Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Tumour Biol ; 36(3): 1871-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25500969

RESUMEN

The migration and adhesion properties of tumors affect their metastatic rate. In the present study, we investigated carcinoembryonic antigen-related cell adhesion molecule (CEACAM) 1, 5, and 6 expression in high nitric oxide (HNO)-adapted lung cancer cells compared to parent cells. We observed high transcript levels of CEACAM 1 (4S, 4L), CEACAM 5, and CEACAM 6 in HNO cells compared to parent cells. However, the surface expression was low in HNO cells. Interestingly, the intracellular protein levels were high for these three CEACAMs. We confirmed these results with immunohistochemical experiments. Further, the adhesion and migration assays showed reduced clumping in HNO-adapted A549 (A549-HNO) cells and faster migration rates, respectively. These results document the altered adhesion and migration properties of cells adapted to HNO. Further, our studies also indicate a dynamic regulation of CEACAM protein expression and surface transport in HNO cells.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/fisiología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Óxido Nítrico/metabolismo , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/genética
2.
Tumour Biol ; 35(3): 2403-15, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24241898

RESUMEN

Our previous studies demonstrate that A549, a human lung adenocarcinoma line, could be adapted to the free radical nitric oxide (NO([Symbol: see text])). NO([Symbol: see text]) has been shown to be overexpressed in human tumors. The original cell line, A549 (parent), and the newly adapted A549-HNO (which has a more aggressive phenotype) serves as a useful model system to study the role of NO([Symbol: see text]) in tumor biology. It is well known that DNA damage response (DDR) is altered in cancer cells and NO([Symbol: see text]) is known to cause DNA damage. Modulations in molecular mechanisms involved in DNA damage response in A549-HNO cells can provide better insights into the enhanced growth behavior of these cells. Thus, here, we carried out a series of time course experiments by treating A549 and A549-HNO cells with NO([Symbol: see text]) donor and examining levels of proteins involved in the DDR pathway. We observed induced expression of key components of DDR pathway in A549-HNO cells. The HNO cells showed sustained expression of key proteins involved in both nonhomologous end joining (NHEJ) and homologous recombination pathways, whereas parent cells only expressed low levels of NHEJ pathway proteins. Further with prolonged NO([Symbol: see text]) exposure, ATR, Chk1, and p53 were activated and upregulated in HNO cells. Activation of p53 results in inhibition of apoptosis through induced Mcl1 expression. It also leads to cell cycle modulation. Interestingly, several reports show that cancer stem cells have enhanced expression of proteins involved in DNA damage response and also activated an antiapoptotic response. Our results here suggest that our HNO adapted A549 cells have increased activation of DNA damage response pathway proteins which can lead to better DNA repair function. Enhanced DDR leads to activation of antiapoptosis response and modulation in the cell cycle which may lead to better survival of these cells under harsh conditions. Thus, our present investigation further supports the hypothesis that HNO exposure leads to survival of these cells.


Asunto(s)
Adaptación Fisiológica/fisiología , Adenocarcinoma/metabolismo , Apoptosis/fisiología , Daño del ADN , Reparación del ADN/fisiología , Neoplasias Pulmonares/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Daño del ADN/fisiología , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/fisiología
3.
Tumour Biol ; 35(3): 2417-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24241959

RESUMEN

Previous work has shown enhanced survival capacity in high nitric oxide (HNO)-adapted tumor cells. In Part I of this series of manuscripts, we have shown that A549-HNO cells demonstrate an improved growth profile under UV and X-ray radiation treatment. These cells exhibit increased expression of proteins involved in DNA damage recognition and repair pathway, both the non-homologous end joining pathway and homologous recombination. These include Ku80, DNA-PK, XLF ligase and MRN complex proteins. Further, the A549-HNO cells show high levels of ATM, ATR, Chk1 and Chk2, and phospho-p53. Activation of these molecules may lead to cell cycle arrest and apoptosis due to DNA damage. This is observed in parent A549 cells in response to NO donor treatment; however, the A549-HNO cells proliferate and inhibit apoptosis. Cell cycle analysis showed slowed progression through S phase which will allow time for DNA repair. Thus, to better understand the increased growth rate in A549-HNO when compared to the parent cell line A549, we studied molecular mechanisms involved in cell cycle regulation in A549-HNO cells. During the initial time period of NO donor treatment, we observe high levels of cyclin/Cdk complexes involved in regulating various stages of the cell cycle. This would lead to bypass of G1-S and G2-M checkpoints. The HNO cells also show much higher expression of Cdc25A. Cdc25A activates Cdk molecules involved in different phases of the cell cycle. In addition, there is enhanced phosphorylation of the Rb protein in HNO cells. This leads to inactivation of Rb/E2F checkpoint regulating G1-S transition. This may lead to faster progression in S phase. Thus, all of these perturbations in HNO cells lead to accelerated cell cycle progression and a higher growth rate. We also assessed expression of cell cycle inhibitors in HNO cells. Interestingly, the HNO cells show a significant decline in p21CIP1 at initial time points, but with prolonged exposure, the levels were much higher than those of the parent cells. This suggests an initial bypass of cell cycle checkpoints as p21CIP1 can inhibit the activity of all cyclin/Cdk complexes. p21CIP1 is also known to inhibit p53-induced apoptosis. This could be important during later phases of the cell cycle to allow time for repair of damaged DNA and thus better survival of HNO cells.


Asunto(s)
Adaptación Fisiológica/fisiología , Adenocarcinoma/metabolismo , Apoptosis/fisiología , Puntos de Control del Ciclo Celular/fisiología , Reparación del ADN/fisiología , Neoplasias Pulmonares/metabolismo , Óxido Nítrico/metabolismo , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Ciclo Celular/fisiología , Línea Celular Tumoral , Daño del ADN/fisiología , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal/fisiología
4.
Tumour Biol ; 33(3): 739-48, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22407532

RESUMEN

Previously, we demonstrated that A549, a human lung cancer cell line, could be adapted to the free radical nitric oxide (NO●). NO● is known to be over expressed in human tumors. The original cell line, A549 (parent), and the newly adapted A549-HNO (which has a more aggressive phenotype) serve as a useful model system to study the biology of NO●. To see if tumor cells can similarly be adapted to any free radical with the same outcome, herein we successfully adapted A549 cells to high levels of hydrogen peroxide (HHP). A549-HHP, the resulting cell line, was more resistant and grew better then the parent cell line, and showed the following characteristics: (1) resistance to hydrogen peroxide, (2) resistance to NO●, (3) growth with and without hydrogen peroxide, and (4) resistance to doxorubicin. Gene chip analysis was used to determine the global gene expression changes between A549-parent and A549-HHP and revealed significant changes in the expression of over 1,700 genes. This gene profile was markedly different from that obtained from the A549-HNO cell line. The mitochondrial DNA content of the A549-HHP line determined by quantitative PCR favored a change for a more anaerobic metabolic profile. Our findings suggest that any free radical can induce resistance to other free radicals; this is especially important given that radiation therapy and many chemotherapeutic agents exert their effect via free radicals. Utilizing this model system to better understand the role of free radicals in tumor biology will help to develop new therapeutic approaches to treat lung cancer.


Asunto(s)
Adaptación Fisiológica , Adenocarcinoma/metabolismo , Peróxido de Hidrógeno/farmacología , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN Mitocondrial , Doxorrubicina/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética
5.
Tumour Biol ; 32(1): 77-85, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20972659

RESUMEN

The free radical nitric oxide (NO) is over-expressed in many tumors, including head and neck squamous cell carcinomas (HNSCC); however, the role NO plays in tumor pathophysiology is still not well understood. We, herein, report the development of an in vitro model system which can be used to probe the role of NO in the carcinogenesis of HNSCC. Five HNSCC cell lines were adapted to a high NO (HNO) environment by gradually introducing increasing concentrations of DETA-NONOate, a nitrogen-based NO donor, to cell media. The adaptation process was carried out until a sufficiently high enough donor concentration was reached which enabled the HNO cells to survive and grow, but which was lethal to the original, unadapted ("parent") cells. The adapted HNO cells exhibited analogous morphology to the parent cells, but grew better than their corresponding parent cells in normal media, on soft agar, and in the presence of hydrogen peroxide, an oxygen-based free radical donor. These results indicate that the HNO cell lines are unique and possess biologically different properties than the parent cell lines from which they originated. The HNO/parent cell lines developed herein may be used as a model system to better understand the role NO plays in HNSCC carcinogenesis.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , Óxido Nítrico/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Modelos Biológicos , Donantes de Óxido Nítrico/farmacología , Compuestos Nitrosos/farmacología , Oxidantes/farmacología , Células Tumorales Cultivadas
6.
Tumour Biol ; 32(1): 87-98, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20963646

RESUMEN

It is not understood why some head and neck squamous cell carcinomas, despite having identical morphology, demonstrate different tumor aggressiveness, including radioresistance. High levels of the free radical nitric oxide (NO) and increased expression of the NO-producing enzyme nitric oxide synthase (NOS) have been implicated in tumor progression. We previously adapted three human tongue cancer cell lines to high NO (HNO) levels by gradually exposing them to increasing concentrations of an NO donor; the HNO cells grew faster than their corresponding untreated ("parent") cells, despite being morphologically identical. Herein we initially characterize the HNO cells and compare the biological properties of the HNO and parent cells. HNO/parent cell line pairs were analyzed for cell cycle distribution, DNA damage, X-ray and ultraviolet radiation response, and expression of key cellular enzymes, including NOS, p53, glutathione S-transferase-pi (GST-pi), apurinic/apyrimidinic endonuclease-1 (APE1), and checkpoint kinases (Chk1, Chk2). While some of these properties were cell line-specific, the HNO cells typically exhibited properties associated with a more aggressive behavior profile than the parent cells (greater S-phase percentage, radioresistance, and elevated expression of GST-pi/APE1/Chk1/Chk2). To correlate these findings with conditions in primary tumors, we examined the NOS, GST-pi, and APE1 expression in human tongue squamous cell carcinomas. A majority of the clinical samples exhibited elevated expression levels of these enzymes. Together, the results herein suggest cancer cells exposed to HNO levels can develop resistance to free radicals by upregulating protective mechanisms, such as GST-pi and APE1. These upregulated defense mechanisms may contribute to their aggressive expression profile.


Asunto(s)
Carcinoma de Células Escamosas/patología , Óxido Nítrico/metabolismo , Neoplasias de la Lengua/patología , Adaptación Fisiológica/efectos de los fármacos , Western Blotting , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Gutatión-S-Transferasa pi/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Técnicas para Inmunoenzimas , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Compuestos Nitrosos/farmacología , Oxidantes/farmacología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias de la Lengua/tratamiento farmacológico , Neoplasias de la Lengua/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta , Rayos X
7.
Tumour Biol ; 31(4): 267-75, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20480412

RESUMEN

Nitric oxide (NO), a free radical, has been implicated in the biology of human cancers, including breast cancer, yet it is still unclear how NO affects tumor development and propagation. We herein gradually adapted four human breast adenocarcinoma cell lines (BT-20, Hs578T, T-47D, and MCF-7) to increasing concentrations of the NO donor DETA-NONOate up to 600 muM. The resulting model system consisted of a set of fully adapted high nitric oxide ("HNO") cell lines that are biologically different from the "parent" cell lines from which they originated. Although each of the four parent and HNO cell lines had identical morphologic appearance, the HNO cells grew faster than their corresponding parent cells and were resistant to both nitrogen- and oxygen-based free radicals. These cell lines serve as a novel tool to study the role of NO in breast cancer progression and potentially can be used to predict the therapeutic response leading to more efficient therapeutic regimens.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Adenocarcinoma/patología , Neoplasias de la Mama/patología , Donantes de Óxido Nítrico/farmacología , Compuestos Nitrosos/farmacología , Adenocarcinoma/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Peróxido de Hidrógeno/toxicidad , Óxido Nítrico/metabolismo
8.
J Photochem Photobiol B ; 82(3): 180-6, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16388964

RESUMEN

The porphyrazines (pzs) are a class of porphyrin derivatives being studied for their use as optical imaging agents and photodynamic therapy (PDT) anti-tumor agents. A previous study revealed that the anionic pz, 18--of the form H2[pz(An;B4-n)], where A is [S(CH2)3CO2-], B is a fused beta',beta'-diisopropyloxy benzo group, with n=2 (trans)--selectively killed tumor cells, while analogous neutral and positively charged pzs lacked this property. In this report, we compare the properties of a suite of three H2[pz(An;B4-n)] pzs containing the same A and B groups as 18, but differing in their values of n: pzs 4 (n=4) and 11 (n=3), and 18 (n=2, trans) exhibit a progressive variation in charge due to the carboxylates, balance between hydrophobic/hydrophilic character, as well as a progressive variation in the singlet oxygen quantum yield (PhiDelta): PhiDelta (18)>PhiDelta (11)>PhiDelta (4). The biological activity of the pzs was tested in human lung carcinoma (A549) and SV40 transformed embryonic (WI-38 VA13) cell lines. Pzs 4 and 11 exhibited significant toxicity in both tumor and normal cells, while 18 showed selective anti-tumor cell activity in a dose-dependent manner. As the number of net negative charges decreased, the compounds became less toxic to normal cells, and the killing effect observed with these compounds was light independent. These observations indicate that the toxicity may have little to do with singlet oxygen quantum yields, but rather is more dependent on the net number of negative charges a pz contains. The study reported herein presents an example of how the porphyrazines can be easily modified to vary their biological behavior and specifically suggest that anionic porphyrazines pzs with lower n (fewer carboxylates, larger hydrophobic core) are more specific tumor killers, while those with larger n (increased net negative charge) are more potent tumor killers.


Asunto(s)
Antineoplásicos/farmacología , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Porfirinas/química , Espectrometría de Fluorescencia , Espectroscopía Infrarroja Corta , Relación Estructura-Actividad
9.
J Med Chem ; 48(26): 8125-33, 2005 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-16366594

RESUMEN

Porphyrazines (pzs), or tetraazaporphyrins, can be viewed as porphyrinic macrocycles in which the porphyrin meso (CH) groups are replaced by nitrogen atoms; as such, it can be anticipated that pzs would show similar biocompatibility and biodistribution to those of porphyrins. However, distinctive chemical and physical features of the pzs differentiate them from either the porphyrins or phthalocyanines, in particular making them excellent candidates as optical imaging/therapeutic agents. The novelty of the pzs requires that we first determine how specific structures selectively alter biological function, leading to the development of "rules" that will be used to predict future biologically functional pzs. In the first of these studies, we present here a correlation of pz charge with biocompatibility for a suite of three pzs-neutral, negative, and positive. Confocal fluorescence microscopy and proliferation/viability measurements disclose that the three pzs differ in their toxicity, uptake, and localization in A549 human lung adenocarcinoma cells and WI-38 VA13 normal cells. Interestingly, the negatively charged pz exhibits selective dark toxicity in pulmonary adenocarcinoma cells.


Asunto(s)
Porfirinas/metabolismo , Porfirinas/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Compuestos Corona/síntesis química , Compuestos Corona/metabolismo , Compuestos Corona/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Microscopía Confocal , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/farmacología , Porfirinas/síntesis química
10.
J Histochem Cytochem ; 60(9): 683-93, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22723525

RESUMEN

Electrocautery and directed energy devices (DEDs) such as lasers, which are used in surgery, result in tissue damage that cannot be readily detected by traditional histological methods, such as hematoxylin and eosin staining. Alternative staining methods, including 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to stain live tissue, have been reported. Despite providing superior detection of damaged tissue relative to the hematoxylin and eosin (H&E) method, the MTT method possesses a number of drawbacks, most notably that it must be carried out on live tissue samples. Herein, we report the development of a novel staining method, "antigen destruction immunohistochemistry" (ADI), which can be carried out on paraffin-embedded tissue. The ADI method takes advantage of epitope loss to define the area of tissue damage and provides many of the benefits of live tissue MTT staining without the drawbacks inherent to that method. In addition, the authors provide data to support the use of antibodies directed at a number of gene products for use in animal tissue for which there are no species-specific antibodies commercially available, as well as an example of a species-specific direct antibody. Data are provided that support the use of this method in many tissue models, as well as evidence that ADI is comparable to the live tissue MTT method.


Asunto(s)
Antígenos/análisis , Inmunohistoquímica/métodos , Animales , Anticuerpos , Especificidad de Anticuerpos , Antígenos/inmunología , Colorantes , Reacciones Cruzadas , Eosina Amarillenta-(YS) , Fijadores , Formaldehído , Hematoxilina , Calor , Adhesión en Parafina , Desnaturalización Proteica , Pliegue de Proteína , Receptor ErbB-2/análisis , Receptor ErbB-2/inmunología , Coloración y Etiquetado/métodos , Porcinos , Sales de Tetrazolio , Tiazoles , Tirosina/análogos & derivados , Tirosina/análisis , Tirosina/inmunología
11.
ChemMedChem ; 3(4): 552-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18076011

RESUMEN

Gastroesophageal reflux disease (GERD) affects both men and women worldwide, with the most common symptom of GERD being frequent heartburn. If left untreated, more serious diseases including esophagitis and/or esophageal cancer may result. GERD has been commonly held to be the result of gastric acid refluxing into the esophagus. Recent work, however, has shown that there are acid-producing cells in the upper aerodigestive tract. In addition, acid-producing bacteria located within the upper gastrointestinal tract and oral cavity may also be a contributing factor in the onset of GERD. Proton pump inhibitors (PPIs) are commonly prescribed for treating GERD; these drugs are designed to stop the production of gastric acid by shutting down the H(+)/K(+)-ATPase enzyme located in parietal cells. PPI treatment is systemic and therefore significantly different than traditional antacids. Although a popular treatment choice, PPIs exhibit substantial interpatient variability and commonly fail to provide a complete cure to the disease. Recent studies have shown that H(+)/K(+)-ATPases are expressed in tissues outside the stomach, and the effects of PPIs in these nongastric tissues have not been fully explored. Likewise, acid-producing bacteria containing proton pumps are present in both the oral cavity and esophagus, and PPI use may also adversely affect these bacteria. The use of PPI therapy is further complicated by the two philosophical approaches to treating this disease: to treat only symptoms or to treat continuously. The latter approach frequently results in unwanted side effects which may be due to the PPIs acting on nongastric tissues or the microbes which colonize the upper aerodigestive tract.


Asunto(s)
Reflujo Gastroesofágico/tratamiento farmacológico , Antiácidos/uso terapéutico , Ácido Gástrico/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/química , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Antagonistas de los Receptores H2 de la Histamina/uso terapéutico , Humanos , Inhibidores de la Bomba de Protones/uso terapéutico , Receptor de Colecistoquinina B/antagonistas & inhibidores
12.
Met Based Drugs ; 2008: 391418, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18274661

RESUMEN

The porphyrazines (pzs), a class of porphyrin analogues, are being investigated for their potential use as tumor imaging/therapeutic agents. We here examine six peripherally-functionalized M[pz(AnB4-n)] pzs with n=4, 3, or 2 (in a trans conformation) and M = H2 or Zn, where A is an [S((CH2)2O)4Me]2 unit and B is a fused beta,beta'-diisopropyloxybenzo group. Cell viability/proliferation assays and fluorescence microscopy were carried out in both tumor and normal cells. Dark toxicity studies disclosed that four of the compounds exhibited toxicity in both normal and tumor cells; one was nontoxic in both normal and tumor cells, and one was selectively toxic to normal cells. Additionally, three of the pzs showed enhanced photo-induced toxicity with these effects in some cases being observed at treatment concentrations of up to ten-fold lower than that needed for a response in Photofrin. All six compounds were preferentially absorbed by tumor cells, suggesting that they have potential as in vitro diagnostic agents and as aids in the isolation and purification of aberrant cells from pathological specimens. In particular, two promising diagnostic candidates have been identified as part of this work.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA