Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Addict Biol ; 27(1): e13107, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34699111

RESUMEN

Hazardous, heavy drinking increases risk for developing alcohol use disorder (AUD), which affects ~7% of adult Americans. Thus, understanding the molecular mechanisms promoting risk for heavy drinking is essential to developing more effective AUD pharmacotherapies than those currently approved by the FDA. Using genome-wide bisulfate sequencing, we identified DNA methylation (DNAm) signals within the nucleus accumbens core (NAcC) that differentiate nonheavy and heavy ethanol-drinking rhesus macaques. One differentially DNAm region (D-DMR) located within the gene neurobeachin (NBEA), which promotes synaptic membrane protein trafficking, was hypermethylated in heavy drinking macaques. A parallel study identified a similar NBEA D-DMR in human NAcC that distinguished alcoholic and nonalcoholic individuals. To investigate the role of NBEA in heavy ethanol drinking, we engineered a viral vector carrying a short hairpin RNA (shRNA) to reduce the expression of NBEA. Using two murine models of ethanol consumption: 4 days of drinking-in-the-dark and 4 weeks of chronic intermittent access, the knockdown of NBEA expression did not alter average ethanol consumption in either model. However, it did lead to a significant increase in the ethanol preference ratio. Following withdrawal, whole-cell patch clamp electrophysiological experiments revealed that Nbea knockdown led to an increase in spontaneous excitatory postsynaptic current amplitude with no alteration in spontaneous inhibitory postsynaptic currents, suggesting a specific role of NBEA in trafficking of glutamatergic receptors. Together, our findings suggest that NBEA could be targeted to modulate the preference for alcohol use.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Alcoholismo/genética , Proteínas Portadoras/genética , Proteínas del Tejido Nervioso/genética , Adulto , Anciano , Animales , Metilación de ADN/efectos de los fármacos , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Núcleo Accumbens/efectos de los fármacos
2.
Elife ; 102021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34494548

RESUMEN

Hypothalamic Kiss1 neurons control gonadotropin-releasing hormone release through the secretion of kisspeptin. Kiss1 neurons serve as a nodal center that conveys essential regulatory cues for the attainment and maintenance of reproductive function. Despite this critical role, the mechanisms that control kisspeptin synthesis and release remain largely unknown. Using Drop-Seq data from the arcuate nucleus of adult mice and in situ hybridization, we identified Nescient Helix-Loop-Helix 2 (Nhlh2), a transcription factor of the basic helix-loop-helix family, to be enriched in Kiss1 neurons. JASPAR analysis revealed several binding sites for NHLH2 in the Kiss1 and Tac2 (neurokinin B) 5' regulatory regions. In vitro luciferase assays evidenced a robust stimulatory action of NHLH2 on human KISS1 and TAC3 promoters. The recruitment of NHLH2 to the KISS1 and TAC3 promoters was further confirmed through chromatin immunoprecipitation. In vivo conditional ablation of Nhlh2 from Kiss1 neurons using Kiss1Cre:Nhlh2fl/fl mice induced a male-specific delay in puberty onset, in line with a decrease in arcuate Kiss1 expression. Females retained normal reproductive function albeit with irregular estrous cycles. Further analysis of male Kiss1Cre:Nhlh2fl/fl mice revealed higher susceptibility to metabolic challenges in the release of luteinizing hormone and impaired response to leptin. Overall, in Kiss1 neurons, Nhlh2 contributes to the metabolic regulation of kisspeptin and NKB synthesis and release, with implications for the timing of puberty onset and regulation of fertility in male mice.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Kisspeptinas/metabolismo , Neuronas/fisiología , Maduración Sexual/fisiología , Animales , Línea Celular , Cromatina , ADN/genética , Estradiol/farmacología , Femenino , Fertilidad , Regulación de la Expresión Génica/efectos de los fármacos , Inmunoprecipitación , Kisspeptinas/genética , Kisspeptinas/farmacología , Leptina/farmacología , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Noqueados , Fragmentos de Péptidos/farmacología , Reacción en Cadena de la Polimerasa/métodos , Factores Sexuales , Sustancia P/análogos & derivados , Sustancia P/farmacología
3.
Environ Health Perspect ; 129(8): 87003, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34383603

RESUMEN

BACKGROUND: The effects of endocrine-disrupting chemicals (EDCs) on fertility and reproductive development represent a rising concern in modern societies. Although the neuroendocrine control of sexual maturation is a major target of EDCs, little is known about the potential role of the hypothalamus in puberty and ovulation disruption transmitted across generations. OBJECTIVES: We hypothesized that developmental exposure to an environmentally relevant dose of EDC mixture could induce multi- and/or transgenerational alterations of sexual maturation and maternal care in female rats through epigenetic reprograming of the hypothalamus. We investigated the transmission of a disrupted reproductive phenotype via the maternal germline or via nongenomic mechanisms involving maternal care. METHODS: Adult female Wistar rats were exposed prior to and during gestation and until the end of lactation to a mixture of the following 13 EDCs: di-n-butyl phthalate (DnBP), di(2-ethylhexyl) phthalate (DEHP), bisphenol A (BPA), vinclozolin, prochloraz, procymidone, linuron, epoxynaxole, dichlorodiphenyldichloroethylene, octyl methoxynimmate, 4-methylbenzylidene camphor (4-MBC), butylparaben, and acetaminophen. Perinatally exposed offspring (F1) were mated with unexposed males to generate germ cell (F2) and transgenerationally exposed (F3 and F4) females. Sexual maturation, maternal behavior, and hypothalamic targets of exposure were studied across generations. RESULTS: Germ cell (F2) and transgenerationally (F3) EDC-exposed females, but not F1, displayed delayed pubertal onset and altered folliculogenesis. We reported a transgenerational alteration of key hypothalamic genes controlling puberty and ovulation (Kiss1, Esr1, and Oxt), and we identified the hypothalamic polycomb group of epigenetic repressors as actors of this mechanism. Furthermore, we found a multigenerational reduction of maternal behavior (F1-F3) induced by a loss in hypothalamic dopaminergic signaling. Using a cross-fostering paradigm, we identified that the reduction in maternal phenotype was normalized in EDC-exposed pups raised by unexposed dams, but no reversal of the pubertal phenotype was achieved. DISCUSSION: Rats developmentally exposed to an EDC mixture exhibited multi- and transgenerational disruption of sexual maturation and maternal care via hypothalamic epigenetic reprogramming. These results raise concerns about the impact of EDC mixtures on future generations. https://doi.org/10.1289/EHP8795.


Asunto(s)
Disruptores Endocrinos , Hipotálamo/efectos de los fármacos , Conducta Materna/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Animales , Disruptores Endocrinos/toxicidad , Epigénesis Genética , Femenino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Ratas Wistar , Maduración Sexual
4.
Front Genet ; 12: 613808, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692826

RESUMEN

The SALL2 transcription factor, an evolutionarily conserved gene through vertebrates, is involved in normal development and neuronal differentiation. In disease, SALL2 is associated with eye, kidney, and brain disorders, but mainly is related to cancer. Some studies support a tumor suppressor role and others an oncogenic role for SALL2, which seems to depend on the cancer type. An additional consideration is tissue-dependent expression of different SALL2 isoforms. Human and mouse SALL2 gene loci contain two promoters, each controlling the expression of a different protein isoform (E1 and E1A). Also, several improvements on the human genome assembly and gene annotation through next-generation sequencing technologies reveal correction and annotation of additional isoforms, obscuring dissection of SALL2 isoform-specific transcriptional targets and functions. We here integrated current data of normal/tumor gene expression databases along with ChIP-seq binding profiles to analyze SALL2 isoforms expression distribution and infer isoform-specific SALL2 targets. We found that the canonical SALL2 E1 isoform is one of the lowest expressed, while the E1A isoform is highly predominant across cell types. To dissect SALL2 isoform-specific targets, we analyzed publicly available ChIP-seq data from Glioblastoma tumor-propagating cells and in-house ChIP-seq datasets performed in SALL2 wild-type and E1A isoform knockout HEK293 cells. Another available ChIP-seq data in HEK293 cells (ENCODE Consortium Phase III) overexpressing a non-canonical SALL2 isoform (short_E1A) was also analyzed. Regardless of cell type, our analysis indicates that the SALL2 long E1 and E1A isoforms, but not short_E1A, are mostly contributing to transcriptional control, and reveals a highly conserved network of brain-specific transcription factors (i.e., SALL3, POU3F2, and NPAS3). Our data integration identified a conserved molecular network in which SALL2 regulates genes associated with neural function, cell differentiation, development, and cell adhesion between others. Also, we identified PODXL as a gene that is likely regulated by SALL2 across tissues. Our study encourages the validation of publicly available ChIP-seq datasets to assess a specific gene/isoform's transcriptional targets. The knowledge of SALL2 isoforms expression and function in different tissue contexts is relevant to understanding its role in disease.

5.
Sci Rep ; 11(1): 1996, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479437

RESUMEN

Female puberty is subject to Polycomb Group (PcG)-dependent transcriptional repression. Kiss1, a puberty-activating gene, is a key target of this silencing mechanism. Using a gain-of-function approach and a systems biology strategy we now show that EED, an essential PcG component, acts in the arcuate nucleus of the hypothalamus to alter the functional organization of a gene network involved in the stimulatory control of puberty. A central node of this network is Kdm6b, which encodes an enzyme that erases the PcG-dependent histone modification H3K27me3. Kiss1 is a first neighbor in the network; genes encoding glutamatergic receptors and potassium channels are second neighbors. By repressing Kdm6b expression, EED increases H3K27me3 abundance at these gene promoters, reducing gene expression throughout a gene network controlling puberty activation. These results indicate that Kdm6b repression is a basic mechanism used by PcG to modulate the biological output of puberty-activating gene networks.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/genética , Kisspeptinas/genética , Complejo Represivo Polycomb 2/genética , Pubertad/genética , Animales , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Neuronas/metabolismo , Sistemas Neurosecretores/crecimiento & desarrollo , Sistemas Neurosecretores/metabolismo , Proteínas del Grupo Polycomb/genética , Regiones Promotoras Genéticas/genética , Pubertad/fisiología , Ratas , Biología de Sistemas
6.
Curr Opin Endocr Metab Res ; 14: 65-72, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32905232

RESUMEN

In the arcuate nucleus (ARC) of the hypothalamus reside two neuronal systems in charge of regulating feeding control and reproductive development. The melanocortin system responds to metabolic fluctuations adjusting food intake, whereas kisspeptin neurons are in charge of the excitatory control of Gonadotropin Hormone Releasing Hormone (GnRH) neurons. While it is known that the melanocortin system regulates GnRH neuronal activity, it was recently demonstrated that kisspeptin neurons not only innervate melanocortin neurons, but also play an active role in the control of metabolism. These two neuronal systems are intricately interconnected forming loops of stimulation and inhibition according to metabolic status. Furthermore, intracellular and epigenetic pathways respond to external environmental signals by changing DNA conformation and gene expression. Here we review the role of Silent mating type Information Regulation 2 homologue 1 (Sirt1), a class III NAD+ dependent protein deacetylase, in the ARC control of pubertal development and feeding behavior.

7.
J Clin Invest ; 130(8): 4486-4500, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32407292

RESUMEN

The identification of loss-of-function mutations in MKRN3 in patients with central precocious puberty in association with the decrease in MKRN3 expression in the medial basal hypothalamus of mice before the initiation of reproductive maturation suggests that MKRN3 is acting as a brake on gonadotropin-releasing hormone (GnRH) secretion during childhood. In the current study, we investigated the mechanism by which MKRN3 prevents premature manifestation of the pubertal process. We showed that, as in mice, MKRN3 expression is high in the hypothalamus of rats and nonhuman primates early in life, decreases as puberty approaches, and is independent of sex steroid hormones. We demonstrated that Mkrn3 is expressed in Kiss1 neurons of the mouse hypothalamic arcuate nucleus and that MKRN3 repressed promoter activity of human KISS1 and TAC3, 2 key stimulators of GnRH secretion. We further showed that MKRN3 has ubiquitinase activity, that this activity is reduced by MKRN3 mutations affecting the RING finger domain, and that these mutations compromised the ability of MKRN3 to repress KISS1 and TAC3 promoter activity. These results indicate that MKRN3 acts to prevent puberty initiation, at least in part, by repressing KISS1 and TAC3 transcription and that this action may involve an MKRN3-directed ubiquitination-mediated mechanism.


Asunto(s)
Kisspeptinas/biosíntesis , Neuronas/metabolismo , Pubertad Precoz/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/patología , Femenino , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Células HEK293 , Humanos , Kisspeptinas/genética , Masculino , Ratones , Neuroquinina B/genética , Neuroquinina B/metabolismo , Neuronas/patología , Regiones Promotoras Genéticas , Pubertad Precoz/genética , Pubertad Precoz/patología , Ratas Sprague-Dawley , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética
8.
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.

9.
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
10.
J Res Adolesc ; 29(1): 54-79, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30869843

RESUMEN

The adolescent transition begins with the onset of puberty which, upstream in the brain, is initiated by the gonadotropin-releasing hormone (GnRH) pulse generator that activates the release of peripheral sex hormones. Substantial research in human and animal models has revealed a myriad of cellular networks and heritable genes that control the GnRH pulse generator allowing the individual to begin the process of reproductive competence and sexual maturation. Here, we review the latest knowledge in neuroendocrine pubertal research with emphasis on genetic and epigenetic mechanisms underlying the pubertal transition.


Asunto(s)
Salud del Adolescente , Epigénesis Genética , Hormonas Esteroides Gonadales/metabolismo , Sistemas Neurosecretores/fisiología , Regiones Promotoras Genéticas/fisiología , Pubertad/genética , Maduración Sexual/genética , Adolescente , Animales , Femenino , Humanos , Kisspeptinas , Hormona Luteinizante , Masculino
11.
Neuroendocrinology ; 109(3): 208-217, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30731454

RESUMEN

To attain sexual competence, all mammalian species go through puberty, a maturational period during which body growth and development of secondary sexual characteristics occur. Puberty begins when the diurnal pulsatile gonadotropin-releasing hormone (GnRH) release from the hypothalamus increases for a prolonged period of time, driving the adenohypophysis to increase the pulsatile release of luteinizing hormone with diurnal periodicity. Increased pubertal GnRH secretion does not appear to be driven by inherent changes in GnRH neuronal activity; rather, it is induced by changes in transsynaptic and glial inputs to GnRH neurons. We now know that these changes involve a reduction in inhibitory transsynaptic inputs combined with increased transsynaptic and glial excitatory inputs to the GnRH neuronal network. Although the pubertal process is known to have a strong genetic component, during the last several years, epigenetics has been implicated as a significant regulatory mechanism through which GnRH release is first repressed before puberty and is involved later on during the increase in GnRH secretion that brings about the pubertal process. According to this concept, a central target of epigenetic regulation is the transcriptional machinery of neurons implicated in stimulating GnRH release. Here, we will briefly review the hormonal changes associated with the advent of female puberty and the role that excitatory transsynaptic inputs have in this process. In addition, we will examine the 3 major groups of epigenetic modifying enzymes expressed in the neuroendocrine hypothalamus, which was recently shown to be involved in pubertal development and progression.


Asunto(s)
Cromatina/metabolismo , Pubertad/metabolismo , Maduración Sexual/fisiología , Animales , Epigénesis Genética , Femenino , Humanos
12.
Clin Epigenetics ; 10(1): 146, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30466473

RESUMEN

BACKGROUND: Recent studies demonstrated that changes in DNA methylation (DNAm) and inactivation of two imprinted genes (MKRN3 and DLK1) alter the onset of female puberty. We aimed to investigate the association of DNAm profiling with the timing of human puberty analyzing the genome-wide DNAm patterns of peripheral blood leukocytes from ten female patients with central precocious puberty (CPP) and 33 healthy girls (15 pre- and 18 post-pubertal). For this purpose, we performed comparisons between the groups: pre- versus post-pubertal, CPP versus pre-pubertal, and CPP versus post-pubertal. RESULTS: Analyzing the methylome changes associated with normal puberty, we identified 120 differentially methylated regions (DMRs) when comparing pre- and post-pubertal healthy girls. Most of these DMRs were hypermethylated in the pubertal group (99%) and located on the X chromosome (74%). Only one genomic region, containing the promoter of ZFP57, was hypomethylated in the pubertal group. ZFP57 is a transcriptional repressor required for both methylation and imprinting of multiple genomic loci. ZFP57 expression in the hypothalamus of female rhesus monkeys increased during peripubertal development, suggesting enhanced repression of downstream ZFP57 target genes. Fourteen other zinc finger (ZNF) genes were related to the hypermethylated DMRs at normal puberty. Analyzing the methylome changes associated with CPP, we demonstrated that the patients with CPP exhibited more hypermethylated CpG sites compared to both pre-pubertal (81%) and pubertal (89%) controls. Forty-eight ZNF genes were identified as having hypermethylated CpG sites in CPP. CONCLUSION: Methylome profiling of girls at normal and precocious puberty revealed a widespread pattern of DNA hypermethylation, indicating that the pubertal process in humans is associated with specific changes in epigenetically driven regulatory control. Moreover, changes in methylation of several ZNF genes appear to be a distinct epigenetic modification underlying the initiation of human puberty.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/genética , Estudio de Asociación del Genoma Completo/métodos , Pubertad Precoz/genética , Factores de Transcripción/genética , Animales , Estudios de Casos y Controles , Niño , Epigénesis Genética , Femenino , Impresión Genómica , Humanos , Macaca mulatta , Linaje , Regiones Promotoras Genéticas , Proteínas Represoras , Dedos de Zinc
13.
Nat Commun ; 9(1): 57, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302059

RESUMEN

Polycomb group (PcG) proteins control the timing of puberty by repressing the Kiss1 gene in hypothalamic arcuate nucleus (ARC) neurons. Here we identify two members of the Trithorax group (TrxG) of modifiers, mixed-lineage leukemia 1 (MLL1), and 3 (MLL3), as central components of an activating epigenetic machinery that dynamically counteracts PcG repression. Preceding puberty, MLL1 changes the chromatin configuration at the promoters of Kiss1 and Tac3, two genes required for puberty to occur, from repressive to permissive. Concomitantly, MLL3 institutes a chromatin structure that changes the functional status of a Kiss1 enhancer from poised to active. RNAi-mediated, ARC-specific Mll1 knockdown reduced Kiss1 and Tac3 expression, whereas CRISPR-Cas9-directed epigenome silencing of the Kiss1 enhancer selectively reduced Kiss1 activity. Both interventions delay puberty and disrupt reproductive cyclicity. Our results demonstrate that an epigenetic switch from transcriptional repression to activation is crucial to the regulatory mechanism controlling the timing of mammalian puberty.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Hipotálamo/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Pubertad/genética , Animales , Sistemas CRISPR-Cas , Cromatina , Epigénesis Genética , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Kisspeptinas/genética , Macaca mulatta , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Taquicininas/genética
14.
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
15.
J Biol Chem ; 285(47): 36471-85, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20843809

RESUMEN

Na(+)-coupled ascorbic acid transporter-2 (SVCT2) activity is impaired at acid pH, but little is known about the molecular determinants that define the transporter pH sensitivity. SVCT2 contains six histidine residues in its primary sequence, three of which are exofacial in the transporter secondary structure model. We used site-directed mutagenesis and treatment with diethylpyrocarbonate to identify histidine residues responsible for SVCT2 pH sensitivity. We conclude that five histidine residues, His(109), His(203), His(206), His(269), and His(413), are central regulators of SVCT2 function, participating to different degrees in modulating pH sensitivity, transporter kinetics, Na(+) cooperativity, conformational stability, and subcellular localization. Our results are compatible with a model in which (i) a single exofacial histidine residue, His(413), localized in the exofacial loop IV that connects transmembrane helices VII-VIII defines the pH sensitivity of SVCT2 through a mechanism involving a marked attenuation of the activation by Na(+) and loss of Na(+) cooperativity, which leads to a decreased V(max) without altering the transport K(m); (ii) exofacial histidine residues His(203), His(206), and His(413) may be involved in maintaining a functional interaction between exofacial loops II and IV and influence the general folding of the transporter; (iii) histidines 203, 206, 269, and 413 affect the transporter kinetics by modulating the apparent transport K(m); and (iv) histidine 109, localized at the center of transmembrane helix I, might be fundamental for the interaction of SVCT2 with the transported substrate ascorbic acid. Thus, histidine residues are central regulators of SVCT2 function.


Asunto(s)
Histidina/metabolismo , Riñón/metabolismo , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Sodio/metabolismo , Simportadores/metabolismo , Ácido Ascórbico/metabolismo , Transporte Biológico , Biotinilación , Membrana Celular/metabolismo , Histidina/química , Histidina/genética , Humanos , Concentración de Iones de Hidrógeno , Riñón/citología , Cinética , Mutagénesis Sitio-Dirigida , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Conformación Proteica , Transportadores de Sodio Acoplados a la Vitamina C , Fracciones Subcelulares , Simportadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...