Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Molecules ; 28(13)2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37446894

RESUMEN

Selenium is a main group element and an essential trace element in human health. It was discovered in selenocysteine (SeC) by Stadtman in 1974. SeC is an encoded natural amino acid hailed as the 21st naturally occurring amino acid (U) present in several enzymes and which exquisitely participates in redox biology. As it turns out, selenium bears a U-shaped toxicity curve wherein too little of the nutrient present in biology leads to disorders; concentrations that are too great, on the other hand, pose toxicity to biological systems. In light of many excellent previous reviews and the corpus of literature, we wanted to offer this current review, in which we present aspects of the clinical and biological literature and justify why we should further investigate Se-containing species in biological and medicinal contexts, especially small molecule-containing species in biomedical research and clinical medicine. Of central interest is how selenium participates in biological signaling pathways. Several clinical medical cases are recounted; these reports are mainly pertinent to human cancer and changes in pathology and cases in which the patients are often terminal. Selenium was an option chosen in light of earlier chemotherapeutic treatment courses which lost their effectiveness. We describe apoptosis, and also ferroptosis, and senescence clearly in the context of selenium. Other contemporary issues in research also compelled us to form this review: issues with CoV-2 SARS infection which abound in the literature, and we described findings with human patients in this context. Laboratory scientific studies and clinical studies dealing with two main divisions of selenium, organic (e.g., methyl selenol) or inorganic selenium (e.g., sodium selenite), are discussed. The future seems bright with the research and clinical possibilities of selenium as a trace element, whose recent experimental clinical treatments have so far involved dosing simply and inexpensively over a set of days, amounts, and time intervals.


Asunto(s)
COVID-19 , Selenio , Oligoelementos , Humanos , Selenio/farmacología , Selenio/metabolismo , Oxidación-Reducción , Selenocisteína/metabolismo , Estrés Oxidativo , Apoptosis , Transducción de Señal
2.
Int J Mol Sci ; 22(21)2021 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-34769276

RESUMEN

Functions of selenium are diverse as antioxidant, anti-inflammation, increased immunity, reduced cancer incidence, blocking tumor invasion and metastasis, and further clinical application as treatment with radiation and chemotherapy. These functions of selenium are mostly related to oxidation and reduction mechanisms of selenium metabolites. Hydrogen selenide from selenite, and methylselenol (MSeH) from Se-methylselenocyteine (MSeC) and methylseleninicacid (MSeA) are the most reactive metabolites produced reactive oxygen species (ROS); furthermore, these metabolites may involve in oxidizing sulfhydryl groups, including glutathione. Selenite also reacted with glutathione and produces hydrogen selenide via selenodiglutathione (SeDG), which induces cytotoxicity as cell apoptosis, ROS production, DNA damage, and adenosine-methionine methylation in the cellular nucleus. However, a more pronounced effect was shown in the subsequent treatment of sodium selenite with chemotherapy and radiation therapy. High doses of sodium selenite were effective to increase radiation therapy and chemotherapy, and further to reduce radiation side effects and drug resistance. In our study, advanced cancer patients can tolerate until 5000 µg of sodium selenite in combination with radiation and chemotherapy since the half-life of sodium selenite may be relatively short, and, further, selenium may accumulates more in cancer cells than that of normal cells, which may be toxic to the cancer cells. Further clinical studies of high amount sodium selenite are required to treat advanced cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Selenito de Sodio/uso terapéutico , Glutatión/análogos & derivados , Glutatión/metabolismo , Humanos , Metanol/análogos & derivados , Metanol/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Compuestos de Organoselenio/metabolismo , Compuestos de Selenio/metabolismo , Selenito de Sodio/metabolismo
3.
Oncol Rep ; 20(3): 557-65, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18695906

RESUMEN

To assess the inhibitory effects of methylselenol on the invasion of murine B16F10 melanoma cells, we carried out in vivo and in vitro experiments using Se-methylselenocysteine (Se-MSC) and selenomethionine (SeMet), respectively. In an animal experiment, the supplementation of drinking water with Se-MSC (4 ppm Se) led to a significant increase in Se levels in the lung, liver and serum in mice. Mice given a mash diet or water supplemented with Se-MSC (2, 4 and 6 ppm Se in the mash diet, and 2 and 4 ppm Se in the drinking water) displayed an almost completely diminished pulmonary metastasis of B16F10 melanoma cells and an enhanced survival, compared to the control mice which were given a basal diet. Treatment with non-cytotoxic concentrations of SeMet (2.5, 5 and 10 microM plus 0.02 U/ml METase, methioninase) induced a substantial decrease in the expression of integrin alphavbeta3, the FN receptor and adhesion ability to vitronectin (VN) and fibronectin (FN) in B16F10 melanoma cells. Moreover, these compounds suppressed gelatinase activity, invasive ability and wound migration in the culture system. SeMet-METase prevented the conversion of pro-MMP-9 to its active form and decreased pro-MMP-2 activities in a zymogram. The pre-treatment of B16F10 melanoma cells with SeMet-METase led to a decrease in pulmonary metastasis and extended survival in mice injected with tumor cells. Collectively, our results indicate that integrin expression is crucial in promoting the metastatic phenotype in murine B16F10 melanoma cells by supporting specific adhesive and invasive properties, suggesting that Se-MSC effectively reduces the metastasis of B16F10 melanoma cells as a nutritional adjuvant. Methylselenol may also contribute to the suppression of integrin expression.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Compuestos de Organoselenio/uso terapéutico , Animales , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisteína/análogos & derivados , Cisteína/farmacología , Suplementos Dietéticos , Femenino , Fibronectinas/metabolismo , Citometría de Flujo , Gelatinasas/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Metanol/análogos & derivados , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Compuestos de Organoselenio/farmacología , Selenocisteína/análogos & derivados , Selenito de Sodio/farmacología , Tasa de Supervivencia , Células Tumorales Cultivadas , Vitronectina/metabolismo , Cicatrización de Heridas/efectos de los fármacos
4.
FEBS Lett ; 581(17): 3303-10, 2007 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-17597617

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a nuclear receptor modulating a variety of biological functions including cancer cell proliferation and differentiation. However, the role of PPARgamma and its ligands in tumor invasion is unclear. To evaluate a possible role for PPARgamma ligands in tumor invasion, we examined whether PPARgamma agonists including pioglitazone, troglitazone, rosiglitazone, and ciglitazone could affect the activity of matrix metalloproteinases (MMPs) in the HT1080 cell line, a well-studied and well-characterized cell line for MMP research. The gelatin zymography assay showed that ciglitazone activated pro-MMP-2 significantly. In addition, ciglitazone increased the expression of MMP-2, which was accompanied by an increase of membrane type 1-MMP (MT1-MMP) expression. The PPARgamma antagonist, GW9662 attenuated the ciglitazone-induced PPARgamma activation but it did not affect the pro-MMP2 activation by ciglitazone, suggesting that the action of ciglitazone on the pro-MMP-2 activation bypassed the PPARgamma pathway. Antioxidants and various inhibitors of signal transduction were used to investigate the mechanism of ciglitazone-induced pro-MMP-2 activation. We found that the sustained production of reactive oxygen species (ROS) was required for pro-MMP-2 activation by ciglitazone. We also found that PB98059, an inhibitor of MEK-ERK, significantly blocked ciglitazone-induced pro-MMP-2 activation and that extracellular signal-regulated kinase (ERK) was hyperphosphorylated by ciglitazone. Moreover, cell invasion was significantly increased by ciglitazone in the HT1080 cell lines, whereas cell motility was not affected. This study suggests that ciglitazone-induced pro-MMP-2 activation increases PPARgamma-independent tumor cell invasion through ROS production and ERK activation in some types of cancer cells.


Asunto(s)
Activación Enzimática/efectos de los fármacos , Precursores Enzimáticos/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosarcoma/patología , Gelatinasas/metabolismo , Metaloendopeptidasas/metabolismo , PPAR gamma/agonistas , Especies Reactivas de Oxígeno/metabolismo , Tiazolidinedionas/farmacología , Fibrosarcoma/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 14 de la Matriz/genética , Invasividad Neoplásica , Células Tumorales Cultivadas
5.
Mol Cancer Ther ; 5(11): 2666-75, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17121913

RESUMEN

Matrix metalloproteinase (MMP)-9 plays a key role in tumor invasion. Inhibitors of MMP-9 were screened from Metasequoia glyptostroboides (Dawn redwood) and one potent inhibitor, isoginkgetin, a biflavonoid, was identified. Noncytotoxic levels of isoginkgetin decreased MMP-9 production profoundly, but up-regulated the level of tissue inhibitor of metalloproteinase (TIMP)-1, an inhibitor of MMP-9, in HT1080 human fibrosarcoma cells. The major mechanism of Ras-dependent MMP-9 production in HT1080 cells was phosphatidylinositol 3-kinase (PI3K)/Akt/nuclear factor-kappaB (NF-kappaB) activation. Expression of dominant-active H-Ras and p85 (a subunit of PI3K) increased MMP-9 activity, whereas dominant-negative forms of these molecules decreased the level of MMP-9. H-Ras did not increase MMP-9 in the presence of a PI3K inhibitor, LY294002, and a NF-kappaB inhibitor, SN50. Further studies showed that isoginkgetin regulated MMP-9 production via PI3K/Akt/NF-kappaB pathway, as evidenced by the findings that isoginkgetin inhibited activities of both Akt and NF-kappaB. PI3K/Akt is a well-known key pathway for cell invasion, and isoginkgetin inhibited HT1080 tumor cell invasion substantially. Isoginkgetin was also quite effective in inhibiting the activities of Akt and MMP-9 in MDA-MB-231 breast carcinomas and B16F10 melanoma. Moreover, isoginkgetin treatment resulted in marked decrease in invasion of these cells. In summary, PI3K/Akt is a major pathway for MMP-9 expression and isoginkgetin markedly decreased MMP-9 expression and invasion through inhibition of this pathway. This suggests that isoginkgetin could be a potential candidate as a therapeutic agent against tumor invasion.


Asunto(s)
Biflavonoides/farmacología , Flavonoides/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Fibrosarcoma/metabolismo , Humanos , Inhibidores de la Metaloproteinasa de la Matriz , Invasividad Neoplásica , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Células Tumorales Cultivadas , Quinasa de Factor Nuclear kappa B
6.
FASEB J ; 16(1): 111-3, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11709494

RESUMEN

The relationship between selenium and signal molecules has not been well elucidated. It was found that physiological concentration of selenite, 3 microM, reduced ASK1 activity and induced PI3-kinase (PI3-K)/Akt pathways in HT1080 cells. Duration of these signal molecules by selenite was much longer than that by growth factors and other stresses. The longer duration time of these signal molecules may be important to maintain normal functions against stresses. Selenite increased cell proliferation through up-regulation of Bcl-2 expression, mitochondrial membrane potential, adenosine triphosphate (ATP) generation, and glucose uptake mediated by PI3-K pathway. High concentration of H2O2 increased an apoptotic signal molecule, ASK1, which resulted in Bcl-2 down-regulation, membrane potential disruption, decreased ATP and glucose uptake, and activation of caspases. However, an antiapoptotic signal molecule, Akt, was activated also by H2O2, but duration of its activation was much shorter. Selenite blocked apoptosis induced by H2O2, which was related to blocking ASK1 and further stimulating PI3-kinase/Akt activities. Selenite blocked mitochondrial membrane potential disruption by 400 mM H2O2. Selenite also blocked caspase-9 and -3 activities and apoptosis induced by 500 microM H2O2, even after mitochondrial membrane potential disruption. These observations demonstrate that selenite increases cell proliferation and maintains cell survival by activating the antiapoptotic signal and blocking the apoptotic signal.


Asunto(s)
Apoptosis/efectos de los fármacos , Peróxido de Hidrógeno/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas , Transducción de Señal/efectos de los fármacos , Selenito de Sodio/farmacología , Adenosina Trifosfato/biosíntesis , Animales , División Celular/efectos de los fármacos , Línea Celular , Activación Enzimática , Glucosa/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , MAP Quinasa Quinasa Quinasa 5 , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas
7.
Mech Ageing Dev ; 123(12): 1597-604, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12470897

RESUMEN

Reactive oxygen species (ROS) during normal metabolism signal cells to stimulate proliferation or to cause cellular damages, depending on a specific concentration. Energy restriction (ER) increases life span in animals, which can explain an effective modulator for reducing oxidative stress. Oxidative stress can result from a decrease in the protection against ROS. The deleterious effects of oxidative stress generally occur after exposure to a relatively high concentration of ROS. Alternatively, it has been suggested that a low concentration of ROS can exert important physiological roles in cellular signaling and proliferation. Signal pathways are crucial for cell survival or death. It is generally acceptable that aged cells have less response to stresses such as ROS than young cells. Oxidative stresses induce JNK and p38 kinase pathways regulated by redox regulatory proteins: thioredoxin and glutathione s-transferase, respectively. Antioxidants such as selenium block apoptosis induced by ROS through blocking apoptotic signal ASK1 and stimulating survival signal Akt activity. Old hepatocytes are more susceptible to ROS-induced apoptosis than young hepatocytes, which is associated with low expression of ERK and Akt kinases. Pharmacological inhibition of ERK and Akt activation in the young cells markedly increase their sensitivity to H(2)O(2), and ER, by preventing loss of ERK and Akt activities, enhances survival of old hepatocytes to a level similar to those of young cells. Expressions of signal pathways such as survival and apoptotic signals can regulate cells' fate and aging process. Further studies on the interaction of signal pathways may change the scientific direction of the study of aging.


Asunto(s)
Envejecimiento/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Animales , Metabolismo Energético/fisiología
8.
Mech Ageing Dev ; 125(5): 351-7, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15130752

RESUMEN

Histone deacetylases (HDACs) such as HDAC1 and HSIR2 have been known to be involved in the regulation of life-span extension. However, its underlying mechanism remains unclear in human. Using the primary human gingival fibroblasts (HGFs) derived from donors of different ages, which exhibit clear features of senescence in aged HGFs, we demonstrated that histone deacetylase, HDAC1 and HSIR2, repressed the ageing through the transcriptional inactivation of p53 and p21 promoters. These results suggest that primary HGFs can be a useful human ageing model, and HDAC1, HSIR2, p53 and p21 may play an important role in ageing process of human beings.


Asunto(s)
Senescencia Celular/fisiología , Fibroblastos/fisiología , Encía/fisiología , Histona Desacetilasas/metabolismo , Sirtuinas/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Células Cultivadas , Senescencia Celular/genética , Regulación de la Expresión Génica/genética , Encía/citología , Histona Desacetilasa 1 , Histona Desacetilasas/genética , Humanos , Regiones Promotoras Genéticas/genética , Sirtuina 1 , Sirtuinas/genética , Transcripción Genética/genética , Proteína p53 Supresora de Tumor/genética
9.
Biochem Pharmacol ; 66(5): 849-58, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12948866

RESUMEN

Alpha-lipoic acid is known to increase insulin sensitivity in vivo and to stimulate glucose uptake into adipose and muscle cells in vitro. In this study, alpha-lipoic acid was demonstrated to stimulate the autophosphorylation of insulin receptor and glucose uptake into 3T3-L1 adipocytes by reducing the thiol reactivity of intracellular proteins. To elucidate mechanism of this effect, role of protein thiol groups and H(2)O(2) in insulin receptor autophosphorylation and glucose uptake was investigated in 3T3-L1 adipocytes following stimulation with alpha-lipoic acid. Alpha-lipoic acid or insulin treatment of adipocytes increased intracellular level of oxidants, decreased thiol reactivity of the insulin receptor beta-subunit, increased tyrosine phosphorylation of the insulin receptor, and enhanced glucose uptake. Alpha-lipoic acid or insulin-stimulated glucose uptake was inhibited (i) by alkylation of intracellular, but not extracellular, thiol groups downstream of insulin receptor activation, and (ii) by diphenylene iodonium at the level of the insulin receptor autophosphorylation. alpha-Lipoic acid also inhibited protein tyrosine phosphatase activity and decreased thiol reactivity of protein tyrosine phosphatase 1B. These findings indicate that oxidants produced by alpha-lipoic acid or insulin are involved in activation of insulin receptor and in inactivation of protein tyrosine phosphatases, which eventually result in elevated glucose uptake into 3T3-L1 adipocytes.


Asunto(s)
Adipocitos/efectos de los fármacos , Antioxidantes/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Receptor de Insulina/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Ácido Tióctico/farmacología , Células 3T3 , Adipocitos/enzimología , Adipocitos/metabolismo , Animales , Glucosa/metabolismo , Insulina/farmacología , Ratones , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores
10.
Mol Cells ; 16(2): 147-53, 2003 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-14651254

RESUMEN

A process termed activation-induced cell death (AICD) is responsible for peripheral T cell tolerance after negative selection of self-reactive T cells, and deletion of hyperactivated T cells following the immune response. Cells in G1 phase of the cell cycle are most susceptible to AICD. We have investigated the relationship between the induction of AICD by phorbol 12-myristate 13-acetate plus ionomycin during the cell cycle and the expression of survivin, an inhibitor of the apoptosis protein (LAP) family. AICD was highly induced in cells of the human T cell line Jurkat E6.1 arrested in G1 phase, whereas survivin was hardly expressed in G1 and instead it was highly expressed in G2/M. Moreover, transient over-expression of survivin in G1 partially blocked the induction of AICD. These results suggest that survivin inhibits the induction of AICD, especially in G1 phase.


Asunto(s)
Muerte Celular/fisiología , Fase G1/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Linfocitos T/inmunología , Carcinógenos/farmacología , Caspasa 3 , Caspasas/metabolismo , Muerte Celular/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis , Ionomicina/farmacología , Células Jurkat , Proteínas de Neoplasias , Survivin , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacología
11.
Mol Cells ; 15(1): 94-101, 2003 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-12661767

RESUMEN

Reactive oxygen species (ROS) are generated during cellular metabolism or by external factors. Recently, it was learned that ROS can stimulate cellular proliferation and act as a second messenger in cellular signaling. We previously reported that hydroxyl radicals might be the signaling molecules. In the present experiment, phenazine methosulfate (PMS) was used to generate superoxide anion intracellularly. Treatment with 3 microM PMS in V79 cells increased cellular proliferation by 50%. PMS also activated the c-Jun N-terminal kinase (JNK) and p38 MAPK, but not extracellular signal-regulated kinase (ERK) 1/2 and ERK5. In particular, increased proliferation was blocked by pretreatment with SB203580 (an inhibitor of p38 MAPK). At the transcriptional level, the phosphorylation of c-Jun and ATF-2, which are mediated by JNK and p38 MAPK, were also increased by treatment with PMS.


Asunto(s)
División Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Especies Reactivas de Oxígeno/farmacología , Superóxidos/farmacología , Factor de Transcripción Activador 2 , Animales , Antioxidantes/farmacología , Ácido Benzoico/farmacología , Línea Celular/citología , Línea Celular/efectos de los fármacos , Cricetinae , Cricetulus , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Deferoxamina/farmacología , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Depuradores de Radicales Libres/farmacología , Imidazoles/farmacología , Quelantes del Hierro/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Pulmón/citología , Metosulfato de Metilfenazonio/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-jun/metabolismo , Piridinas/farmacología , Factores de Transcripción/metabolismo , Vitamina K 3/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
12.
J Biochem Mol Biol ; 36(1): 128-37, 2003 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-12542983

RESUMEN

Matrix metalloproteinases (MMPs), zinc dependent proteolytic enzymes, cleave extracellular matrix (ECM: collagen, laminin, firbronectin, etc) as well as non-matrix substrates (growth factors, cell surface receptors, etc). The deregulation of MMPs is involved in many diseases, such as tumor metastasis, rheumatoid arthritis, and periodontal disease. Metastasis is the major cause of death among cancer patients. In this review, we will focus on the roles of MMPs in tumor metastasis. The process of metastasis involves a cascade of linked, sequential steps that involve multiple host-tumor interactions. Specifically, MMPs are involved in many steps of tumor metastasis. These include tumor invasion, migration, host immune escape, extravasation, angiogenesis, and tumor growth. Therefore, without MMPs, the tumor cell cannot perform successful metastasis. The activities of MMPs are tightly regulated at the gene transcription levels, zymogen activation by proteolysis, and inhibition of active forms by endogenous inhibitors, tissue inhibitor of metalloproteinase (TIMP), and RECK. The detailed regulations of MMPs are described in this review.


Asunto(s)
Metaloproteinasas de la Matriz/fisiología , Metástasis de la Neoplasia/fisiopatología , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/enzimología , Animales , Movimiento Celular , Regulación Enzimológica de la Expresión Génica , Humanos , Neoplasias/inmunología , Neovascularización Patológica/inmunología
13.
Food Chem Toxicol ; 50(7): 2565-74, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22266358

RESUMEN

The clinical use of cisplatin (cis-diamminedichloroplatinum II) has been limited by the frequent emergence of cisplatin-resistant cell populations and numerous other adverse effects. Therefore, new agents are required to improve the therapy and health of cancer patients. Oral administration of ginsenoside Rg3 significantly inhibited tumor growth and promoted the anti-neoplastic efficacy of cisplatin in mice inoculated with CT-26 colon cancer cells. In addition, Rg3 administration remarkably inhibited cisplatin-induced nephrotoxicity, hepatotoxicity and oxidative stress. In cell-based experiments, Rg3 inhibited cisplatin-induced cytotoxicity in LLC-RK1 kidney and NCTC1469 liver cells but not in CT-26 cancer cells and significantly decreased cisplatin-induced intracellular ROS levels in these cells. In normal cells with cytoplasmically localized Nrf2 and negligible levels of HO-1 and NQO-1, Rg3 substantially decreased cisplatin-induced elevation in HO-1/NQO-1 levels and inhibited cisplatin-induced translocation of Nrf2 into the nucleus. In chemoresistant cancer cells with high levels of HO-1/NQO-1 and nuclear Nrf2, both basal and cisplatin-induced levels of HO-1/NQO-1 and nuclear Nrf2 were decreased by Rg3 treatment, thereby enhancing the susceptibility of cancer cells to cisplatin. Collectively, Rg3 promotes the efficacy of cisplatin by inhibiting HO-1 and NQO-1 expression in cancer cells and protects the kidney and liver against tissue damage by preventing cisplatin-induced intracellular ROS generation.


Asunto(s)
Cisplatino/uso terapéutico , Ginsenósidos/farmacología , Hemo-Oxigenasa 1/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/uso terapéutico , Línea Celular , Humanos
15.
J Cell Physiol ; 212(2): 386-400, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17348006

RESUMEN

Melanoma is a highly metastatic cancer resistant to current chemotherapeutic and radiotherapeutic approaches. Several studies have shown that interactions between cancer cells and the extracellular matrix (ECM) are critical for the survival and invasion of metastatic cancer cells. In this study, we examine the effects of methylselenol generated from selenomethionine (SeMet) by methioninase (METase) on cell proliferation, adhesion, and expression of integrins in murine melanoma B16F10 cells, which are metastatic in the lungs of syngeneic C57BL/6J mice. Combined treatment with SeMet-METase decreased the expression of integrins alpha(4), beta(1), alpha(nu), and beta(3), and inhibited melanoma-ECM adhesion. Caspase-mediated apoptosis was induced following loss of cell adherence. Phosphorylation of focal adhesion kinase (FAK) and Akt, related to integrin-mediated survival, were decreased upon treatment with SeMet-METase while phosphorylation of p38, PKC-delta, and IkappaBalpha increased. In the presence of specific inhibitors of p38, PKC-delta, and NF-kappaB, expression of integrins and cell adhesion to ECM were maintained and cell apoptosis was prevented in SeMet-METase-treated melanoma cells. Treatment with caspase inhibitors restored cell viability and blocked poly (ADP-ribose) polymerase (PARP) cleavage, but did not restore integrin expression and cell adhesion to ECMs reduced by SeMet-METase. Based on these results, we propose that combined treatment with SeMet-METase induces caspase-mediated apoptosis in melanoma cells by altering integrin expression and adhesion. Furthermore, activation of p38, PKC-delta, and NF-kappaB is a prerequisite for the down-regulation of integrin expression, followed by detachment-mediated apoptosis.


Asunto(s)
Antineoplásicos/metabolismo , Apoptosis , Liasas de Carbono-Azufre/metabolismo , Caspasas/metabolismo , Integrinas/metabolismo , Melanoma Experimental/metabolismo , Compuestos Organometálicos/metabolismo , Selenometionina/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Liasas de Carbono-Azufre/farmacología , Inhibidores de Caspasas , Adhesión Celular , Ciclo Celular , Proliferación Celular , Forma de la Célula , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Proteínas I-kappa B/metabolismo , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Metanol/análogos & derivados , Ratones , Compuestos de Organoselenio , Fosforilación , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Selenometionina/farmacología , Transducción de Señal , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología , Selenito de Sodio/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Carcinogenesis ; 28(4): 837-47, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17071627

RESUMEN

Selenium, an essential biological trace element, reduces the incidence of cancer. Our previous studies show that selenite inhibits tumor invasion by suppressing the expression of matrix metalloproteinases (MMP) -2 and -9. Methylseleninic acid (MSeA), an immediate precursor of methylselenol, inhibits tumor cell growth in vitro and mammary carcinogenesis in vivo. In this study, we demonstrate that MSeA suppresses pro-MMP-2 activation in a dose-dependent manner induced by 12-O-tetradecanoylphorbol-13-acetate (PMA), and further decreases the invasiveness of HT1080 tumor cells. Membrane type-1-MMP (MT1-MMP) is a crucial element in the process of pro-MMP-2 activation. Pro-MMP-2 binds MT1-MMP, using tissue inhibitor of metalloproteinase-2 (TIMP-2) as an adaptor, by forming a trimolecular complex on the cell surface. MSeA blocked MT1-MMP in a dose-dependent manner, but not TIMP-2 expression. MMP-9 and TIMP-1 levels were not affected by MSeA. Selenite induced a decrease in protein levels of both pro-MMPs -9 and -2, but not active forms of pro-MMP-2. MT1-MMP expression is regulated by NF-kappaB. Our data show that the effect of MSeA on MT1-MMP expression is mediated through suppression of NF-kappaB activity. Methylselenol generated by selenomethionine (SeMet) and methioninase (METase) inhibited pro-MMP-2 activation induced by PMA, confirming the effect of MSeA on pro-MMP-2 activity. Moreover, ROS production induced by PMA was partly decreased in the presence of MSeA. This suppression of ROS production may be related to diminished NF-kappaB activity. Thus, our results suggest that MSeA blocks tumor invasion in vitro via inhibiting pro-MMP-2 activation mediated by suppression of MT1-MMP expression, which is regulated by the NF-kappaB signal pathway.


Asunto(s)
Carcinógenos/farmacología , Fibrosarcoma/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , FN-kappa B/metabolismo , Compuestos de Organoselenio/farmacología , Acetato de Tetradecanoilforbol/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática/efectos de los fármacos , Precursores Enzimáticos/metabolismo , Fibrosarcoma/patología , Humanos , Metaloproteinasa 14 de la Matriz/genética , FN-kappa B/genética , Invasividad Neoplásica , Especies Reactivas de Oxígeno/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
17.
Development ; 134(4): 769-77, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17259304

RESUMEN

Activin/Nodal signaling is essential for germ-layer formation and axial patterning during embryogenesis. Recent evidence has demonstrated that the intra- or extracellular inhibition of this signaling is crucial for ectoderm specification and correct positioning of mesoderm and endoderm. Here, we analyzed the function of Xenopus serum response factor (XSRF) in establishing germ layers during early development. XSRF transcripts are restricted to the animal pole ectoderm in Xenopus early embryos. Ectopic expression of XSRF RNA suppresses mesoderm induction, both in the marginal zone in vivo and caused by Activin/Nodal signals in animal caps. Dominant-negative mutant or antisense morpholino oligonucleotide-mediated inhibition of XSRF function expands the expression of mesendodermal genes toward the ectodermal territory and enhances the inducing activity of the Activin signal. SRF interacts with Smad2 and FAST-1, and inhibits the formation of the Smad2-FAST-1 complex induced by Activin. These results suggest that XSRF might act to ensure proper mesoderm induction in the appropriate region by inhibiting the mesoderm-inducing signals during early embryogenesis.


Asunto(s)
Activinas/metabolismo , Gástrula/metabolismo , Factor de Respuesta Sérica/fisiología , Transducción de Señal , Animales , Ectodermo/química , Embrión no Mamífero , Inducción Embrionaria , Factores de Transcripción Forkhead/metabolismo , Mesodermo/citología , Complejos Multiproteicos , ARN Mensajero/análisis , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Proteína Smad2/metabolismo , Xenopus , Proteínas de Xenopus/metabolismo
18.
J Cell Physiol ; 209(3): 775-85, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16972255

RESUMEN

Matrix Metalloproteinases (MMPs) are crucial enzymes for ultraviolet irradiation-induced photoaging in human skin. Ultraviolet B (UVB) stimulates dermal fibroblasts to increase MMP-1 and -3 expression and extracellular matrix (ECM) degradation in photoaging. We investigated whether phosphatase and tensin homolog (PTEN)/Akt pathway is involved in secretions of MMP-1 and -3 in human dermal fibroblasts. The increase in MMP-1 and -3 expression and secretion occurred along with the increase in PTEN and Akt phosphorylation by UVB irradiation in a dose- and time-dependent manner. However, treatment with a casein kinase 2 inhibitor, 5,6-dichloro-1-beta-D-ribofuranosyl-benzimidazole, inhibited their phosphorylations and MMP-1 and -3 secretions. Transfection of wild-type PTEN (Wt-PTEN) decreased basal and UVB-induced MMP-1 and -3 secretions, as well as activator protein-1 (AP-1) activity, while transfection of small interference RNA of PTEN (siRNA-PTEN), phosphatase-inactive PTEN (C124S-PTEN), or lipid phosphatase-inactive PTEN (G129E-PTEN) increased basal or UVB-induced MMP-1 and -3 secretions and AP-1 activity. Transfection of constitutively active Akt (Myr-Akt) also increased basal or UVB-induced MMP-1 and -3 secretions, as well as AP-1 activity. However, transfection of kinase-inactive Akt (K179M-Akt) decreased their secretions, but showed no significant change of AP-1 activity without UVB irradiation, and a significant increase of AP-1 activity with UVB irradiation. Treatment with the phosphatidylinositol 3-kinase inhibitors, LY294002 or wortmannin, downregulated basal and UVB-induced MMP-1 and -3 secretions. In conclusion, UVB irradiation increases PTEN and Akt phosphorylation in human dermal fibroblasts, and these inhibition of PTEN and activation of Akt by phosphorylation are involved in UVB-induced MMP-1 and -3 secretions partly through upregulation of AP-1 activity.


Asunto(s)
Dermis/citología , Fibroblastos , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Células Cultivadas , Inducción Enzimática , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibroblastos/efectos de la radiación , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Factor de Transcripción AP-1/metabolismo , Rayos Ultravioleta
19.
J Cell Biochem ; 98(4): 851-60, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16453289

RESUMEN

ADP-ribosyltransferases (ARTs) are a family of enzymes that catalyze the covalent transfer of an ADP-ribose moiety, derived from NAD, to an amino acid of an acceptor protein, thereby altering its function. To date, little information is available on the protein target specificity of different ART family members. ART2 is a T-cell-specific transferase, attached to the cell surface by a glycosylphosphatidylinositol (GPI) anchor, and also found in serum. Here we investigated the role of ART2 localization in serum or on the cell surface, or solubilized with detergents or enzymes, on its target protein specificity. We found that detergent solubilization of cell membranes, or release of ART2 by phosphoinositide-specific phospholipase C treatment, altered the ability of ART2 to ADP-ribosylate high or low molecular weight histone proteins. Similarly, soluble recombinant ART2 (lacking the GPI anchor) showed a different histone specificity than did cell-bound ART2. When soluble ART2 was incubated with serum proteins in the presence of [32P]-labeled NAD, several serum proteins were ADP-ribosylated in a thiol-specific manner. Mass spectrometry of labeled proteins identified albumin and transferrin as ADP-ribosylated proteins in serum. Collectively, these studies reveal that the membrane or solution environment of ART2 plays a pivotal role in determining its substrate specificity.


Asunto(s)
ADP Ribosa Transferasas/química , Membrana Celular/enzimología , Linfocitos T/enzimología , ADP Ribosa Transferasas/genética , Adenosina Difosfato/química , Adenosina Difosfato/metabolismo , Animales , Células COS , Chlorocebus aethiops , Ratones , NAD/química , NAD/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Solubilidad , Especificidad por Sustrato/fisiología
20.
Biochem Biophys Res Commun ; 326(1): 7-17, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15567145

RESUMEN

Many fundamental processes, including oncogenesis, have implicated HECT domain proteins with ubiquitin ligase activity. The protein human upstream regulatory element binding protein 1 (hUREB1) is a HECT domain protein whose function is not defined yet. Here, we investigate the function of hUREB1 as a ubiquitin-protein ligase in human colorectal cells. Ectopic expression of the HECT domain of hUREB1 reduces the protein level and transcriptional activity of the p53 tumor suppressor, which is abrogated by the deletion in the HECT domain or point mutations in the essential residues of the HECT domain. The ubiquitination and destabilization of p53 is observed in cells treated with the protease inhibitor MG132, implying that the HECT domain of hUREB1 suppresses the transcriptional activity of p53 through a ubiquitin-dependent degradation pathway. Based on the results of Northern blot analysis, RT-PCR, and immunohistochemical analyses, the over-expression of hUREB1 is associated with colorectal carcinoma. Moreover, protein levels of hUREB1 and p53 were inversely correlated. These findings suggest that hUREB1 can function, at least in part, as a negative regulator of p53 during the colorectal carcinoma progression through the ubiquitination pathway mediated by the HECT domain.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fosfoproteínas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosfoproteínas/química , Fosfoproteínas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA