 9.17; p = 0.013), necrosis ≥ 30% (OR, 4.51; 95% CI, 1.62 to 12.55; p =0.002), and presence of calcification (OR, 3.97; 95% CI, 1.16 to 13.55; p = 0.035). Morphologic features associated with bone metastasis included necrosis ≥ 30% (OR, 4.639; 95% CI, 1.98 to 10.82; p < 0.001) and T 3 to 4 stage (OR, 2.53; 95% CI, 1.07 to 6.00; p = 0.031). CONCLUSIONS: We found that necrosis ≥ 30% was associated with pulmonary adenocarcinoma with brain and bone metastasis at initial chest CT morphologic feature. To validate these results, further research should be conducted.


Asunto(s)
Adenocarcinoma , Neoplasias Óseas , Neoplasias Encefálicas , Neoplasias Pulmonares , Tomografía Computarizada por Rayos X , Adenocarcinoma/secundario , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Óseas/secundario , Encéfalo , Neoplasias Encefálicas/secundario , Femenino , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Estudios Retrospectivos
12.
Int J Oncol ; 28(6): 1401-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16685442

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are known to enhance the responsiveness of tumor cells toward chemotherapeutic drugs and radiation. However, the precise mechanism of synergistic enhancement in tumoricidal activity is not clearly known. Herein, we demonstrate that the combination treatment of arsenic trioxide (As2O3) and sulindac resulted in a synergistic augmentation of cytotoxicity toward NCI-H157 lung cancer cells, which was revealed as apoptosis accompanied by chromatin fragmentation and an increase in sub-G0/G1 fraction. In addition, combination treatment with As2O3 and sulindac increased the catalytic activity of caspase-3, -8, and -9 along with induction of Fas/FasL expression and cytosolic release of cytochrome c. Pharmacologic scavenging study of reactive oxygen species (ROS) revealed that synergistic augmentation of cytotoxicity was achieved by generation of ROS, which might modulate the expression of Bcl-2 family proteins, the activity of caspase-3, and mitochondrial membrane potential transition.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Neoplasias Pulmonares/patología , Óxidos/farmacología , Sulindac/farmacología , Trióxido de Arsénico , Línea Celular Tumoral , Citometría de Flujo , Inhibidores de Crecimiento/farmacología , Humanos , Peróxido de Hidrógeno/metabolismo
13.
Exp Mol Med ; 38(6): 616-24, 2006 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-17202837

RESUMEN

Trichostatin A (TSA), originally developed as an antifungal agent, is one of potent histone deacetylase (HDAC) inhibitors, which are known to cause growth arrest and apoptosis induction of transformed cells, including urinary bladder, breast, prostate, ovary, and colon cancers. However, the effect of HDAC inhibitors on human non-small cell lung cancer cells is not clearly known yet. Herein, we demonstrated that treatment of TSA resulted in a significant decrease of the viability of H157 cells in a dose-dependent manner, which was revealed as apoptosis accompanying with nuclear fragmentation and an increase in sub-G0/G1 fraction. In addition, it induced the expression of Fas/FasL, which further triggered the activation of caspase-8. Catalytic activation of caspase-9 and decreased expression of anti-apoptotic Bcl-2 and Bcl-XL proteins were observed in TSA-treated cells. Catalytic activation of caspase-3 by TSA was further confirmed by cleavage of pro-caspase-3 and intracellular substrates, including poly (ADP-ribose) polymerase (PARP) and inhibitor of caspase-activated deoxyribonuclease (ICAD). In addition, a characteristic phenomenon of mitochondrial dysfunction, including mitochondrial membrane potential transition and release of mitochondrial cytochrome c into the cytosol was apparent in TSA-treated cells. Taken together, our data indicate that inhibition of HDAC by TSA induces the apoptosis of H157 cells through signaling cascade of Fas/FasL-mediated extrinsic and mitochondria-mediated intrinsic caspases pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Receptores de Muerte Celular/metabolismo , Acetilación , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Catálisis , Línea Celular Tumoral , Activación Enzimática , Histonas/metabolismo , Humanos , Isoformas de Proteínas/metabolismo , Transducción de Señal
15.
Asia Pac J Clin Oncol ; 12(1): e118-24, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24289233

RESUMEN

AIMS: Lung cancer remains the leading cause of cancer mortality worldwide. Despite their poor prognosis, patients with lung cancer are increasingly being admitted to the medical intensive care unit (MICU) for treatment of critical illnesses. The aim of this study was to assess the outcome of patients with lung cancer who are admitted to an MICU and to identify the measurable predictors of their MICU outcome. METHODS: We conducted retrospective analysis on 97 patients with lung cancer admitted to the MICU between 2007 and 2011. RESULTS: The mean age ± standard deviation was 71.8 ± 6.8 years. Of the 97 patients (82 male), 73 patients (75%) had non-small cell lung cancer stage IIIB, IV and 24 patients (25%) had small cell lung cancer. The intensive care unit mortality and in-hospital mortality rates were 53.6 and 61.8%. The main reasons for MICU admission were pneumonia (n = 51) and complication of cancer management (n = 45). The predictors of poor MICU outcome were history of diabetes mellitus (P = 0.028), Acute Physiology and Chronic Health Evaluation II score (P = 0.018), need for mechanical ventilation (P = 0.014), use of vasoactive agents (P < 0.0001), the presence of acute renal failure (P < 0.0001) and presence of multiorgan failure (P < 0.0001). CONCLUSIONS: We found that in-hospital mortality was not influenced by age, sex or performance status score of patients with lung cancer but increased with the severity of organ failure at MICU admission.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Mortalidad Hospitalaria , Unidades de Cuidados Intensivos/estadística & datos numéricos , Neoplasias Pulmonares/mortalidad , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Anciano , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Carcinoma Pulmonar de Células Pequeñas/complicaciones , Carcinoma Pulmonar de Células Pequeñas/patología
16.
Oncotarget ; 7(35): 57213-57227, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27528025

RESUMEN

The non-steroidal anti-inflammatory drugs (NSAIDs) celecoxib and sulindac have been reported to suppress lung cancer migration and invasion. The class III deacetylase sirtuin 1 (SIRT1) possesses both pro- and anticarcinogenic properties. However, its role in inhibition of lung cancer cell epithelial-mesenchymal transition (EMT) by NSAIDs is not clearly known. We attempted to investigate the potential use of NSAIDs as inhibitors of TGF-ß1-induced EMT in A549 cells, and the underlying mechanisms of suppression of lung cancer migration and invasion by celecoxib and sulindac. We demonstrated that celecoxib and sulindac were effective in preventing TGF-ß1-induced EMT, as indicated by upregulation of the epithelial marker, E-cadherin, and downregulation of mesenchymal markers and transcription factors. Moreover, celecoxib and sulindac could inhibit TGF-ß1-enhanced migration and invasion of A549 cells. SIRT1 downregulation enhanced the reversal of TGF-ß1-induced EMT by celecoxib or sulindac. In contrast, SIRT1 upregulation promoted TGF-ß1-induced EMT. Taken together, these results indicate that celecoxib and sulindac can inhibit TGF-ß1-induced EMT and suppress lung cancer cell migration and invasion via downregulation of SIRT1. Our findings implicate overexpressed SIRT1 as a potential therapeutic target to reverse TGF-ß1-induced EMT and to prevent lung cancer cell migration and invasion.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Celecoxib/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Pulmonares/patología , Sirtuina 1/metabolismo , Sulindac/farmacología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Células A549 , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Celecoxib/administración & dosificación , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Invasividad Neoplásica , Sulindac/administración & dosificación
17.
Tumori ; 101(3): e96-8, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-25908039

RESUMEN

First-line epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) for patients with advanced non-small cell lung cancer who were selected on the basis of EGFR mutations have improved the progression-free survival with acceptable toxicity compared to standard chemotherapy. However, acquired resistance to EGFR-TKIs has been identified as an important clinical problem. Transformation to small cell lung cancer (SCLC) is a rare mechanism of resistance to EGFR-TKI therapy. We describe the case of a 61-year-old man who presented transformation from adenocarcinoma to SCLC as the manifestation of acquired resistance after EGFR-TKI treatment. He underwent chemotherapy with etoposide and cisplatin and achieved a complete response.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos , Receptores ErbB/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Cisplatino/administración & dosificación , Etopósido/administración & dosificación , Humanos , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad , Inducción de Remisión , Carcinoma Pulmonar de Células Pequeñas/genética
18.
Korean J Intern Med ; 30(1): 56-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25589836

RESUMEN

BACKGROUND/AIMS: Pleuropulmonary paragonimiasis produces no specific symptoms or radiologic findings, allowing for the possibility of misdiagnosis. We evaluated the specific clinical and pleural fluid features of pleuropulmonary paragonimiasis masquerading as pleural tuberculosis. METHODS: We retrospectively analyzed the clinical and radiologic characteristics of 20 patients diagnosed with pleuropulmonary paragonimiasis between 2001 and 2011. RESULTS: In total, 17 patients presented with respiratory symptoms, including dyspnea (30%), hemoptysis (20%), cough (20%), and pleuritic chest pain (15%). Chest radiographs revealed intrapulmonary parenchymal lesions, including air-space consolidation (30%), nodular opacities (20%), cystic lesions (15%), ground-glass opacities (10%), and pneumothorax (5%). A pleural fluid examination revealed eosinophilia, low glucose levels, and high lactate dehydrogenase (LDH) levels in 87%, 76%, and 88% of the patients, respectively. These traits helped to distinguish pleuropulmonary paragonimiasis from other pleural diseases such as parapneumonic effusion, malignancy, and pleural tuberculosis. CONCLUSIONS: Pleuropulmonary paragonimiasis is often initially misdiagnosed as other pleural diseases. Therefore, it is important to establish the correct diagnosis. In patients with unexplained pleural effusion living in paragonimiasis-endemic areas, pleural fluid obtained by thoracentesis should be examined to distinguish pleuropulmonary paragonimiasis. When marked eosinophilia, high LDH levels, and low glucose levels are identified in pleural fluid, physicians could consider a diagnosis of pleuropulmonary paragonimiasis.


Asunto(s)
Enfermedades Pulmonares Parasitarias/diagnóstico , Paragonimiasis/diagnóstico , Paragonimus westermani/aislamiento & purificación , Derrame Pleural/diagnóstico , Tuberculosis Pleural/diagnóstico , Adolescente , Adulto , Anciano , Animales , Biomarcadores/análisis , Niño , Preescolar , Diagnóstico Diferencial , Ensayo de Inmunoadsorción Enzimática , Eosinofilia/diagnóstico , Eosinofilia/parasitología , Femenino , Glucosa/análisis , Humanos , L-Lactato Deshidrogenasa/análisis , Enfermedades Pulmonares Parasitarias/diagnóstico por imagen , Enfermedades Pulmonares Parasitarias/metabolismo , Enfermedades Pulmonares Parasitarias/parasitología , Masculino , Persona de Mediana Edad , Paracentesis , Paragonimiasis/diagnóstico por imagen , Paragonimiasis/metabolismo , Paragonimiasis/parasitología , Derrame Pleural/diagnóstico por imagen , Derrame Pleural/metabolismo , Derrame Pleural/parasitología , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Tomografía Computarizada por Rayos X , Adulto Joven
19.
Cancer Res Treat ; 47(1): 90-100, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25520153

RESUMEN

PURPOSE: Non-steroidal anti-inflammatory drugs (NSAIDs) and statins are potential chemopreventive or chemotherapeutic agents. The mechanism underlying the deregulation of survivin by NSAIDs and statins in human non-small cell lung cancer cells has not been elucidated. In this study, we investigated the synergistic interaction of sulindac and simvastatin in lung cancer A549 cells. MATERIALS AND METHODS: Cell viability was measured by an MTT assay, while the expression of apoptotic markers, AKT, and survivin in response to sulindac and simvastatin was examined by Western blotting. DNA fragmentation by apoptosis was analyzed by flow cytometry in A549 cells. Reactive oxygen species (ROS) generation was measured by flow cytometry using H2DCFDA and MitoSOX Red, and the effects of pretreatment with N-acetylcysteine were tested. The effects of AKT on survivin expression in sulindac- and simvastatin-treated cells were assessed. Survivin was knocked down or overexpressed to determine its role in apoptosis induced by sulindac and simvastatin. RESULTS: Sulindac and simvastatin synergistically augmented apoptotic activity and intracellular ROS production in A549 cells. Inhibition of AKT by siRNA or LY294002 inhibited survivin, while AKT overexpression markedly increased survivin expression, even in the presence of sulindac and simvastatin. Moreover, survivin siRNA enhanced sulindac- and simvastatininduced apoptosis. In contrast, survivin upregulation protected against sulindac- and simvastatin-induced apoptosis. CONCLUSION: Combined treatment with sulindac and simvastatin augmented their apoptotic potential in lung cancer cells through AKT signaling-dependent downregulation of survivin. These results indicate that sulindac and simvastatin may be clinically promising therapies for the prevention of lung cancer.

20.
Tuberc Respir Dis (Seoul) ; 78(3): 272-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26175784

RESUMEN

Malignant melanoma occurs most frequently on the skin. However, it can also arise in other organs and tissues of the body. Primary pulmonary malignant melanoma is a very rare non-epithelial neoplasm accounting for 0.01% of all primary pulmonary tumors. The treatment of choice is surgical resection of the tumor with an oncologically adequate margin as in lobectomy or pneumonectomy. The prognosis of this condition is rather poor. Based on previous data, its 5-year survival is at least 10%. Here, we report a case of an 82-year-old woman whose primary pulmonary melanoma was detected incidentally.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA