Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Intervalo de año de publicación
1.
Front Pharmacol ; 11: 211, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194425

RESUMEN

Since the early studies of William J. McCormick in the 1950s, vitamin C has been proposed as a candidate for the treatment of cancer. A number of reports have shown that pharmacological concentrations of vitamin C selectively kill cancer cells in vitro and decrease the growth rates of a number of human tumor xenografts in immunodeficient mice. However, up to the date there is still doubt regarding this possible therapeutic role of vitamin C in cancer, mainly because high dose administration in cancer patients has not showed a clear antitumor activity. These apparent controversial findings highlight the fact that we lack information on the interactions that occurs between cancer cells and vitamin C, and if these transformed cells can uptake, metabolize and compartmentalize vitamin C like normal human cells do. The role of SVCTs and GLUTs transporters, which uptake the reduced form and the oxidized form of vitamin C, respectively, has been recently highlighted in the context of cancer showing that the relationship between vitamin C and cancer might be more complex than previously thought. In this review, we analyze the state of art of the effect of vitamin C on cancer cells in vitro and in vivo, and relate it to the capacity of cancer cells in acquiring, metabolize and compartmentalize this nutrient, with its implications on the potential therapeutic role of vitamin C in cancer.

2.
Data Brief ; 25: 103972, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31249848

RESUMEN

The data presented in this article are related to the research paper entitled "Increased expression of mitochondrial sodium-coupled ascorbic acid transporter-2 (mitSVCT2) as a central feature in breast cancer", available in Free Radical Biology and Medicine Journal [1]. In this article, we examined the SVCT2 transporter expression in various breast cancer cell lines using RT-PCR and Western blot assays. In addition, we analyzed the subcellular localization of SVCT2 by immunofluorescence colocalization assays and cellular fractionation experiments. Finally, an analysis of different cancer tissue microarrays immunostained for SVCT2 and imaged by The Human Protein Atlas (https://www.proteinatlas.org) is presented.

3.
J Physiol Biochem ; 75(2): 209-215, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31020623

RESUMEN

The mammary gland increases energy requirements during pregnancy and lactation to support epithelial proliferation and milk nutrients synthesis. Lactose, the principal carbohydrate of the milk, is synthetized in the Golgi of mammary epithelial cells by lactose synthase from glucose and UPD galactose. We studied the temporal changes in the expression of GLUT1 and GLUT8 in mammary gland and their association with lactose synthesis and proliferation in BALB/c mice. Six groups were used: virgin, pregnant at 2 and 17 days, lactating at 2 and 10 days, and weaning at 2 days. Temporal expression of GLUT1 and GLUT8 transporters by qPCR, western blot and immunohistochemistry, and its association with lactalbumin, Ki67, and cytokeratin 18 within mammary tissue was studied, along with subcellular localization. GLUT1 and GLUT8 transporters increased their expression during mammary gland progression, reaching 20-fold increasing in GLUT1 mRNA at lactation (p < 0.05) and 2-fold at protein level for GLUT1 and GLUT8 (p < 0.05 and 0.01, respectively). The temporal expression pattern was shared with cytokeratin 18 and Ki67 (p < 0.01). Endogenous GLUT8 partially co-localized with 58 K protein and α-lactalbumin in mammary tissue and with Golgi membrane-associated protein 130 in isolated epithelial cells. The spatial-temporal synchrony between expression of GLUT8/GLUT1 and alveolar cell proliferation, and its localization in cis-Golgi associated to lactose synthase complex, suggest that both transporters are involved in glucose uptake into this organelle, supporting lactose synthesis.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Aparato de Golgi/metabolismo , Glándulas Mamarias Animales/metabolismo , Animales , Células Epiteliales/inmunología , Femenino , Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Transportador de Glucosa de Tipo 1/genética , Queratina-18/metabolismo , Lactalbúmina/metabolismo , Lactancia , Lactosa/biosíntesis , Lactosa Sintasa/metabolismo , Ratones , Ratones Endogámicos BALB C , Péptidos/metabolismo , Embarazo , ARN Mensajero/metabolismo , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Factores de Tiempo , Destete
4.
Free Radic Biol Med ; 135: 283-292, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30902760

RESUMEN

The potential role of vitamin C in cancer prevention and treatment remains controversial. While normal human cells obtain vitamin C as ascorbic acid, the prevalent form of vitamin C in vivo, the uptake mechanisms by which cancer cells acquire vitamin C has remained unclear. The aim of this study is to characterize how breast cancer cells acquire vitamin C. For this, we determined the expression of vitamin C transporters in normal and breast cancer tissue samples, and in ZR-75, MCF-7, MDA-231 and MDA-468 breast cancer cell lines. At the same time, reduced (AA) and oxidized (DHA) forms of vitamin C uptake experiments were performed in all cell lines. We show here that human breast cancer tissues differentially express a form of SVCT2 transporter, that is systematically absent in normal breast tissues and it is increased in breast tumors. In fact, estrogen receptor negative breast cancer tissue, exhibit the most elevated SVCT2 expression levels. Despite this, our analysis in breast cancer cell lines showed that these cells are not able to uptake ascorbic acid and depend on glucose transporter for the acquisition of vitamin C by a bystander effect. This is consistent with our observations that this form of SVCT2 is completely absent from the plasma membrane and is overexpressed in mitochondria of breast cancer cells, where it mediates ascorbic acid transport. This work shows that breast cancer cells acquire vitamin C in its oxidized form and are capable of accumulated high concentrations of the reduced form. Augmented expression of an SVCT2 mitochondrial form appears to be a common hallmark across all human cancers and might have implications in cancer cells survival capacity against pro-oxidant environments.


Asunto(s)
Neoplasias de la Mama/genética , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Transportadores de Sodio Acoplados a la Vitamina C/genética , Ácido Ascórbico/metabolismo , Neoplasias de la Mama/patología , Efecto Espectador , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células MCF-7 , Mitocondrias/patología , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Sodio/metabolismo
5.
Free Radic Biol Med ; 108: 655-667, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28419867

RESUMEN

The liver has an extraordinary regenerative capacity in response to partial hepatectomy (PHx), which develops with neither tissue inflammation response nor alterations in the whole organism. This process is highly coordinated and it has been associated with changes in glutathione (GSH) metabolism. However, there are no reports indicating ascorbic acid (AA) levels after partial hepatectomy. AA and GSH act integrally as an antioxidant system that protects cells and tissues from oxidative damage and imbalance observed in a variety of diseases that affect the liver. Although rat hepatocytes are able to synthesize AA and GSH, which are the providers of AA for the whole organism, they also acquire AA from extracellular sources through the sodium-coupled ascorbic acid transporter-1 (SVCT1). Here, we show that hepatocytes from rat livers subjected to PHx increase their GSH and AA levels from 1 to 7 days post hepatectomy, whose peaks precede the peak in cell proliferation observed at 3 days post-hepatectomy. The increase in both antioxidants was associated with higher expression of the enzymes involved in their synthesis, such as the modifier subunit of enzyme glutamine cysteine ligase (GCLM), glutathione synthetase (GS), gulonolactonase (GLN) and gulonolactone oxidase (GULO). Importantly, rat hepatocytes, that normally exhibit kinetic evidence indicating only SVCT1-mediated transport of AA, lost more than 90% of their capacity to transport it at day 1 after PHx without evidence of recovery at day 7. This observation was in agreement with loss of SVCT1 protein expression, which was undetectable in hepatocytes as early as 2h after PHx, with partial recovery at day 7, when the regenerated liver weight returns to normal. We conclude that after PHx, rat hepatocytes enhance their antioxidant capacity by increasing GSH and AA levels prior to the proliferative peak. GSH and AA are increased by de novo synthesis, however paradoxically hepatocytes from rat subjected to PHx also suppress their capacity to acquire AA from extracellular sources through SVCT1.


Asunto(s)
Ácido Ascórbico/metabolismo , Glutatión/metabolismo , Hepatocitos/fisiología , Hígado/fisiología , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Animales , Antioxidantes/metabolismo , Proliferación Celular , Regulación de la Expresión Génica , Hepatectomía , Hígado/cirugía , Regeneración Hepática , Oxidación-Reducción , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Transportadores de Sodio Acoplados a la Vitamina C/genética
6.
Free Radic Biol Med ; 85: 183-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25933589

RESUMEN

Ascorbic acid is transported into cells by the sodium-coupled vitamin C transporters (SVCTs). Recently, we obtained evidence of differential regulation of SVCT expression in response to acute oxidative stress in cells from species that differ in their capacity to synthesize vitamin C, with a marked decrease in SVCT1 mRNA and protein levels in rat hepatoma cells that was not observed in human hepatoma cells. To better understand the regulatory aspects involved, we performed a structural and functional analysis of the proximal promoter of the SVCT1 rat gene. We cloned a 1476-bp segment containing the proximal promoter of the rat SVCT1 gene and generated deletion-derived truncated promoters of decreasing sizes and mutant promoters by modification of consensus binding sites for transcription factors by site-directed mutagenesis. We next analyzed their capacity to direct the transcription of a reporter gene after transfection into rat H4IIE and human HepG2 hepatoma cells, in experiments involving the coexpression of transcription factors whose consensus binding sequences are present in the SVCT1 promoter. This analysis revealed the presence of two critical cis-regulatory elements of the transcriptional activity of the rat SVCT1 gene promoter, sites containing consensus sequences for the binding of the transcription factors Bach1 and HNF4 that are not present in equivalent locations in the human SVCT1 gene promoter. Moreover, a consensus site for HNF1 that is crucial for the regulation of the human SVCT1 promoter is present in the SVCT1 rat promoter but has no effect on its transcriptional activity. These findings imply that regulation of vitamin C metabolism in the rat, a species with the capacity to synthesize large amounts of ascorbic acid, may differ from that of humans, a species that must obtain ascorbic acid from the diet through a transport mechanism that depends on proper SVCT1 expression.


Asunto(s)
Secuencias Reguladoras de Ácidos Nucleicos , Transportadores de Sodio Acoplados a la Vitamina C/genética , Animales , Línea Celular Tumoral , Humanos , Regiones Promotoras Genéticas , Ratas , Especificidad de la Especie
7.
Rev Med Chil ; 142(1): 20-6, 2014 Jan.
Artículo en Español | MEDLINE | ID: mdl-24861110

RESUMEN

BACKGROUND: Biliary cholesterol is transported by vesicles and micelles. Cholesterol microcrystals are derived from thermodynamically unstable vesicles. In experimental animals vitamin C deficiency leads to a super-saturation of biliary cholesterol and to the formation of gallstones. AIM: To search for a possible relationship between serum levels of vitamin C and the formation of cholesterol gallstones in patients with cholelithiasis. MATERIAL AND METHODS: Thirteen patients with cholelithiasis and a programmed surgical intervention were treated with 2 g/day of vitamin C per os for two weeks before surgery. Forty nine patients subjected to a cholecystectomy not supplemented with vitamin C were studied as controls. Plasma concentrations of vitamin C and lipid profiles were measured. The cholesterol saturation index, crystallization time, cholesterol and phospholipid content in vesicles and micelles, separated by gel filtration chromatography, were studied in bile samples obtained from the gallbladder. RESULTS: Vitamin C supplementation did not change significantly plasma lipids and bile lipid concentrations. However, in supplemented patients, significant reductions in vesicular cholesterol content (6.5 ± 4.8% compared to 17.9 ± 14.0% in the control group; p < 0.05) and vesicular cholesterol/phospholipid ratio (0.71 ± 0.53 compared to 1.36 ± 1.15 in controls; p < 0.05), were observed. CONCLUSIONS: Vitamin C administration may modify bile cholesterol crystallization process, the first step in cholesterol gallstone formation.


Asunto(s)
Ácido Ascórbico/administración & dosificación , Colelitiasis/etiología , Colesterol/metabolismo , Lípidos/análisis , Ácido Ascórbico/análisis , Ácidos y Sales Biliares/química , Estudios de Casos y Controles , Colelitiasis/química , Colesterol/análisis , Cristalización , Femenino , Humanos , Metabolismo de los Lípidos , Masculino , Micelas , Persona de Mediana Edad
8.
Free Radic Biol Med ; 70: 241-54, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24594434

RESUMEN

Despite the fundamental importance of the redox metabolism of mitochondria under normal and pathological conditions, our knowledge regarding the transport of vitamin C across mitochondrial membranes remains far from complete. We report here that human HEK-293 cells express a mitochondrial low-affinity ascorbic acid transporter that molecularly corresponds to SVCT2, a member of the sodium-coupled ascorbic acid transporter family 2. The transporter SVCT1 is absent from HEK-293 cells. Confocal colocalization experiments with anti-SVCT2 and anti-organelle protein markers revealed that most of the SVCT2 immunoreactivity was associated with mitochondria, with minor colocalization at the endoplasmic reticulum and very low immunoreactivity at the plasma membrane. Immunoblotting of proteins extracted from highly purified mitochondrial fractions confirmed that SVCT2 protein was associated with mitochondria, and transport analysis revealed a sigmoidal ascorbic acid concentration curve with an apparent ascorbic acid transport Km of 0.6mM. Use of SVCT2 siRNA for silencing SVCT2 expression produced a major decrease in mitochondrial SVCT2 immunoreactivity, and immunoblotting revealed decreased SVCT2 protein expression by approximately 75%. Most importantly, the decreased protein expression was accompanied by a concomitant decrease in the mitochondrial ascorbic acid transport rate. Further studies using HEK-293 cells overexpressing SVCT2 at the plasma membrane revealed that the altered kinetic properties of mitochondrial SVCT2 are due to the ionic intracellular microenvironment (low in sodium and high in potassium), with potassium acting as a concentration-dependent inhibitor of SVCT2. We discarded the participation of two glucose transporters previously described as mitochondrial dehydroascorbic acid transporters; GLUT1 is absent from mitochondria and GLUT10 is not expressed in HEK-293 cells. Overall, our data indicate that intracellular SVCT2 is localized in mitochondria, is sensitive to an intracellular microenvironment low in sodium and high in potassium, and functions as a low-affinity ascorbic acid transporter. We propose that the mitochondrial localization of SVCT2 is a property shared across cells, tissues, and species.


Asunto(s)
Ácido Ascórbico/metabolismo , Transporte Biológico/genética , Mitocondrias/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Radicales Libres/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Oxidación-Reducción , ARN Interferente Pequeño , Transportadores de Sodio Acoplados a la Vitamina C/genética
9.
Rev. méd. Chile ; 142(1): 20-26, ene. 2014. graf, tab
Artículo en Español | LILACS | ID: lil-708846

RESUMEN

Background: Biliary cholesterol is transported by vesicles and micelles. Cholesterol microcrystals are derived from thermodynamically unstable vesicles. In experimental animals vitamin C deficiency leads to a super-saturation of biliary cholesterol and to the formation of gallstones. Aim: To search for a possible relationship between serum levels of vitamin C and the formation of cholesterol gallstones in patients with cholelithiasis. Material and Methods: Thirteen patients with cholelithiasis and a programmed surgical intervention were treated with 2 g/day of vitamin C per os for two weeks before surgery. Forty nine patients subjected to a cholecystectomy not supplemented with vitamin C were studied as controls. Plasma concentrations of vitamin C and lipid profiles were measured. The cholesterol saturation index, crystallization time, cholesterol and phospholipid content in vesicles and micelles, separated by gel filtration chromatography, were studied in bile samples obtained from the gallbladder. Results: Vitamin C supplementation did not change significantly plasma lipids and bile lipid concentrations. However, in supplemented patients, significant reductions in vesicular cholesterol content (6.5 ± 4.8% compared to 17.9 ± 14.0% in the control group; p < 0.05) and vesicular cholesterol/phospholipid ratio (0.71 ± 0.53 compared to 1.36 ± 1.15 in controls; p < 0.05), were observed. Conclusions: Vitamin C administration may modify bile cholesterol crystallization process, the first step in cholesterol gallstone formation.


Asunto(s)
Femenino , Humanos , Masculino , Persona de Mediana Edad , Ácido Ascórbico/administración & dosificación , Colelitiasis/etiología , Colesterol/metabolismo , Lípidos/análisis , Ácido Ascórbico/análisis , Ácidos y Sales Biliares/química , Estudios de Casos y Controles , Colelitiasis/química , Colesterol/análisis , Cristalización , Metabolismo de los Lípidos , Micelas
10.
Free Radic Biol Med ; 52(9): 1874-87, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22348976

RESUMEN

Although there is in vivo evidence suggesting a role for glutathione in the metabolism and tissue distribution of vitamin C, no connection with the vitamin C transport systems has been reported. We show here that disruption of glutathione metabolism with buthionine-(S,R)-sulfoximine (BSO) produced a sustained blockade of ascorbic acid transport in rat hepatocytes and rat hepatoma cells. Rat hepatocytes expressed the Na(+)-coupled ascorbic acid transporter-1 (SVCT1), while hepatoma cells expressed the transporters SVCT1 and SVCT2. BSO-treated rat hepatoma cells showed a two order of magnitude decrease in SVCT1 and SVCT2 mRNA levels, undetectable SVCT1 and SVCT2 protein expression, and lacked the capacity to transport ascorbic acid, effects that were fully reversible on glutathione repletion. Interestingly, although SVCT1 mRNA levels remained unchanged in rat hepatocytes made glutathione deficient by in vivo BSO treatment, SVCT1 protein was absent from the plasma membrane and the cells lacked the capacity to transport ascorbic acid. The specificity of the BSO treatment was indicated by the finding that transport of oxidized vitamin C (dehydroascorbic acid) and glucose transporter expression were unaffected by BSO treatment. Moreover, glutathione depletion failed to affect ascorbic acid transport, and SVCT1 and SVCT2 expression in human hepatoma cells. Therefore, our data indicate an essential role for glutathione in controlling vitamin C metabolism in rat hepatocytes and rat hepatoma cells, two cell types capable of synthesizing ascorbic acid, by regulating the expression and subcellular localization of the transporters involved in the acquisition of ascorbic acid from extracellular sources, an effect not observed in human cells incapable of synthesizing ascorbic acid.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Glutatión/metabolismo , Hepatocitos/metabolismo , Neoplasias Hepáticas/metabolismo , Transportadores de Sodio Acoplados a la Vitamina C/metabolismo , Animales , Ácido Ascórbico/administración & dosificación , Secuencia de Bases , Butionina Sulfoximina/farmacología , Carcinoma Hepatocelular/patología , Cartilla de ADN , Glutatión/antagonistas & inhibidores , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/patología , Ratas , Ratas Sprague-Dawley
11.
In. Vivaldi Cichero, Ennio; Hernández Poblete, Glenn. Síndrome de shock: aspectos históricos, hemodinámicos, bioquímicos y clínicos. Santiago de Chile, Universidad de Concepción. Facultad de Ciencias Biológicas, mar. 1995. p.15-48, ilus.
Monografía en Español | LILACS | ID: lil-173374
12.
In. Vivaldi Cichero, Ennio; Hernández Poblete, Glenn. Síndrome de shock: aspectos históricos, hemodinámicos, bioquímicos y clínicos. Santiago de Chile, Universidad de Concepción. Facultad de Ciencias Biológicas, mar. 1995. p.107-43.
Monografía en Español | LILACS | ID: lil-173377
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...