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.
Artigo em Inglês | MEDLINE | ID: mdl-38936976

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-26184135

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Aspirina/análogos & derivados , Neoplasias da Mama/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica , NF-kappa B/antagonistas & inibidores , Receptor ErbB-2/deficiência , Receptores de Estrogênio/deficiência , Animais , Apoptose/efeitos dos fármacos , Aspirina/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Genes Reporter , Humanos , Concentração Inibidora 50 , Injeções Subcutâneas , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Nus , NF-kappa B/genética , NF-kappa B/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Receptor ErbB-2/genética , Receptores de Estrogênio/genética , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Bioorg Med Chem Lett ; 21(23): 7146-50, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22001089

RESUMO

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.


Assuntos
Antineoplásicos , Apoptose/efeitos dos fármacos , Organofosfatos/síntese química , Organofosfatos/farmacologia , Acetilação , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Neoplasias/tratamento farmacológico , Organofosfatos/química
4.
Cells ; 10(11)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34831416

RESUMO

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.


Assuntos
Reprogramação Celular , Macrófagos/enzimologia , Macrófagos/patologia , Neoplasias/patologia , Neoplasias/terapia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Animais , Plasticidade Celular , Humanos
5.
Biochem Pharmacol ; 176: 113899, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32145264

RESUMO

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.


Assuntos
Macrófagos/metabolismo , Redes e Vias Metabólicas , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais , Animais , Citocinas/metabolismo , Humanos , Ativação de Macrófagos , Macrófagos/classificação , Neoplasias/patologia , Óxido Nítrico Sintase Tipo II/metabolismo
6.
Biochem Pharmacol ; 176: 113857, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32061771

RESUMO

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.


Assuntos
Aspirina/análogos & derivados , Proliferação de Células/efeitos dos fármacos , Dissulfetos/farmacologia , Nitratos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Aspirina/química , Aspirina/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Dissulfetos/química , Proteína Forkhead Box M1/metabolismo , Masculino , Camundongos Nus , Camundongos SCID , NF-kappa B/metabolismo , Nitratos/química , Óxido Nítrico Sintase Tipo II/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Espécies Reativas de Oxigênio/metabolismo , Carga Tumoral/efeitos dos fármacos
7.
Mol Cell Biol ; 22(2): 567-77, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11756552

RESUMO

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.


Assuntos
Encéfalo/enzimologia , Neuritos/ultraestrutura , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Clonagem Molecular , DNA Complementar/genética , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Mutação , Neurônios/enzimologia , Neurônios/ultraestrutura , Fosforilação , Proteínas Serina-Treonina Quinases/química , Homologia de Sequência de Aminoácidos , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases Ativadas por p21 , Proteínas rac de Ligação ao GTP/metabolismo
8.
Cancer Res ; 63(22): 7613-8, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14633677

RESUMO

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.


Assuntos
Aspirina/análogos & derivados , Aspirina/farmacologia , Neoplasias do Colo/prevenção & controle , Proteínas do Citoesqueleto/fisiologia , Isoenzimas/biossíntese , NF-kappa B/antagonistas & inibidores , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Prostaglandina-Endoperóxido Sintases/biossíntese , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Aspirina/metabolismo , Núcleo Celular/metabolismo , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , GMP Cíclico/metabolismo , Ciclo-Oxigenase 2 , Proteínas do Citoesqueleto/antagonistas & inibidores , DNA de Neoplasias/metabolismo , Células HT29 , Humanos , Proteínas de Membrana , NF-kappa B/metabolismo , Doadores de Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transativadores/antagonistas & inibidores , beta Catenina
9.
Oncogene ; 22(38): 5986-94, 2003 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-12955077

RESUMO

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.


Assuntos
Apoptose/fisiologia , Proteínas de Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Ligação a DNA , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitógenos/farmacologia , Proteína do Retinoblastoma/metabolismo , Sítios de Ligação , Células Cultivadas , Meios de Cultura Livres de Soro , Ciclina D , Ciclinas/metabolismo , Fatores de Transcrição E2F , Humanos , Células Jurkat/efeitos dos fármacos , Células Jurkat/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Mitógenos/metabolismo , Mutação , Fosforilação , Proteína do Retinoblastoma/antagonistas & inibidores , Proteína do Retinoblastoma/genética , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Receptor fas/metabolismo , Receptor fas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno
10.
Redox Biol ; 6: 334-343, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26335399

RESUMO

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.


Assuntos
Regulação Neoplásica da Expressão Gênica , NF-kappa B/genética , Neoplasias/genética , Óxido Nítrico Sintase Tipo II/genética , Animais , Antineoplásicos/uso terapêutico , Efeito Espectador , GMP Cíclico/metabolismo , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , NF-kappa B/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Polimorfismo Genético , Transdução de Sinais , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa