Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 407
Filtrar
1.
Biomolecules ; 10(5)2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32365821

RESUMEN

Cystathionine-ß-synthase (CBS), the first (and rate-limiting) enzyme in the transsulfuration pathway, is an important mammalian enzyme in health and disease. Its biochemical functions under physiological conditions include the metabolism of homocysteine (a cytotoxic molecule and cardiovascular risk factor) and the generation of hydrogen sulfide (H2S), a gaseous biological mediator with multiple regulatory roles in the vascular, nervous, and immune system. CBS is up-regulated in several diseases, including Down syndrome and many forms of cancer; in these conditions, the preclinical data indicate that inhibition or inactivation of CBS exerts beneficial effects. This article overviews the current information on the expression, tissue distribution, physiological roles, and biochemistry of CBS, followed by a comprehensive overview of direct and indirect approaches to inhibit the enzyme. Among the small-molecule CBS inhibitors, the review highlights the specificity and selectivity problems related to many of the commonly used "CBS inhibitors" (e.g., aminooxyacetic acid) and provides a comprehensive review of their pharmacological actions under physiological conditions and in various disease models.


Asunto(s)
Cistationina betasintasa/metabolismo , Síndrome de Down/metabolismo , Neoplasias/metabolismo , Animales , Cistationina betasintasa/antagonistas & inhibidores , Cistationina betasintasa/genética , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/enzimología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Sulfuro de Hidrógeno/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Tacrolimus/uso terapéutico
2.
Sci Rep ; 10(1): 3181, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32081899

RESUMEN

Phosphorylation of Munc18-1 (Stxbp1), a presynaptic organizer of synaptic vesicle fusion, is a powerful mechanism to regulate synaptic strength. Munc18-1 is a proposed substrate for the Down Syndrome-related kinase dual-specificity tyrosine phosphorylation-regulate kinase 1a (Dyrk1a) and mutations in both genes cause intellectual disability. However, the functional consequences of Dyrk1a-dependent phosphorylation of Munc18-1 for synapse function are unknown. Here, we show that the proposed Munc18-1 phosphorylation site, T479, is among the highly constrained phosphorylation sites in the coding regions of the gene and is also located within a larger constrained coding region. We confirm that Dyrk1a phosphorylates Munc18-1 at T479. Patch-clamp physiology in conditional null mutant hippocampal neurons expressing Cre and either wildtype, or mutants mimicking or preventing phosphorylation, revealed that synaptic transmission is similar among the three groups: frequency/amplitude of mEPSCs, evoked EPSCs, paired pulse plasticity, rundown kinetics upon intense activity and the readily releasable pool. However, synapses expressing the phosphomimic mutant responded to intense activity with more pronounced facilitation. These data indicate that Dyrk1a-dependent Munc18-1 phosphorylation has a minor impact on synaptic transmission, only after intense activity, and that the role of genetic variation in both genes in intellectual disability may be through different mechanisms.


Asunto(s)
Síndrome de Down/enzimología , Proteínas Munc18/genética , Mutación/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transmisión Sináptica , Animales , Supervivencia Celular , Células HEK293 , Humanos , Ratones , Proteínas Munc18/deficiencia , Proteínas Munc18/metabolismo , Neuronas/metabolismo , Neuronas/patología , Sistemas de Lectura Abierta/genética , Fosforilación , Treonina/metabolismo
3.
Int J Mol Sci ; 21(4)2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32092951

RESUMEN

Epigallocatechin gallate (EGCG) is an inhibitor of DYRK1A, a serine/threonine kinase considered to be a major contributor of cognitive dysfunctions in Down syndrome (DS). Two clinical trials in adult patients with DS have shown the safety and efficacy to improve cognitive phenotypes using commercial green tea extract containing EGCG (45% content). In the present study, we performed a preclinical study using FontUp®, a new nutritional supplement with a chocolate taste specifically formulated for the nutritional needs of patients with DS and enriched with a standardized amount of EGCG in young mice overexpressing Dyrk1A (TgBACDyrk1A). This preparation is differential with previous one used, because its green tea extract has been purified to up 94% EGCG of total catechins. We analyzed the in vitro effect of green tea catechins not only for EGCG, but for others residually contained in FontUp®, on DYRK1A kinase activity. Like EGCG, epicatechin gallate was a noncompetitive inhibitor against ATP, molecular docking computations confirming these results. Oral FontUp® normalized brain and plasma biomarkers deregulated in TgBACDyrk1A, without negative effect on liver and cardiac functions. We compared the bioavailability of EGCG in plasma and brain of mice and have demonstrated that EGCG had well crossed the blood-brain barrier.


Asunto(s)
Encéfalo/efectos de los fármacos , Catequina/análogos & derivados , Síndrome de Down/dietoterapia , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Té/química , Animales , Antioxidantes/administración & dosificación , Antioxidantes/uso terapéutico , Disponibilidad Biológica , Biomarcadores/sangre , Biomarcadores/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/ultraestructura , Catequina/administración & dosificación , Catequina/efectos adversos , Catequina/química , Catequina/uso terapéutico , Suplementos Dietéticos , Síndrome de Down/sangre , Síndrome de Down/enzimología , Síndrome de Down/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Polifenoles/análisis , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Regulación hacia Arriba
4.
Neurotox Res ; 37(3): 714-723, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31802378

RESUMEN

Down syndrome (DS) is a developmental disorder that results from the trisomy of chromosome 21. DS patients show several abnormalities including cognitive deficits. Here, we show enhanced activation of the extracellular signal-regulated kinase (ERK), a kinase that critically regulates synaptic plasticity and memory, in a hippocampal cell line derived from trisomy 16 mouse foetus. In addition, these cells show enhanced activation of p38 mitogen-activated protein kinase (p38 MAPK). The hyper-activation of ERK and p38 MAPK is significantly reduced by a small peptide, Gly-Pro-Glu (GPE), derived from insulin-like growth factor-1. In addition, the trisomic cells show reduced level of inhibitory phosphorylation of glycogen synthase kinase-3ß (GSK-3ß), which is enhanced by GPE. Furthermore, the trisomic cells do not show ERK activation in response to KCl depolarization or forskolin treatment. Importantly, ERK activation by these stimuli is observed after GPE treatment of the cells. These results suggest that GPE may help reduce aberrant signalling in the trisomic neurons by affecting MAPK and GSK-3ß activation.


Asunto(s)
Síndrome de Down/enzimología , Síndrome de Down/prevención & control , Glucógeno Sintasa Quinasas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oligopéptidos/administración & dosificación , Animales , Línea Celular , Modelos Animales de Enfermedad , Feto/citología , Humanos , Ratones , Transducción de Señal/efectos de los fármacos
5.
Prenat Diagn ; 39(1): 3-9, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30474229

RESUMEN

Published studies indicate the MTHFR C677T and A1298C polymorphisms are associated with abnormal homocysteine levels, which may cause various pregnancy complications and birth defects. However, the results obtained from different studies have been inconsistent. Therefore, this meta-analysis explores the association between MTHFR polymorphisms and birth defects and adverse pregnancy outcomes. The PubMed, ScienceDirect, Embase, and China Biology Medicine literature databases and ClinicalTrials were searched. Analyses of public bias, meta-regression, subgroups, and sensitivity were used to ensure the robustness of our results. MTHFR C677T was significantly associated with recurrent pregnancy loss in developing countries (odds ratio [OR], 1.34; 95% confidence interval [CI], 1.20-1.50) but not in developed countries (OR, 0.87; 95% CI, 0.68-1.11). No significant relationship was found between MTHFR A1298C and recurrent pregnancy loss (OR, 1.04; 95% CI, 0.93-1.18). MTHFR C677T and A1298C were not associated with preeclampsia (OR, 1.06; 95% CI, 0.97-1.16 and OR, 1.16; 95% CI, 0.97-1.39, respectively), and C677T was not associated with placental abruption (OR, 1.03; 95% CI, 0.87-1.21), intrauterine growth retardation (OR, 1.02; 95% CI, 0.90-1.15), or congenital heart disease (OR, 1.05; 95% CI, 0.89-1.25). MTHFR C677T, but not A1298C, was associated with neural tube defects (OR, 1.24; 95% CI, 1.08-1.42) and Down syndrome (OR, 1.65; 95% CI, 1.39-1.95). CONCLUSION: Although MTHFR C677T and A1298C are significantly associated with some types of congenital defects and adverse pregnancy outcomes, the impact of these polymorphisms is moderate.


Asunto(s)
Anomalías Congénitas/enzimología , Anomalías Congénitas/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo Genético , Resultado del Embarazo/genética , Aborto Habitual/enzimología , Aborto Habitual/genética , Ensayos Clínicos como Asunto , Países en Desarrollo , Síndrome de Down/enzimología , Síndrome de Down/genética , Femenino , Retardo del Crecimiento Fetal/enzimología , Retardo del Crecimiento Fetal/genética , Humanos , Defectos del Tubo Neural/enzimología , Defectos del Tubo Neural/genética , Oportunidad Relativa , Embarazo
6.
Sci Rep ; 8(1): 2859, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29434250

RESUMEN

Inhibition of DYRK1A kinase, produced by chromosome 21 and consequently overproduced in trisomy 21 subjects, has been suggested as a therapeutic approach to treating the cognitive deficiencies observed in Down syndrome (DS). We now report the synthesis and potent DYRK1A inhibitory activities of fluoro derivatives of 3,5-di(polyhydroxyaryl)-7-azaindoles (F-DANDYs). One of these compounds (3-(4-fluorophenyl)-5-(3,4-dihydroxyphenyl)-1H-pyrrolo[2,3-b]pyridine, 5a) was selected for in vivo studies of cognitive rescuing effects in a standard mouse model of DS (Ts65Dn line). Using the Morris water maze task, Ts65Dn mice treated i.p. with 20 mg/kg of 5a performed significantly better than Ts65Dn mice treated with placebo, confirming the promnesiant effect of 5a in the trisomic mice. Overall, these results demonstrate for the first time that selective and competitive inhibition of DYRK1A kinase by the F-DANDY derivative 5a may provide a viable treatment strategy for combating the memory and learning deficiencies encountered in DS.


Asunto(s)
Síndrome de Down/psicología , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Piridinas/administración & dosificación , Animales , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Síndrome de Down/enzimología , Humanos , Inyecciones Intraperitoneales , Discapacidades para el Aprendizaje/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/química , Piridinas/farmacología
7.
Physiol Behav ; 177: 230-241, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28478033

RESUMEN

Down syndrome (DS) is caused by three copies of human chromosome 21 (Hsa21) and results in phenotypes including intellectual disability and skeletal deficits. Ts65Dn mice have three copies of ~50% of the genes homologous to Hsa21 and display phenotypes associated with DS, including cognitive deficits and skeletal abnormalities. DYRK1A is found in three copies in humans with Trisomy 21 and in Ts65Dn mice, and is involved in a number of critical pathways including neurological development and osteoclastogenesis. Epigallocatechin-3-gallate (EGCG), the main polyphenol in green tea, inhibits Dyrk1a activity. We have previously shown that EGCG treatment (~10mg/kg/day) improves skeletal abnormalities in Ts65Dn mice, yet the same dose, as well as ~20mg/kg/day did not rescue deficits in the Morris water maze spatial learning task (MWM), novel object recognition (NOR) or balance beam task (BB). In contrast, a recent study reported that an EGCG-containing supplement with a dose of 2-3mg per day (~40-60mg/kg/day) improved hippocampal-dependent task deficits in Ts65Dn mice. The current study investigated if an EGCG dosage similar to that study would yield similar improvements in either cognitive or skeletal deficits. Ts65Dn mice and euploid littermates were given EGCG [0.4mg/mL] or a water control, with treatments yielding average daily intakes of ~50mg/kg/day EGCG, and tested on the multivariate concentric square field (MCSF)-which assesses activity, exploratory behavior, risk assessment, risk taking, and shelter seeking-and NOR, BB, and MWM. EGCG treatment failed to improve cognitive deficits; EGCG also produced several detrimental effects on skeleton in both genotypes. In a refined HPLC-based assay, its first application in Ts65Dn mice, EGCG treatment significantly reduced kinase activity in femora but not in the cerebral cortex, cerebellum, or hippocampus. Counter to expectation, 9-week-old Ts65Dn mice exhibited a decrease in Dyrk1a protein levels in Western blot analysis in the cerebellum. The lack of beneficial therapeutic behavioral effects and potentially detrimental skeletal effects of EGCG found in Ts65Dn mice emphasize the importance of identifying dosages of EGCG that reliably improve DS phenotypes and linking those effects to actions of EGCG (or EGCG-containing supplements) in specific targets in brain and bone.


Asunto(s)
Catequina/análogos & derivados , Cognición/efectos de los fármacos , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/patología , Fémur/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Administración Oral , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Catequina/farmacología , Cognición/fisiología , Modelos Animales de Enfermedad , Síndrome de Down/enzimología , Síndrome de Down/psicología , Fémur/diagnóstico por imagen , Fémur/enzimología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C3H , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Fenotipo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Distribución Aleatoria , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Insuficiencia del Tratamiento
8.
Neurobiol Dis ; 103: 11-23, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28359846

RESUMEN

Neurogenesis impairment is considered a major determinant of the intellectual disability that characterizes Down syndrome (DS), a genetic condition caused by triplication of chromosome 21. Previous evidence obtained in the Ts65Dn mouse model of DS showed that the triplicated gene APP (amyloid precursor protein) is critically involved in neurogenesis alterations. In particular, excessive levels of AICD (amyloid precursor protein intracellular domain) resulting from APP cleavage by gamma-secretase increase the transcription of Ptch1, a Sonic Hedgehog (Shh) receptor that keeps the mitogenic Shh pathway repressed. Previous evidence showed that neonatal treatment with ELND006, an inhibitor of gamma-secretase, reinstates the Shh pathway and fully restores neurogenesis in Ts65Dn pups. In the framework of potential therapies for DS, it is extremely important to establish whether the positive effects of early intervention are retained after treatment cessation. Therefore, the goal of the current study was to establish whether early treatment with ELND006 leaves an enduring trace in the brain of Ts65Dn mice. Ts65Dn and euploid pups were treated with ELND006 in the postnatal period P3-P15 and the outcome of treatment was examined at ~one month after treatment cessation. We found that in treated Ts65Dn mice the pool of proliferating cells in the hippocampal dentate gyrus (DG) and total number of granule neurons were still restored as was the number of pre- and postsynaptic terminals in the stratum lucidum of CA3, the site of termination of the mossy fibers from the DG. Accordingly, patch-clamp recording from field CA3 showed functional normalization of the input to CA3. Unlike in field CA3, the number of pre- and postsynaptic terminals in the DG of treated Ts65Dn mice was no longer fully restored. The finding that many of the positive effects of neonatal treatment were retained after treatment cessation provides proof of principle demonstration of the efficacy of early inhibition of gamma-secretase for the improvement of brain development in DS.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Modelos Animales de Enfermedad , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/enzimología , Hipocampo/enzimología , Pirazoles/uso terapéutico , Quinolinas/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Animales Recién Nacidos , Síndrome de Down/patología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirazoles/farmacología , Quinolinas/farmacología , Factores de Tiempo , Resultado del Tratamiento
9.
Biol Pharm Bull ; 40(3): 327-333, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28250274

RESUMEN

Amyloid-ß peptide (Aß) accumulation is a triggering event leading to the Alzheimer's disease (AD) pathological cascade. Almost all familial AD-linked gene mutations increase Aß production and accelerate the onset of AD. The Swedish mutation of amyloid precursor protein (APP) affects ß-secretase activity and increases Aß production up to ca. 6-fold in cultured cells; the onset age is around 50. Down syndrome (DS) patients with chromosome 21 trisomy present AD-like pathologies at earlier ages (40s) compared with sporadic AD patients, because APP gene expression is 1.5-fold higher than that in healthy people, thus causing a 1.5-fold increase in Aß production. However, when comparing the causal relationship of Aß accumulation with the onset age between the above two populations, early DS pathogenesis does not appear to be accounted for by the increased Aß production alone. In this study, we found that neprilysin, a major Aß-degrading enzyme, was downregulated in DS patient-derived fibroblasts, compared with healthy people-derived fibroblasts. Treatment with harmine, an inhibitor of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), which is located in the DS critical region of chromosome 21, and gene knockdown of DYRK1A, upregulated neprilysin in fibroblasts. These results suggest that a decrease in the Aß catabolic rate may be, at least in part, one of the causes for accelerated AD-like pathogenesis in DS patients if a similar event occurs in the brains, and that neprilysin activity may be regulated directly or indirectly by DYRK1A-mediated phosphorylation. DYRK1A inhibition may be a promising disease-modifying therapy for AD via neprilysin upregulation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Síndrome de Down/metabolismo , Fibroblastos/metabolismo , Neprilisina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Enfermedad de Alzheimer/enzimología , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/patología , Línea Celular , Cromosomas Humanos Par 21 , Síndrome de Down/enzimología , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Fibroblastos/enzimología , Harmina/farmacología , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Tirosina/metabolismo
10.
Dis Model Mech ; 9(8): 839-48, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27483355

RESUMEN

DYRK1A is important in neuronal development and function, and its excessive activity is considered a significant pathogenic factor in Down syndrome and Alzheimer's disease. Thus, inhibition of DYRK1A has been suggested to be a new strategy to modify the disease. Very few compounds, however, have been reported to act as inhibitors, and their potential clinical uses require further evaluation. Here, we newly identify CX-4945, the safety of which has been already proven in the clinical setting, as a potent inhibitor of DYRK1A that acts in an ATP-competitive manner. The inhibitory potency of CX-4945 on DYRK1A (IC50=6.8 nM) in vitro was higher than that of harmine, INDY or proINDY, which are well-known potent inhibitors of DYRK1A. CX-4945 effectively reverses the aberrant phosphorylation of Tau, amyloid precursor protein (APP) and presenilin 1 (PS1) in mammalian cells. To our surprise, feeding with CX-4945 significantly restored the neurological and phenotypic defects induced by the overexpression of minibrain, an ortholog of human DYRK1A, in the Drosophila model. Moreover, oral administration of CX-4945 acutely suppressed Tau hyperphosphorylation in the hippocampus of DYRK1A-overexpressing mice. Our research results demonstrate that CX-4945 is a potent DYRK1A inhibitor and also suggest that it has therapeutic potential for DYRK1A-associated diseases.


Asunto(s)
Síndrome de Down/tratamiento farmacológico , Síndrome de Down/enzimología , Naftiridinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/farmacología , Administración Oral , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Calcineurina/metabolismo , Modelos Animales de Enfermedad , Síndrome de Down/patología , Drosophila melanogaster/metabolismo , Células HEK293 , Hipocampo/patología , Humanos , Ratones Endogámicos C57BL , Modelos Moleculares , Factores de Transcripción NFATC/metabolismo , Naftiridinas/administración & dosificación , Naftiridinas/química , Naftiridinas/farmacología , Neuronas/efectos de los fármacos , Neuronas/patología , Fenazinas , Fenotipo , Fosforilación/efectos de los fármacos , Presenilina-1 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas tau/metabolismo
11.
Mutat Res ; 787: 1-6, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26926955

RESUMEN

Despite that advanced maternal age at conception represents the major risk factor for the birth of a child with Down syndrome (DS), most of DS babies are born from women aging less than 35 years. Studies performed in peripheral lymphocytes of those women revealed several markers of global genome instability, including an increased frequency of micronuclei, shorter telomeres and impaired global DNA methylation. Furthermore, young mothers of DS individuals (MDS) are at increased risk to develop dementia later in life, suggesting that they might be "biologically older" than mothers of euploid babies of similar age. Mutations in folate pathway genes, and particularly in the methylenetetrahydrofolate reductase (MTHFR) one, have been often associated with maternal risk for a DS birth as well as with risk of dementia in the elderly. Recent studies pointed out that also changes in MTHFR methylation levels can contribute to human disease, but nothing is known about MTHFR methylation in MDS tissues. We investigated MTHFR promoter methylation in DNA extracted from perypheral lymphocytes of 40 MDS and 44 matched control women that coinceived their children before 35 years of age, observing a significantly increased MTHFR promoter methylation in the first group (33.3 ± 8.1% vs. 28.3 ± 5.8%; p=0.001). In addition, the frequency of micronucleated lymphocytes was available from the women included in the study, was higher in MDS than control mothers (16.1 ± 8.6‰ vs. 10.5 ± 4.3‰; p=0.0004), and correlated with MTHFR promoter methylation levels (r=0.33; p=0.006). Present data suggest that MTHFR epimutations are likely to contribute to the increased genomic instability observed in cells from MDS, and could play a role in the risk of birth of a child with DS as well as in the onset of age related diseases in those women.


Asunto(s)
Metilación de ADN , Síndrome de Down/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Adulto , Estudios de Casos y Controles , Síndrome de Down/enzimología , Femenino , Regulación Enzimológica de la Expresión Génica , Estudios de Asociación Genética , Humanos , Persona de Mediana Edad , Factores de Riesgo , Análisis de Secuencia de ADN
12.
Hum Mol Genet ; 25(22): 4856-4869, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28172997

RESUMEN

Trisomy 21 (Ts21) affects craniofacial precursors in individuals with Down syndrome (DS). The resultant craniofacial features in all individuals with Ts21 may significantly affect breathing, eating and speaking. Using mouse models of DS, we have traced the origin of DS-associated craniofacial abnormalities to deficiencies in neural crest cell (NCC) craniofacial precursors early in development. Hypothetically, three copies of Dyrk1a (dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A), a trisomic gene found in most humans with DS and mouse models of DS, may significantly affect craniofacial structure. We hypothesized that we could improve DS-related craniofacial abnormalities in mouse models using a Dyrk1a inhibitor or by normalizing Dyrk1a gene dosage. In vitro and in vivo treatment with Epigallocatechin-3-gallate (EGCG), a Dyrk1a inhibitor, modulated trisomic NCC deficiencies at embryonic time points. Furthermore, prenatal EGCG treatment normalized some craniofacial phenotypes, including cranial vault in adult Ts65Dn mice. Normalization of Dyrk1a copy number in an otherwise trisomic Ts65Dn mice normalized many dimensions of the cranial vault, but did not correct all craniofacial anatomy. These data underscore the complexity of the gene­phenotype relationship in trisomy and suggest that changes in Dyrk1a expression play an important role in morphogenesis and growth of the cranial vault. These results suggest that a temporally specific prenatal therapy may be an effective way to ameliorate some craniofacial anatomical changes associated with DS.


Asunto(s)
Catequina/análogos & derivados , Síndrome de Down/terapia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Animales , Catequina/farmacología , Anomalías Craneofaciales/enzimología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/terapia , Modelos Animales de Enfermedad , Síndrome de Down/enzimología , Síndrome de Down/genética , Femenino , Dosificación de Gen , Ratones , Fenotipo , Fosforilación , Embarazo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo
13.
Clin Biochem ; 49(1-2): 61-5, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26340803

RESUMEN

OBJECTIVES: Oxidative stress (OS) may play a critical role in cell aging and neurologic disorders that are often seen in Down syndrome (DS) patients. The aim of this study was to determine the antioxidant enzyme level and lipoperoxidation status in blood from DS children. DESIGN AND METHODS: In a cross-sectional study, we recruited a total of 36 DS children and 40 healthy controls (HCs). All subjects were free of infection according to the C reactive protein (CRP) value and routine peripheral blood profile. The activities of total superoxide dismutases (SODs), extracellular glutathione peroxidase (GPx3),malondialdehyde (MDA) and nitric oxide synthase (NOS) concentrations in peripheral blood were measured by spectrophotometric methods. The relationship of SOD and GPx3 was analyzed in the two groups. RESULTS: The two groups were similar with respect to age, gender and peripheral blood profiles. The total SOD activity was significantly increased, while the GPx3 activity was significantly reduced in the DS group compared to the HCs (p=0.000, p=0.033 respectively). The MDA level was higher in DS children (p=0.013). There was no significant difference in NOS between DS and HCs (p=0.708). A significant negative correlation between GPx3 and SOD activity was identified in DS (r=-0.14, p=0.018) but not in the HC group. CONCLUSIONS: Abnormal redox metabolism takes place in DS individuals. Reducing GPx3 may be a compensatory mechanism of protection against intracellular OS. Moreover, monitoring of decreases in GPx3 activity may be a useful biomarker for evaluating OS in DS patients.


Asunto(s)
Catalasa/metabolismo , Síndrome de Down/sangre , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido , Malondialdehído/metabolismo , Superóxido Dismutasa/metabolismo , Estudios de Casos y Controles , Niño , Síndrome de Down/enzimología , Humanos
14.
Cardiovasc Toxicol ; 16(2): 182-92, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25962911

RESUMEN

The intracardiac synthesis of anthracycline alcohol metabolites by aldo-keto reductases (AKRs) contributes to the pathogenesis of anthracycline-related cardiotoxicity. AKR7A2 is the most abundant anthracycline reductase in hearts from donors with and without Down syndrome (DS), and its expression varies between individuals (≈tenfold). We investigated whether DNA methylation impacts AKR7A2 expression in hearts from donors with (n = 11) and without DS (n = 30). Linear models were used to test for associations between methylation status and cardiac AKR7A2 expression. In hearts from donors without DS, DNA methylation status at CpG site -865 correlated with AKR7A2 mRNA (Pearson's regression coefficient, r = -0.4051, P = 0.0264) and AKR7A2 protein expression (r = -0.5818, P = 0.0071). In heart tissue from donors with DS, DNA methylation status at CpG site -232 correlated with AKR7A2 protein expression (r = 0.8659, P = 0.0025). Multiple linear regression modeling revealed that methylation at several CpG sites is associated with the synthesis of cardiotoxic daunorubicinol. AKR7A2 methylation status in lymphoblastoid cell lines from donors with and without DS was examined to explore potential parallelisms between cardiac tissue and lymphoid cells. These results suggest that DNA methylation impacts AKR7A2 expression and the synthesis of cardiotoxic daunorubicinol.


Asunto(s)
Aldehído Reductasa/metabolismo , Antraciclinas/metabolismo , Metilación de ADN/fisiología , Síndrome de Down/enzimología , Miocardio/enzimología , Anciano , Síndrome de Down/diagnóstico , Femenino , Corazón/fisiología , Humanos , Masculino , Persona de Mediana Edad
15.
J Clin Lab Anal ; 30(3): 196-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25546171

RESUMEN

Cryptorchidism represents a risk factor for infertility and germ cell testicular neoplasia. An increased rate of cryptorchidism has been reported in subjects with Down's syndrome. Cyclic nucleotide phosphodiesterases (PDEs) are important messengers that regulate and mediate a number of cellular responses to extracellular signals, such as neurotransmitters and hormones. PDE4B, cAMP-specific (PDE4B) gene which maps to chromosome 1p31.3 appears to be involved in schizophrenia, chronic psychiatric illness, learning, memory, and mood disturbances. Expression of PDE4 enzymes have been studied in testes of cryptorchid rats. Expression of PDE4B protein examination showed marked degenerative changes in the epithelial lining of the seminiferous tubules. These findings led us to evaluate PDE4 mRNA expression in leukocytes of peripheral blood of five men with DS and cryptorchidism and eleven subjects with DS without cryptorchidism compared with healthy men (controls) by quantitative Real Time PCR (qRT-PCR). This study showed that the PDE4B gene was downexpressed in men with DS and cryptorchidism compared to normal controls and DS without cryptorchidism. A lower expression of the PDE4B gene may be involved in the neurological abnormalities in subjects with Down's syndrome. Moreover, PDE4B gene may be involved in the testicular abnormalities of men with DS and cryptorchidism.


Asunto(s)
Criptorquidismo/complicaciones , Criptorquidismo/enzimología , Síndrome de Down/complicaciones , Síndrome de Down/enzimología , Adulto , Estudios de Casos y Controles , Criptorquidismo/genética , AMP Cíclico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Síndrome de Down/genética , Humanos , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo
16.
J Neonatal Perinatal Med ; 8(2): 77-84, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26410429

RESUMEN

UNLABELLED: Down syndrome is the most frequent aneuploidy in live births, with an overall frequency of 1/600-700 births. The overexpression of cystathionine ß-synthase is thought to participate in the presentation of some phenotypes observed in Down syndrome. OBJECTIVE: The aim of this study was to compare the expression levels of cystathionine ß-synthase and histopathological observations from placentas of infants with Down syndrome and healthy newborns. MATERIALS AND METHODS: Six placentas of fetuses/infants with Down syndrome and sixteen placentas of healthy fetuses were studied. Cystathionine ß-synthase mRNA and protein expression were performed by real-time PCR and immunohistochemistry, respectively. RESULTS: We observed an increase in cystathionine ß-synthase mRNA expression (p = 0.0465) and protein levels (p = 0.009) in placentas of fetus/infants with Down syndrome compared with controls. Significantly more circinate edges (p = 0.0007) and trophoblast inclusions (p = 0.0037) were observed in the group with Down syndrome compared with control group. CONCLUSION: The results demonstrate overexpression of cystathionine ß-synthase mRNA and protein in placentas of fetuses/infants with trisomy 21. Further histological abnormalities were found in placentas of patients with Down syndrome, suggesting an alteration in the development of placenta.


Asunto(s)
Cistationina betasintasa/metabolismo , Síndrome de Down/enzimología , MicroARNs/metabolismo , Placenta/enzimología , Colombia/epidemiología , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Recién Nacido , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Neurosci Lett ; 604: 91-6, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26240993

RESUMEN

In addition to nervous system, cardiovascular and respiratory systems are primarily affected in Down syndrome (DS). The Ts65Dn mouse model is widely used to recapitulate cognitive dysfunction in DS. While these mice consistently show failure in learning and memory along with functional and structural abnormalities in the hippocampal region, the underlying mechanisms behind cognitive dysfunction remain to be fully elucidated. Convergent evidence implicates chronic episodes of hypoxemia in cognitive dysfunction in people with DS. Using an infra-red detection system to assess oxygen saturation in free-moving mice, we assessed arterial blood oxygenation in both adolescent and adult Ts65Dn mice and found a significant increase in the incidence of hypoxemia in both groups. Notably, the severity of hypoxemia increased during the dark cycle, suggesting a link between hypoxemia and increased motor activity. Postmortem analysis showed significant increase in the expression of mitochondrial Cox4i2, the terminal enzyme of the mitochondrial respiratory chain and oxygen response element. Altogether these data suggest early and chronic occurrence of hypoxemia in the Ts65Dn mouse model of DS, which can contribute to cognitive dysfunction in these mice.


Asunto(s)
Síndrome de Down/sangre , Síndrome de Down/enzimología , Hipoxia/sangre , Hipoxia/enzimología , Oxígeno/sangre , Factores de Edad , Animales , Oscuridad , Giro Dentado/enzimología , Síndrome de Down/genética , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Ratones Mutantes , Mitocondrias/enzimología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
18.
Pharmacol Ther ; 151: 87-98, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25795597

RESUMEN

Protein kinases are one of the most studied drug targets in current pharmacological research, as evidenced by the vast number of kinase-targeting agents enrolled in active clinical trials. Dual-specificity Tyrosine phosphorylation-Regulated Kinase 1A (DYRK1A) has been much less studied compared to many other kinases. DYRK1A primary function occurs during early development, where this protein regulates cellular processes related to proliferation and differentiation of neuronal progenitor cells. Although most extensively characterised for its role in brain development, DYRK1A is over-expressed in a variety of diseases including a number of human malignancies, such as haematological and brain cancers. Here we review the accumulating molecular studies that support our understanding of how DYRK1A signalling could underlie these pathological functions. The relevance of DYRK1A in a number of diseases is also substantiated with intensive drug discovery efforts to develop potent and selective inhibitors of DYRK1A. Several classes of DYRK1A inhibitors have recently been disclosed and some molecules are promising leads to develop DYRK1A inhibitors as drugs for DYRK1A-dependent diseases.


Asunto(s)
Neoplasias/enzimología , Enfermedades Neurodegenerativas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Empalme Alternativo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Apoptosis , Benzotiazoles/farmacología , Benzotiazoles/uso terapéutico , Ciclo Celular , Diferenciación Celular , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/enzimología , Activación Enzimática , Dosificación de Gen , Harmina/farmacología , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Neoplasias/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Piridazinas/farmacología , Piridazinas/uso terapéutico , Receptores Notch/metabolismo , Transducción de Señal
19.
Histopathology ; 67(2): 235-44, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25556590

RESUMEN

AIMS: The majority of patients with Down syndrome (DS), trisomy 21, have morphologically abnormal thymuses and present with intrinsic immunological abnormalities affecting mainly the cellular immune response. The aim of this study was to examine whether the expression of functionally important molecules is altered in thymic stromal cells in patients with DS. METHODS AND RESULTS: We analysed thymic tissues from patients with trisomy 13 (n = 4), trisomy 18 (n = 14) and trisomy 21 (n = 13) for histological alterations, and for the expression of functionally important molecules such as ß5t, a thymoproteasome subunit, and cathepsins L and S. In patients with trisomy 13 and trisomy 18, the thymus was morphologically normal or showed only mild depletion of cortical thymocytes. In contrast, the thymus showed variable histological changes in patients with trisomy 21; six of 13 cases showed severe depletion of thymocytes accompanied by the disappearance of thymic lobular architecture. In such thymuses, spindle-shaped keratin-positive cells were densely distributed, and expression of ß5t, but not of cathepsin L, was markedly decreased. CONCLUSIONS: The present study suggests that abnormal thymic architecture and decreased expression of functionally important molecules in thymic stromal cells may be involved in immunological abnormalities in DS patients.


Asunto(s)
Síndrome de Down/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Catepsina L/metabolismo , Catepsinas/metabolismo , Preescolar , Trastornos de los Cromosomas/enzimología , Cromosomas Humanos Par 13/enzimología , Cromosomas Humanos Par 18/enzimología , Síndrome de Down/inmunología , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunidad Celular/fisiología , Lactante , Recién Nacido , Masculino , Coloración y Etiquetado , Células del Estroma/patología , Timo/enzimología , Timo/patología , Trisomía , Síndrome de la Trisomía 13 , Síndrome de la Trisomía 18
20.
Biochim Biophys Acta ; 1842(7): 1144-53, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24735980

RESUMEN

Down syndrome (DS) is the most frequent genetic cause of intellectual disability characterized by the presence of three copies of chromosome 21 (Chr21). Individuals with DS have sufficient neuropathology for a diagnosis of Alzheimer's disease (AD) after the age of 40years. The aim of our study is to gain new insights in the molecular mechanisms impaired in DS subjects that eventually lead to the development of dementia. We evaluate the PI3K/Akt/mTOR axis in the frontal cortex from DS cases (under the age of 40years) and DS with AD neuropathology compared with age-matched controls (Young and Old). The PI3K/Akt/mTOR axis may control several key pathways involved in AD that, if aberrantly regulated, affect amyloid beta (Aß) deposition and tau phosphorylation. Our results show a hyperactivation of PI3K/Akt/mTOR axis in individuals with DS, with and without AD pathology, in comparison with respective controls. The PI3K/Akt/mTOR deregulation results in decreased autophagy, inhibition of IRS1 and GSK3ß activity. Moreover, our data suggest that aberrant activation of the PI3K/Akt/mTOR axis acts in parallel to RCAN1 in phosphorylating tau, in DS and DS/AD. In conclusion, this study provides insights into the neuropathological mechanisms that may be engaged during the development of AD in DS. We suggest that deregulation of this signaling cascade is already evident in young DS cases and persist in the presence of AD pathology. The impairment of the PI3K/Akt/mTOR axis in DS population might represent a key-contributing factor to the neurodegenerative process that culminates in Alzheimer-like dementia.


Asunto(s)
Síndrome de Down/metabolismo , Síndrome de Down/patología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Adulto , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Autofagia/fisiología , Estudios de Casos y Controles , Proteínas de Unión al ADN , Síndrome de Down/enzimología , Femenino , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Proteínas Musculares/metabolismo , Fosforilación , Adulto Joven , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...