Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Pharmacol Exp Ther ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38936976

RESUMEN

Estrogen receptor (ER)-negative breast cancers are known to be aggressive and unresponsive to anti-estrogen therapy, and triple negative breast cancers are associated with poor prognosis and metastasis. Thus, new targeted therapies are needed. FOXM1 is abundantly expressed in human cancers and implicated in protecting tumor cells from oxidative stress by reducing the levels of intracellular reactive oxygen species (ROS). Aspirin, a prototypical anti-cancer agent with deleterious side effects, has been modified to release nitric oxide and hydrogen sulfide, called NOSH-aspirin (NOSH-ASA), generating a 'safer' class of new anti-inflammatory agents. We evaluated NOSH-ASA against (ER)-negative breast cancer using cell lines and a xenograft mouse model. NOSH-ASA strongly inhibited growth of MDA-MB-231 and SKBR3 breast cancer cells with low IC50s of 90{plus minus}5 and 82{plus minus}5 nM, respectively, with marginal effects on a normal breast epithelial cell line. NOSH-ASA inhibited cell proliferation, caused G0/G1 phase arrest, increased apoptosis, and was associated with increases in ROS. In MDA-MB-231 cell xenografts, NOSH-ASA reduced tumor size markedly, which was associated with reduced proliferation (decreased PCNA expression), induction of apoptosis (increased TUNEL positive cells), and increased ROS, while NF-kB and FoxM1 that were high in untreated xenografts were significantly reduced. mRNA data for FoxM1, p21 and CyclinD1 corroborated with the respective protein expressions and arrest of cells. Taken together, these molecular events contribute to NOSH-ASA mediated growth inhibition and apoptotic death of (ER)-negative breast cells in vitro and in vivo. Additionally, as a ROS-inducer and FOXM1-inhibitor, NOSH-ASA has potential as a targeted therapy. Significance Statement In this investigation, we examined the cellular effects and xenograft tumor inhibitory potential of NOSH-aspirin, an NO and H2S-donating hybrid, against ER-negative breast cancer, which currently lacks effective therapeutic options. The induction of reactive oxygen species and subsequent downregulation of FOXM1 represents a plausible mechanism contributing to the observed decrease in cell proliferation and concurrent increase in apoptosis. NOSH-ASA demonstrated a remarkable reduction in tumor size by 90% without inducing any observable gross toxicity, underscoring its promising translational potential.

2.
Molecules ; 20(7): 12481-99, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26184135

RESUMEN

Estrogen receptor negative (ER(-)) breast cancer is aggressive, responds poorly to current treatments and has a poor prognosis. The NF-κB signaling pathway is implicated in ER(-) tumorigenesis. Aspirin (ASA) is chemopreventive against ER(+) but not for ER(-) breast cancers. Nitric oxide-releasing aspirin (NO-ASA) is a safer ASA where ASA is linked to an NO-releasing moiety through a spacer. In vitro, we investigated anti-proliferation effects of NO-ASA (para- and meta-isomers) against ER(-) breast cancer cells MDA-MB-231 and SK-BR-23, effects on NF-κB signaling, and reactive oxygen species by standard techniques. In vivo, effects of NO-ASA were evaluated in a mouse xenograft model using MDA-MB-231 cells. p-NO-ASA inhibited the growth of MDA-MB-231 and SK-BR-3 cells at 24 h, the respective IC50s were 13 ± 2 and 17 ± 2 µM; ASA had an IC50 of >3000 µM in both cell lines. The IC50s for m-NO-ASA in MDA-MB-231 and SK-BR-3 were 173 ± 15 and 185 ± 12 µM, respectively, therefore, implying p-NO-ASA as a stronger inhibitor of growth p-NO-ASA reduced cell growth by inhibiting proliferation, inducing apoptosis and causing G0/G1 cell cycle block. Activation of NF-κB was inhibited by both isomers as demonstrated by decreases in NF-κB-DNA binding and luciferase activity at 24 h, However, m-NO-ASA produced transient effects at 3 h such as increased NF-κB-DNA-binding, increased levels of nuclear p50, even though both isomers inhibited IκB degradation. Increase in nuclear p50 by m-NO-ASA was associated with translocation of p50 in to the nucleus as observed by immunoflouresence at 3 h. NO-ASA induced reactive oxygen species (ROS) as evidenced by overall increases in both H2DCFDA (2',7'-dichlorodihydrofluorescein) and DHE (dihydroethidium)-derived fluorescence. Inhibition of ROS by N-acetyl-cysteine reversed the m-NO-ASA-mediated translocation of p50 in to the nucleus. In xenografts, p-NO-ASA inhibited tumor growth by inhibiting proliferation (PCNA and tumor volume), inducing apoptosis (TUNEL positive cells) and reducing NF-κB expression. Both isomers inhibit cancer cells, inhibit NF-κB pathway and induce ROS, and have potential as anticancer compounds.


Asunto(s)
Antineoplásicos/farmacología , Aspirina/análogos & derivados , Neoplasias de la Mama/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , FN-kappa B/antagonistas & inhibidores , Receptor ErbB-2/deficiencia , Receptores de Estrógenos/deficiencia , Animales , Apoptosis/efectos de los fármacos , Aspirina/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Genes Reporteros , Humanos , Concentración 50 Inhibidora , Inyecciones Subcutáneas , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Receptor ErbB-2/genética , Receptores de Estrógenos/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Bioorg Med Chem Lett ; 21(23): 7146-50, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22001089

RESUMEN

Several acetyl-protected hydroxybenzyl diethyl phosphates (EHBPs) that are capable of forming quinone methide intermediates were synthesized and their cell growth inhibitory properties were evaluated in four different human cancer cell lines. Compounds 1, 1a, and 1b, corresponding to (4-acetyloxybenzyl diethylphosphate), (3-methyl-4-acetyloxybenzyl diethylphosphate), and (3-chloro-4-acetyloxybenzyl diethylphosphate), were significantly more potent than compounds 2 and 3, (2-acetyloxybenzyl diethylphosphate) and (3-acetyloxybenzyl diethylphosphate), respectively. Using HT-29 human colon cancer cells, compounds 1 and 3 increased apoptosis, inhibited proliferation, and caused a G(2)/M block in the cell cycle. Our data suggest that these compounds merit further investigation as potential anti-cancer agents.


Asunto(s)
Antineoplásicos , Apoptosis/efectos de los fármacos , Organofosfatos/síntesis química , Organofosfatos/farmacología , Acetilación , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Neoplasias/tratamiento farmacológico , Organofosfatos/química
4.
Cells ; 10(11)2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34831416

RESUMEN

Nitric oxide and its production by iNOS is an established mechanism critical to tumor promotion or suppression. Macrophages have important roles in immunity, development, and progression of cancer and have a controversial role in pro- and antitumoral effects. The tumor microenvironment consists of tumor-associated macrophages (TAM), among other cell types that influence the fate of the growing tumor. Depending on the microenvironment and various cues, macrophages polarize into a continuum represented by the M1-like pro-inflammatory phenotype or the anti-inflammatory M2-like phenotype; these two are predominant, while there are subsets and intermediates. Manipulating their plasticity through programming or reprogramming of M2-like to M1-like phenotypes presents the opportunity to maximize tumoricidal defenses. The dual role of iNOS-derived NO also influences TAM activity by repolarization to tumoricidal M1-type phenotype. Regulatory pathways and immunomodulation achieve this through miRNA that may inhibit the immunosuppressive tumor microenvironment. This review summarizes the classical physiology of macrophages and polarization, iNOS activities, and evidence towards TAM reprogramming with current information in glioblastoma and melanoma models, and the immunomodulatory and therapeutic options using iNOS or NO-dependent strategies.


Asunto(s)
Reprogramación Celular , Macrófagos/enzimología , Macrófagos/patología , Neoplasias/patología , Neoplasias/terapia , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Animales , Plasticidad de la Célula , Humanos
5.
Biochem Pharmacol ; 176: 113899, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32145264

RESUMEN

Nitric oxide (NO) and its pro and anti-tumor activities are dual roles that continue to be debated in cancer biology. The cell situations in the tumor and within the tumor microenvironment also have roles involving NO. In early tumorigenic events, macrophages in the tumor microenvironment promote tumor cell death, and later are reprogramed to support the growth of tumor, through regulatory events involving NO and several stimulatory signals. These two opposing and active phenotypes of tumor associated macrophages known as the M1 or anti-tumorigenic state and M2 or pro-tumorigenic state show differences in metabolic pathways such as glycolysis and arginine utilization, signaling pathways and cytokine induction including iNOS expression, therefore contributing to their function. Polarization of M2 to M1 macrophages, inhibition of M2 state, or reprogramming via NO in combination with other signals may determine or alter tumor kinetics. These strategies and an overview are presented.


Asunto(s)
Macrófagos/metabolismo , Redes y Vías Metabólicas , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Citocinas/metabolismo , Humanos , Activación de Macrófagos , Macrófagos/clasificación , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo
6.
Biochem Pharmacol ; 176: 113857, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32061771

RESUMEN

Pancreatic cancer has poor survival rates and largely ineffective therapies. Aspirin is the prototypical anti-cancer agent but its long-term use is associated with significant side effects. NOSH-aspirin belongs to a new class of anti-inflammatory agents that were designed to be safer alternatives by releasing nitric oxide and hydrogen sulfide. In this study we evaluated the effects of NOSH-aspirin against pancreatic cancer using cell lines and a xenograft mouse model. NOSH-aspirin inhibited growth of MIA PaCa-2 and BxPC-3 pancreatic cancer cells with IC50s of 47 ± 5, and 57 ± 4 nM, respectively, while it did not inhibit growth of a normal pancreatic epithelial cell line at these concentrations. NOSH-aspirin inhibited cell proliferation, caused G0/G1 phase cycle arrest, leading to increased apoptosis. Treated cells displayed increases in reactive oxygen species (ROS) and caspase-3 activity. In MIA PaCa-2 cell xenografts, NOSH-aspirin significantly reduced tumor growth and tumor mass. Growth inhibition was due to reduced proliferation (decreased PCNA expression) and induction of apoptosis (increased TUNEL positive cells). Expressions of ROS, iNOS, and mutated p53 were increased; while that of NF-κB and FoxM1 that were high in vehicle-treated xenografts were significantly inhibited by NOSH-aspirin. Taken together, these molecular events and signaling pathways contribute to NOSH-aspirin mediated growth inhibition and apoptotic death of pancreatic cancer cells in vitro and in vivo.


Asunto(s)
Aspirina/análogos & derivados , Proliferación Celular/efectos de los fármacos , Disulfuros/farmacología , Nitratos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Aspirina/química , Aspirina/farmacología , Caspasa 3/metabolismo , Línea Celular Tumoral , Disulfuros/química , Proteína Forkhead Box M1/metabolismo , Masculino , Ratones Desnudos , Ratones SCID , FN-kappa B/metabolismo , Nitratos/química , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Carga Tumoral/efectos de los fármacos
7.
Mol Cell Biol ; 22(2): 567-77, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11756552

RESUMEN

We have characterized a new member of the mammalian PAK family of serine/threonine kinases, PAK5, which is a novel target of the Rho GTPases Cdc42 and Rac. The kinase domain and GTPase-binding domain (GBD) of PAK5 are most closely related in sequence to those of mammalian PAK4. Outside of these domains, however, PAK5 is completely different in sequence from any known mammalian proteins. PAK5 does share considerable sequence homology with the Drosophila MBT protein (for "mushroom body tiny"), however, which is thought to play a role in development of cells in Drosophila brain. Interestingly, PAK5 is highly expressed in mammalian brain and is not expressed in most other tissues. We have found that PAK5, like Cdc42, promotes the induction of filopodia. In N1E-115 neuroblastoma cells, expression of PAK5 also triggered the induction of neurite-like processes, and a dominant-negative PAK5 mutant inhibited neurite outgrowth. Expression of activated PAK1 caused no noticeable changes in these cells. An activated mutant of PAK5 had an even more dramatic effect than wild-type PAK5, indicating that the morphologic changes induced by PAK5 are directly related to its kinase activity. Although PAK5 activates the JNK pathway, dominant-negative JNK did not inhibit neurite outgrowth. In contrast, the induction of neurites by PAK5 was abolished by expression of activated RhoA. Previous work has shown that Cdc42 and Rac promote neurite outgrowth by a pathway that is antagonistic to Rho. Our results suggest, therefore, that PAK5 operates downstream to Cdc42 and Rac and antagonizes Rho in the pathway, leading to neurite development.


Asunto(s)
Encéfalo/enzimología , Neuritas/ultraestructura , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , ADN Complementario/genética , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Mutación , Neuronas/enzimología , Neuronas/ultraestructura , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Homología de Secuencia de Aminoácido , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas , Proteínas de Unión al GTP rac/metabolismo
8.
Cancer Res ; 63(22): 7613-8, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-14633677

RESUMEN

Nitric oxide (NO)-releasing aspirin (ASA), consisting of a traditional ASA molecule to which a NO-donating moiety is covalently bound, is a promising colon cancer chemopreventive agent. NO-ASA inhibits colon cancer cell growth more potently than ASA by inhibiting cell proliferation and enhancing cell killing. We examined in cultured human colon cancer cells the effect of NO-ASA on the beta-catenin/T-cell factor signaling pathway, nuclear factor-kappaB, and NO synthase 2 and on cyclooxygenase (COX) expression, all presumed to participate in colon carcinogenesis. Besides inhibiting cell growth, NO-ASA inhibited the beta-catenin/T-cell factor signaling pathway (IC(50), 1.1 microM), nuclear factor-kappaB DNA binding (IC(50), 7.5 microM), and NO synthase 2 expression (IC(50), 2 microM). Interestingly, NO-ASA induced COX-2 expression, although it had no effect on COX-1. COX-2 induction was accompanied by increased prostaglandin E(2) production. These effects occurred at NO-ASA concentrations below or near its IC(50) for cell growth (IC(50), 2-50 microM). The metabolism of NO-ASA by these cells is characterized by a rapid deacetylation step and the formation of a conjugate with glutathione. NO-ASA had no effect on intracellular cyclic GMP concentrations. We propose a model incorporating the pleiotropic effects of NO-ASA on cell signaling and postulate that collectively these effects may contribute to its strong chemopreventive effect.


Asunto(s)
Aspirina/análogos & derivados , Aspirina/farmacología , Neoplasias del Colon/prevención & control , Proteínas del Citoesqueleto/fisiología , Isoenzimas/biosíntesis , FN-kappa B/antagonistas & inhibidores , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Transactivadores/fisiología , Factores de Transcripción/fisiología , Aspirina/metabolismo , Núcleo Celular/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , GMP Cíclico/metabolismo , Ciclooxigenasa 2 , Proteínas del Citoesqueleto/antagonistas & inhibidores , ADN de Neoplasias/metabolismo , Células HT29 , Humanos , Proteínas de la Membrana , FN-kappa B/metabolismo , Donantes de Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transactivadores/antagonistas & inhibidores , beta Catenina
9.
Oncogene ; 22(38): 5986-94, 2003 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-12955077

RESUMEN

Inactivation of the retinoblastoma (Rb) tumor suppressor protein is essential for the G1/S transition during mammalian cell cycle progression. Although Rb is inactivated by phosphorylation by cyclins D and E and their associated kinases during cell cycle progression, we find that Rb is inactivated upon apoptotic stimulation by Fas through the mediation of p38 kinase, independent of cyclins and cyclin-dependent kinases (cdks). Inactivation by p38 kinase coincided with increased phosphorylation of Rb leading to dissociation of E2F and increased transcriptional activity; such p38-mediated changes in Rb function occurred only during Fas stimulation but not mitogenic progression. p38 kinase targets Rb preferentially and had minimal effects on p107 and had no effect on p130 function. We also find that phosphorylation site mutants of Rb (PSM7LP and PSM9-Rb) that cannot be inactivated by cdks can be targeted by Fas and p38 kinase, suggesting that Rb inactivation by these kinases is biochemically and functionally distinct. It appears that Rb inactivation is achieved by different kinase cascades in response to mitogenic and apoptotic signals.


Asunto(s)
Apoptosis/fisiología , Proteínas de Ciclo Celular , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Unión al ADN , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mitógenos/farmacología , Proteína de Retinoblastoma/metabolismo , Sitios de Unión , Células Cultivadas , Medio de Cultivo Libre de Suero , Ciclina D , Ciclinas/metabolismo , Factores de Transcripción E2F , Humanos , Células Jurkat/efectos de los fármacos , Células Jurkat/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Mitógenos/metabolismo , Mutación , Fosforilación , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/genética , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Receptor fas/metabolismo , Receptor fas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
10.
Redox Biol ; 6: 334-343, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26335399

RESUMEN

Nitric oxide (NO) is one of the 10 smallest molecules found in nature. It is a simple gaseous free radical whose predominant functions is that of a messenger through cGMP. In mammals, NO is synthesized by the enzyme nitric oxide synthase (NOS) of which there are three isoforms. Neuronal (nNOS, NOS1) and endothelial (eNOS, NOS3) are constitutive calcium-dependent forms of the enzyme that regulate neural and vascular function respectively. The third isoform (iNOS, NOS2), is calcium-independent and is inducible. In many tumors, iNOS expression is high, however, the role of iNOS during tumor development is very complex and quite perplexing, with both promoting and inhibiting actions having been described. This review will aim to summarize the dual actions of iNOS-derived NO showing that the microenvironment of the tumor is a contributing factor to these observations and ultimately to cellular outcomes.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , FN-kappa B/genética , Neoplasias/genética , Óxido Nítrico Sintasa de Tipo II/genética , Animales , Antineoplásicos/uso terapéutico , Efecto Espectador , GMP Cíclico/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , FN-kappa B/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Polimorfismo Genético , Transducción de Señal , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
11.
Drug Des Devel Ther ; 9: 4873-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26346117

RESUMEN

Colorectal cancer (CRC) is the second leading cause of death due to cancer and the third most common cancer in men and women in the USA. Nuclear factor kappa B (NF-κB) is known to be activated in CRC and is strongly implicated in its development and progression. Therefore, activated NF-κB constitutes a bona fide target for drug development in this type of malignancy. Many epidemiological and interventional studies have established nonsteroidal anti-inflammatory drugs (NSAIDs) as a viable chemopreventive strategy against CRC. Our previous studies have shown that several novel hydrogen sulfide-releasing NSAIDs are promising anticancer agents and are safer derivatives of NSAIDs. In this study, we examined the growth inhibitory effect of a novel H2S-releasing naproxen (HS-NAP), which has a repertoire as a cardiovascular-safe NSAID, for its effects on cell proliferation, cell cycle phase transitions, and apoptosis using HT-29 human colon cancer cells. We also investigated its effect as a chemo-preventive agent in a xenograft mouse model. HS-NAP suppressed the growth of HT-29 cells by induction of G0/G1 arrest and apoptosis and downregulated NF-κB. Tumor xenografts in mice were significantly reduced in volume. The decrease in tumor mass was associated with a reduction of cell proliferation, induction of apoptosis, and decreases in NF-κB levels in vivo. Therefore, HS-NAP demonstrates strong anticancer potential in CRC.


Asunto(s)
Anticarcinógenos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/prevención & control , Sulfuro de Hidrógeno/farmacología , FN-kappa B/metabolismo , Naproxeno/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Anticarcinógenos/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Células HT29 , Humanos , Sulfuro de Hidrógeno/metabolismo , Masculino , Ratones Desnudos , Naproxeno/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Drug Des Devel Ther ; 7: 389-96, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23690679

RESUMEN

BACKGROUND: The Wnt/ß-catenin/T cell factor (TCF) signaling pathway is important in the development of nonmelanoma skin cancers (NMSCs). Nitric-oxide-releasing nonsteroidal anti-inflammatory drugs (NO-NSAIDs) are chemopreventive agents consisting of a traditional NSAID attached to an NO-releasing moiety through a chemical spacer. Previously we showed that an aromatic spacer enhanced the potency of a particular NO-NSAID compared to an aliphatic spacer. METHODS: We synthesized an NO-releasing NSAID with an aromatic spacer (flurbiprofen benzyl nitrate, NBS-242), and using the human skin cancer cell line A-431, we evaluated its effects on cell kinetics, Wnt/ß-catenin, cyclin D1, and caspase-3. RESULTS: NBS-242 inhibited the growth of A-431 cancer cells, being ~15-fold more potent than flurbiprofen and up to 5-fold more potent than NO-flurbiprofen with an aliphatic spacer, the half maximal inhibitory concentrations (IC50) for growth inhibition being 60 ± 4 µM, 320 ± 20 µM, and 880 ± 65 µM for NBS-242, NO-flurbiprofen, and flurbiprofen, respectively. This effect was associated with inhibition of proliferation, accumulation of cells in the G0/G1 phase of the cell cycle, and an increase in apoptotic cell population. NBS-242 cleaved ß-catenin both in the cytoplasm and the nucleus of A-431 cells. NBS-242 activated caspase-3 whose activation was reflected in the cleavage of procaspase-3. To test the functional consequence of ß-catenin cleavage, we determined the expression of cyclin D1, a Wnt-response gene. NBS-242 reduced cyclin D1 levels in a concentration dependent manner. CONCLUSION: These findings establish a strong inhibitory effect of NBS-242 in A-431 human epidermoid carcinoma cells. NBS-242 modulates parameters that are important in determining cellular mass.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Flurbiprofeno/análogos & derivados , Nitratos/farmacología , beta Catenina/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Caspasa 3/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Flurbiprofeno/farmacología , Humanos , Óxido Nítrico/fisiología , Vía de Señalización Wnt/efectos de los fármacos
13.
Leuk Res ; 37(10): 1302-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23896061

RESUMEN

Hydrogen sulfide-releasing aspirin (HS-ASA) is a novel compound with potential against cancer. It inhibited the growth of Jurkat T-leukemia cells with an IC50 of 1.9 ± 0.2 µM whereas that of ASA was >5000 µM. It dose-dependently inhibited proliferation and induced apoptosis in these cells, causing a G0/G1 cell cycle arrest. HS-ASA down-regulated ß-catenin protein levels and reduced mRNA and protein expression of ß-catenin/TCF downstream target genes cyclinD1 and c-myc. Aspirin up to 5 mM had no effect on ß-catenin expression. HS-ASA also increased caspase-3 protein levels and dose-dependently increased its activity. These effects were substantially blocked by z-VAD-fmk, a pan-caspase inhibitor.


Asunto(s)
Aspirina/química , Aspirina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Sulfuro de Hidrógeno/química , beta Catenina/genética , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Células Jurkat , beta Catenina/metabolismo
14.
Biochem Pharmacol ; 83(6): 723-32, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22209867

RESUMEN

Hormone-dependent estrogen receptor positive (ER+) breast cancers generally respond well to anti-estrogen therapy. Unfortunately, hormone-independent estrogen receptor negative (ER-) breast cancers are aggressive, respond poorly to current treatments and have a poor prognosis. New approaches and targets are needed for the prevention and treatment of ER- breast cancer. The NF-κB signaling pathway is strongly implicated in ER- tumor genesis, constituting a possible target for treatment. Hydrogen sulfide-releasing aspirin (HS-ASA), a novel and safer derivative of aspirin, has shown promise as an anti-cancer agent. We examined the growth inhibitory effect of HS-ASA via alterations in cell proliferation, cell cycle phase transitions, and apoptosis, using MDA-MB-231 cells as a model of triple negative breast cancer. Tumor xenografts in mice, representing human ER- breast cancer, were evaluated for reduction in tumor size, followed by immunohistochemical analysis for proliferation, apoptosis and expression of NF-κB. HS-ASA suppressed the growth of MDA-MB-231 cells by induction of G(0)/G(1) arrest and apoptosis, down-regulation of NF-κB, reduction of thioredoxin reductase activity, and increased levels reactive oxygen species. Tumor xenografts in mice, were significantly reduced in volume and mass by HS-ASA treatment. The decrease in tumor mass was associated with inhibition of cell proliferation, induction of apoptosis and decrease in NF-κB levels in vivo. HS-ASA has anti-cancer potential against ER- breast cancer and merits further study.


Asunto(s)
Antineoplásicos/farmacología , Aspirina/análogos & derivados , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , FN-kappa B/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Tionas/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Aspirina/química , Aspirina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Sulfuro de Hidrógeno/química , Ratones , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/genética , Transducción de Señal/efectos de los fármacos , Tionas/química , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biochem Pharmacol ; 83(6): 733-40, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22209714

RESUMEN

The balance between phase-I carcinogen-activating and phase-II detoxifying xenobiotic metabolizing enzymes is critical to determining an individual's risk for cancer. We evaluated the effect of Hydrogen sulfide-releasing aspirin (HS-ASA) on xenobiotic metabolizing enzymes in HT-29 human colon and Hepa 1c1c7 mouse liver adenocarcinoma cells and in Wistar rats. HS-ASA inhibited the growth of HT-29 and Hepa 1c1c7 cells, with an IC(50) of 3.2 ± 0.3 µM and 4.2 ± 0.4 µM, respectively. The IC(50) for ASA in both cell lines was greater than 5000 µM at 24h. In these cell lines, HS-ASA caused a dose-dependent increase in activity and expression of the phase-II enzymes glutathione S-transferase (GST) and NAD(P)H:quinoneoxireductase (NQO1). It also caused an increase in UDP-glucuronosyltransferase (UGT) expression. The levels of CYP 1A1 a phase-I enzyme was increased by HS-ASA in both cell lines. Pretreatment of cells with NaF, an esterase inhibitor, abrogated the HS-ASA-mediated increases in NQO1 enzyme activity. HS-ASA increased the protein levels of the transcription factor Nrf2, which is a regulator of the phase-II enzymes. In vivo, HS-ASA at 100mg/kg/day had no effect on rat's weights; it induced a 3.4-fold and 1.4-fold increase in hepatic GST and NQO1 enzyme activities, respectively. GST and NQO1 protein levels were also increased. In contrast to that in cultured cells, CYP 1A1 protein levels were not altered in vivo. Therefore, HS-ASA induces phase-II enzymes, at least in part, through the action of H(2)S and by modulating Nrf2; these effects may be part of its mechanism of action against carcinogenesis.


Asunto(s)
Aspirina/análogos & derivados , Citocromo P-450 CYP1A1/metabolismo , Glutatión Transferasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Fase II de la Desintoxicación Metabólica , Fase I de la Desintoxicación Metabólica , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias/enzimología , Tionas/farmacología , Animales , Aspirina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/metabolismo , Células HT29 , Humanos , Concentración 50 Inhibidora , Hígado/enzimología , Masculino , Ratones , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , Ratas , Ratas Wistar , Fluoruro de Sodio/farmacología
16.
Biochem Pharmacol ; 83(6): 715-22, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22222427

RESUMEN

Hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (HS-NSAIDs) are an emerging novel class of compounds with significant anti-inflammatory properties. They consist of a traditional NSAID to which an H(2)S-releasing moiety is covalently attached. We examined the effects of four different HS-NSAIDs on the growth properties of eleven different human cancer cell lines of six different tissue origins. Human colon, breast, pancreatic, prostate, lung, and leukemia cancer cell lines were treated with HS-aspirin, -sulindac, -iburofen, -naproxen, and their traditional counterparts. HS-NSAIDs inhibited the growth of all cancer cell lines studied, with potencies of 28- to >3000-fold greater than that of their traditional counterparts. HS-aspirin (HS-ASA) was consistently the most potent. HS-NSAIDs inhibited cell proliferation, induced apoptosis, and caused G(0)/G(1) cell cycle block. Metabolism of HS-ASA by colon cells showed that the acetyl group of ASA was hydrolyzed rapidly, followed by hydrolysis of the ester bond linking the salicylate anion to the H(2)S releasing moiety, producing salicylic acid and ADT-OH from which H(2)S is released. In reconstitution studies, ASA and ADT-OH were individually less active than the intact HS-ASA towards cell growth inhibition. Additionally, the combination of these two components representing a fairly close approximation to the intact HS-ASA, was 95-fold less active than the intact HS-ASA for growth inhibition. Taken together, these results demonstrate that HS-NSAIDs have potential anti-growth activity against a wide variety of human cancer cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos/farmacocinética , Aspirina/análogos & derivados , Proliferación Celular/efectos de los fármacos , Inhibidores de Crecimiento/farmacocinética , Sulfuro de Hidrógeno/metabolismo , Ibuprofeno/análogos & derivados , Naproxeno/análogos & derivados , Sulindac/análogos & derivados , Tionas/farmacocinética , Antiinflamatorios no Esteroideos/química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Aspirina/química , Aspirina/farmacocinética , Línea Celular Tumoral , Inhibidores de Crecimiento/química , Humanos , Sulfuro de Hidrógeno/química , Ibuprofeno/química , Ibuprofeno/farmacocinética , Naproxeno/química , Naproxeno/farmacocinética , Sulindac/química , Sulindac/farmacocinética , Tionas/química
17.
Biochem Pharmacol ; 79(12): 1759-71, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20188076

RESUMEN

NO-donating aspirin (NO-ASA, para isomer) has been reported to exhibit strong growth inhibitory effect in Jurkat T-acute lymphoblastic leukemia (T-ALL) cells mediated in part by beta-catenin degradation and caspase activation, but the mechanism(s) still remains unclear. In this study, DNA oligoarrays with 263 genes were used to examine the gene expression profiles relating to stress and drug metabolism, and characterize the stress responses at IC(50) and subIC(50) concentrations of p-NO-ASA (20 and 10microM, respectively) in Jurkat T cells. A total of 22 genes related to heat shock response, apoptosis signaling, detoxifiers and Phase II enzymes, and regulators of cell growth were altered in expression by array analysis based on the expression fold change criteria of > or =1.5-fold or < or =0.65-fold. Real time quantitative RT-PCR confirmed that 20microM p-NO-ASA strongly upregulated the mRNA levels of two heat shock genes HSPA1A (41.5+/-7.01-fold) and HSPA6 (100.4+/-8.11-fold), and FOS (16.2+/-3.2-fold), moderately upregulated HSPH1 (1.71+/-0.43-fold), FMO4 (4.5+/-1.67-fold), CASP9 (1.77+/-0.03-fold), DDIT3 (5.6+/-0.51-fold), and downregulated NF-kappaB1 (0.54+/-0.01-fold) and CCND1 (0.69+/-0.06-fold). Protein levels of Hsp70, the product of HSPA1A, and fos were increased in p-NO-ASA-treated Jurkat T and HT-29 colon cancer cells in a dose-dependent manner. Silencing of Hsp70 enhanced the growth inhibitory effect of p-NO-ASA at low concentrations. The altered gene expression patterns by NO-ASA in Jurkat T cells suggest mechanisms for carcinogen metabolism, anti-proliferative activity and possible chemoprotective activity in T-ALL.


Asunto(s)
Antineoplásicos/farmacología , Aspirina/análogos & derivados , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Leucemia de Células T/tratamiento farmacológico , Antineoplásicos/química , Aspirina/química , Aspirina/farmacología , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Silenciador del Gen , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Células Jurkat , Estructura Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Interferencia de ARN
18.
Biochem Pharmacol ; 80(11): 1641-9, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20797387

RESUMEN

ß-Catenin is a central player of the Wnt signaling pathway that regulates cell-cell adhesion and may promote leukemia cell proliferation. We examined whether JS-K, an NO-donating prodrug, modulates the Wnt/ß-catenin/TCF-4 signaling pathway in Jurkat T-Acute Lymphoblastic Leukemia cells. JS-K inhibited Jurkat T cell growth in a concentration and time-dependent manner. The IC(50)s for cell growth inhibition were 14±0.7 and 9±1.2µM at 24 and 48h, respectively. Treatment of the cells with JS-K for 24h, caused a dose-dependent increase in apoptosis from 16±3.3% at 10µM to 74.8±2% at 100µM and a decrease in proliferation. This growth inhibition was also due, in part, to alterations in the different phases of the cell cycle. JS-K exhibited a dose-dependent cytotoxicity as measured by LDH release at 24h. However, between 2 and 8h, LDH release was less than 20% for any indicated JS-K concentration. The ß-catenin/TCF-4 transcriptional inhibitory activity was reduced by 32±8, 63±5, and 93±2% at 2, 10, and 25µM JS-K, respectively, based on luciferase reporter assays. JS-K reduced nuclear ß-catenin and cyclin D1 protein levels, but cytosolic ß-catenin expression did not change. Based on a time-course assay of S-nitrosylation of proteins by a biotin switch assay, S-nitrsolyation of nuclear ß-catenin was determined to precede its degradation. A comparison of the S-nitrosylated nuclear ß-catenin to the total nuclear ß-catenin showed that ß-catenin protein levels were degraded at 24h, while S-nitrosylation of ß-catenin occurred earlier at 0-6h. The NO scavenger PTIO abrogated the JS-K mediated degradation of ß-catenin demonstrating the need for NO.


Asunto(s)
Compuestos Azo/farmacología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Óxido Nítrico/metabolismo , Piperazinas/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Profármacos/farmacología , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , beta Catenina/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Inhibidores de Crecimiento/farmacología , Humanos , Células Jurkat , Óxido Nítrico/fisiología , Compuestos Nitrosos/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción 4 , Proteínas Wnt/fisiología
19.
Biochem Pharmacol ; 78(10): 1298-304, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19576865

RESUMEN

There is current evidence implicating the Wnt/beta-catenin/TCF pathway in breast cancer. We investigated the effect of para- and meta-positional isomers of nitric oxide-releasing aspirin (NO-ASA), and aspirin (ASA) on MCF-7 human breast cancer cell growth and beta-catenin/TCF signaling. The p- and m-NO-ASA isomers strongly inhibited cell growth and beta-catenin/TCF transcriptional activity compared to ASA; the IC50s for growth inhibition were 57+/-4, 193+/-10 and >5000microM, and for transcriptional inhibition they were 12+/-1.8, 75+/-6.5 and >5000microM for p-, m-NO-ASA and ASA, respectively. p-NO-ASA reduced the expression of Wnt/beta-catenin downstream target gene cyclin D1, and total cellular beta-catenin levels. COX-2 expression was induced by p-NO-ASA, protein kinase C inhibitors reversed this induction. p-NO-ASA blocked the cell cycle transition at S to G2/M phase. These studies suggest a targeted chemopreventive/chemotherapeutic potential for NO-ASA against breast cancer.


Asunto(s)
Anticarcinógenos/farmacología , Aspirina/análogos & derivados , Neoplasias de la Mama/prevención & control , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Factores de Transcripción/biosíntesis , Proteínas Wnt/biosíntesis , beta Catenina/biosíntesis , Anticarcinógenos/química , Aspirina/química , Aspirina/farmacología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina D1/genética , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Inducción Enzimática , Femenino , Genes Reporteros , Humanos , Concentración 50 Inhibidora , Isomerismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción 4 , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Proteínas Wnt/genética , beta Catenina/genética
20.
Plasmid ; 56(1): 24-34, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16545871

RESUMEN

Plasmid pCR1 is a cryptic plasmid harboured by Corynebacterium renale. It is the smallest corynebacterial plasmid known to date. Although its natural host is animal corynebacteria, it can replicate in several strains of soil corynebacteria. It can also replicate in Escherichia coli, in which it is stably maintained. The copy number of pCR1 in this host is higher than that of pUC19, with which it shows unidirectional incompatibility. It is also incompatible with pBK2, a plasmid bearing the common corynebacterial replicon pBL1. Its size is 1488bp, as revealed by DNA sequencing. A total of eight open reading frames (ORF) were detected in this plasmid, the largest of which codes for a putative Rep protein of predicted molecular mass of 21kDa. The plasmid pCR1 can be mobilized by the plasmid R6K from E. coli to other corynebacteria. Sequence analysis revealed the presence of an oriT homologous to that of R64. An E. coli plasmid pKL1 shows more than 90% identity with pCR1. Like many coryenbacterial plasmids, pCR1 also replicates by rolling circle mode.


Asunto(s)
Corynebacterium/genética , Plásmidos/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Escherichia coli/metabolismo , Vectores Genéticos , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Plásmidos/química , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA