Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Sci Rep ; 6: 24108, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-27063143

RESUMEN

Mitochondria-targeted compounds are emerging as a new class of drugs that can potentially alter the pathophysiology of those diseases where mitochondrial dysfunction plays a critical role. We have synthesized a novel mitochondria-targeted esculetin (Mito-Esc) with an aim to investigate its effect during oxidative stress-induced endothelial cell death and angiotensin (Ang)-II-induced atherosclerosis in ApoE(-/-) mice. Mito-Esc but not natural esculetin treatment significantly inhibited H2O2- and Ang-II-induced cell death in human aortic endothelial cells by enhancing NO production via AMPK-mediated eNOS phosphorylation. While L-NAME (NOS inhibitor) significantly abrogated Mito-Esc-mediated protective effects, Compound c (inhibitor of AMPK) significantly decreased Mito-Esc-mediated increase in NO production. Notably, Mito-Esc promoted mitochondrial biogenesis by enhancing SIRT3 expression through AMPK activation; and restored H2O2-induced inhibition of mitochondrial respiration. siSIRT3 treatment not only completely reversed Mito-Esc-mediated mitochondrial biogenetic marker expressions but also caused endothelial cell death. Furthermore, Mito-Esc administration to ApoE(-/-) mice greatly alleviated Ang-II-induced atheromatous plaque formation, monocyte infiltration and serum pro-inflammatory cytokines levels. We conclude that Mito-Esc is preferentially taken up by the mitochondria and preserves endothelial cell survival during oxidative stress by modulating NO generation via AMPK. Also, Mito-Esc-induced SIRT3 plays a pivotal role in mediating mitochondrial biogenesis and perhaps contributes to its anti-atherogenic effects.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antioxidantes/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Sirtuina 3/metabolismo , Umbeliferonas/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Angiotensina II/toxicidad , Animales , Antioxidantes/uso terapéutico , Aorta/citología , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apoptosis/efectos de los fármacos , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Noqueados , Microscopía Confocal , NG-Nitroarginina Metil Éster/toxicidad , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Sirtuina 3/antagonistas & inhibidores , Sirtuina 3/genética , Umbeliferonas/química , Umbeliferonas/uso terapéutico
2.
FEBS J ; 282(20): 3971-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26236947

RESUMEN

Recent studies have highlighted the involvement of metadherin (MTDH), an oncogenic protein, in promoting cancer progression, metastasis and chemoresistance in many cancers including mammary carcinomas. However, the molecular regulation of MTDH is still not completely understood. In this study we document that AMP activated protein kinase (AMPK) activation-induced anti-proliferative effects are, in part, mediated by inhibiting MTDH expression in MDA-MB-231 and BT-549 triple negative breast cancer (TNBC) cells. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR), an AMPK activator, caused growth arrest, inhibition of migration and invasion of TNBC cells. Intriguingly, AICAR or metformin treatment resulted in significant downregulation of MTDH expression via inhibiting c-Myc expression. In contrast, treatment of cells with compound C, an inhibitor of AMPK, increased both c-Myc and MTDH expressions in TNBC cells. Also, AMPK activation caused increased glycogen synthase kinase 3ß (GSK3ß) activity by inhibiting the inactive phosphorylation at Ser9, on the one hand, and activation of sirtuin1 (SIRT1) by inhibiting Ser47 phosphorylation, as evidenced by deacetylation of p53, on the other hand. Moreover, AMPK-induced GSK3ß and SIRT1 activities were found to be responsible for inhibiting c-Myc-mediated upregulation of MTDH, as LiCl (an inhibitor of GSK3ß) and EX-527 (an inhibitor of SIRT1) reversed AICAR-mediated downregulation of c-Myc and MTDH expressions. Similar results were observed with siSIRT1 treatment. Furthermore, AICAR and EX-527 treatments caused increased cell death under MTDH-depleted conditions. Finally, we uncovered a novel regulation of MTDH expression and showed that AMPK activation by inducing GSK3ß and SIRT1 downregulates MTDH expression via inhibiting c-Myc in TNBC cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma/metabolismo , Moléculas de Adhesión Celular/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Sirtuina 1/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/química , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/enzimología , Adenocarcinoma/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/enzimología , Moléculas de Adhesión Celular/agonistas , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/química , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hipoglucemiantes/farmacología , Proteínas de la Membrana , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , Proteínas de Unión al ARN , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/química , Sirtuina 1/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/enzimología
3.
Diabetes ; 64(6): 2028-41, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25552600

RESUMEN

Monocyte-to-macrophage differentiation is a critical event that accentuates atherosclerosis by promoting an inflammatory environment within the vessel wall. In this study, we investigated the molecular mechanisms responsible for monocyte-to-macrophage differentiation and, subsequently, the effect of metformin in regressing angiotensin II (Ang-II)-mediated atheromatous plaque formation in ApoE(-/-) mice. AMPK activity was dose and time dependently downregulated during phorbol myristate acetate (PMA)-induced monocyte-to-macrophage differentiation, which was accompanied by an upregulation of proinflammatory cytokine production. Of note, AMPK activators metformin and AICAR significantly attenuated PMA-induced monocyte-to-macrophage differentiation and proinflammatory cytokine production. However, inhibition of AMPK activity alone by compound C was ineffective in promoting monocyte-to-macrophage differentiation in the absence of PMA. On the other hand, inhibition of c-Jun N-terminal kinase activity inhibited PMA-induced inflammation but not differentiation, suggesting that inflammation and differentiation are independent events. In contrast, inhibition of STAT3 activity inhibited both inflammation and monocyte-to-macrophage differentiation. By decreasing STAT3 phosphorylation, metformin and AICAR through increased AMPK activation caused inhibition of monocyte-to-macrophage differentiation. Metformin attenuated Ang-II-induced atheromatous plaque formation and aortic aneurysm in ApoE(-/-) mice partly by reducing monocyte infiltration. We conclude that the AMPK-STAT3 axis plays a pivotal role in regulating monocyte-to-macrophage differentiation and that by decreasing STAT3 phosphorylation through increased AMPK activity, AMPK activators inhibit monocyte-to-macrophage differentiation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aterosclerosis/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Metformina/farmacología , Metformina/uso terapéutico , Monocitos/citología , Monocitos/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ácidos Polimetacrílicos/farmacología
4.
PLoS One ; 9(9): e108890, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25268751

RESUMEN

Statins are increasingly being recognized as anti-cancer agents against various cancers including breast cancer. To understand the molecular pathways targeted by fluvastatin and its differential sensitivity against metastatic breast cancer cells, we analyzed protein alterations in MDA-MB-231 cells treated with fluvastatin using 2-DE in combination with LC-MS/MS. Results revealed dys-regulation of 39 protein spots corresponding to 35 different proteins. To determine the relevance of altered protein profiles with breast cancer cell death, we mapped these proteins to major pathways involved in the regulation of cell-to-cell signaling and interaction, cell cycle, Rho GDI and proteasomal pathways using IPA analysis. Highly interconnected sub networks showed that vimentin and ERK1/2 proteins play a central role in controlling the expression of altered proteins. Fluvastatin treatment caused proteolysis of vimentin, a marker of epithelial to mesenchymal transition. This effect of fluvastatin was reversed in the presence of mevalonate, a downstream product of HMG-CoA and caspase-3 inhibitor. Interestingly, fluvastatin neither caused an appreciable cell death nor did modulate vimentin expression in normal mammary epithelial cells. In conclusion, fluvastatin alters levels of cytoskeletal proteins, primarily targeting vimentin through increased caspase-3- mediated proteolysis, thereby suggesting a role for vimentin in statin-induced breast cancer cell death.


Asunto(s)
Anticolesterolemiantes/toxicidad , Apoptosis/efectos de los fármacos , Electroforesis en Gel Bidimensional , Ácidos Grasos Monoinsaturados/toxicidad , Indoles/toxicidad , Proteoma/análisis , Acilcoenzima A/metabolismo , Neoplasias de la Mama , Caspasa 3/química , Caspasa 3/metabolismo , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Fluvastatina , Humanos , Redes y Vías Metabólicas , Ácido Mevalónico/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Espectrometría de Masas en Tándem , Vimentina/metabolismo
5.
FEBS J ; 281(16): 3719-38, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24964743

RESUMEN

Accumulating evidence from in vitro, in vivo, clinical and epidemiological studies shows promising results for the use of statins against many cancers including breast carcinoma. However, the molecular mechanisms responsible for the anti-proliferative and anti-invasive properties of statins still remain elusive. In this study, we investigated the involvement of nitric oxide, iron homeostasis and antioxidant defence mechanisms in mediating the anti-proliferative and anti-invasive properties of hydrophobic statins in MDA-MB-231, MDA-MB-453 and BT-549 metastatic triple negative breast cancer cells. Fluvastatin and simvastatin significantly increased cytotoxicity which was reversed with mevalonate. Interestingly, fluvastatin downregulated transferrin receptor (TfR1), with a concomitant depletion of intracellular iron levels in these cells. Statin-induced effects were mimicked by geranylgeranyl transferase inhibitor (GGTI-298) but not farnesyl transferase inhibitor (FTI-277). Further, it was observed that TfR1 downregulation is mediated by increased nitric oxide levels via inducible nitric oxide synthase (iNOS) expression. NOS inhibitors (asymmetric dimethylarginine and 1400W) counteracted and sepiapterin, a precursor of tetrahydrobiopterin, exacerbated statin-induced depletion of intracellular iron levels. Notably, fluvastatin increased manganese superoxide dismutase (by repressing the transcription factor DNA damage-binding protein 2), catalase and glutathione which, in turn, diminished H2 O2 levels. Fluvastatin-induced downregulation of TfR1, matrix metalloproteinase-2, -9 and inhibition of invasion were reversed in the presence of aminotriazole, a specific inhibitor of catalase. Finally, we conclude that fluvastatin, by altering iron homeostasis, nitric oxide generation and antioxidant defence mechanisms, induces triple negative breast cancer cell death.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/metabolismo , Proliferación Celular/efectos de los fármacos , Ácidos Grasos Monoinsaturados/farmacología , Indoles/farmacología , Hierro/metabolismo , Óxido Nítrico/metabolismo , Simvastatina/farmacología , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptosis , Transporte Biológico/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Fluvastatina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , Ácido Mevalónico/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Transcripción Genética , Neoplasias de la Mama Triple Negativas
6.
PLoS One ; 9(3): e89351, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24622734

RESUMEN

Although the anti-cancer effects of curcumin has been shown in various cancer cell types, in vitro, pre-clinical and clinical studies showed only a limited efficacy, even at high doses. This is presumably due to low bioavailability in both plasma and tissues, particularly due to poor intracellular accumulation. A variety of methods have been developed to achieve the selective targeting of drugs to cells and mitochondrion. We used a novel approach by conjugation of curcumin to lipophilic triphenylphosphonium (TPP) cation to facilitate delivery of curcumin to mitochondria. TPP is selectively taken up by mitochondria driven by the membrane potential by several hundred folds. In this study, three mitocurcuminoids (mitocurcuminoids-1, 2, and 3) were successfully synthesized by tagging TPP to curcumin at different positions. ESI-MS analysis showed significantly higher uptake of the mitocurcuminoids in mitochondria as compared to curcumin in MCF-7 breast cancer cells. All three mitocurcuminoids exhibited significant cytotoxicity to MCF-7, MDA-MB-231, SKNSH, DU-145, and HeLa cancer cells with minimal effect on normal mammary epithelial cells (MCF-10A). The IC50 was much lower for mitocurcuminoids when compared to curcumin. The mitocurcuminoids induced significant ROS generation, a drop in ΔØm, cell-cycle arrest and apoptosis. They inhibited Akt and STAT3 phosphorylation and increased ERK phosphorylation. Mitocurcuminoids also showed upregulation of pro-apoptotic BNIP3 expression. In conclusion, the results of this study indicated that mitocurcuminoids show substantial promise for further development as a potential agent for the treatment of various cancers.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Curcumina/farmacología , Curcumina/farmacocinética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Disponibilidad Biológica , Transporte Biológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Curcumina/química , Curcumina/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/metabolismo
7.
Indian J Biochem Biophys ; 48(1): 29-34, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21469599

RESUMEN

Bacterial organophosphate hydrolases (OPH) have been shown to hydrolyze structurally diverse group of organophosphate (OP) compounds and nerve agents. Due to broad substrate range and unusual catalytic properties, the OPH has successfully been used to develop eco-friendly strategies for detection and decontamination of OP compounds. However, their usage has failed to gain necessary acceptance, due to short half-life of the enzyme and loss of activity during process development. In the present study, we report a simple procedure for immobilization of OPH on biocompatible gelatin pads. The covalent coupling of OPH using glutaraldehyde spacer has been found to dramatically improve the enzyme stability. There is no apparent loss of OPH activity in OPH-gelatin pads stored at room temperature for more than six months. As revealed by a number of kinetic parameters, the catalytic properties of immobilized enzyme are found to be comparable to the free enzyme. Further, the OPH-gelatin pads effectively eliminate OP insecticide methyl parathion and nerve agent sarin.


Asunto(s)
Enzimas Inmovilizadas/química , Gelatina/química , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Sarín/química , Estabilidad de Enzimas , Escherichia coli/enzimología , Escherichia coli/genética , Hidrólisis , Insecticidas/envenenamiento , Metil Paratión/química , Compuestos Organofosforados/química , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/aislamiento & purificación , Especificidad por Sustrato
8.
Biosens Bioelectron ; 26(2): 689-95, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20674329

RESUMEN

A novel highly sensitive biosensor for the direct and simultaneous determination of superoxide anion radical (O2-) and nitrite (NO2-) was developed by incorporation of carbon nanotube (CNT) solubilized in nafion in polypyrrole (PPy) matrix on Pt electrode followed by immobilization of Cu,ZnSOD (SOD1) on it. The CNT/PPy nanocomposite electrode enhanced the immobilization of SOD1 and promoted the electron transfer of SOD1 minimizing its fouling effect. The surface morphological images of PPy and CNT-PPy nanocomposite on Pt electrode were obtained by scanning electron microscopy exhibiting highly microporous structures. The electrochemical behavior of the biosensor investigated by cyclic voltammetry revealed that the SOD1 immobilized electrode showed characteristic of SOD1 quasi-reversible redox peaks with a formal potential of +0.065 V vs. Ag/AgCl. The biosensor exhibited a linear response over the concentration range from 0.1 to 750 µM, with a detection limit of 0.1±0.03 µM for O2- and a corresponding linear range of 0.5-2000 µM, with a detection limit of 0.5±0.025 µM for NO2-. In addition, the biosensor exhibited high sensitivity, good reproducibility and retained stability over 30 days. This modified electrode was quite effective not only in detecting O2- and NO2- independently but also determining the concentration of O2- and NO2- simultaneously in vitro and from cancer cells.


Asunto(s)
Técnicas Biosensibles/instrumentación , Conductometría/instrumentación , Nanotubos de Carbono/química , Nitritos/análisis , Polímeros/química , Pirroles/química , Superóxido Dismutasa/química , Superóxidos/análisis , Mezclas Complejas/análisis , Enzimas Inmovilizadas/química , Diseño de Equipo , Análisis de Falla de Equipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA