Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Eur J Med Chem ; 265: 116098, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38171148

ABSTRACT

Overexpression of the chromosome 21 DYRK1A gene induces morphological defects and cognitive impairments in individuals with Down syndrome (DS) and in DS mice models. Aging neurons of specific brain regions of patients with Alzheimer's disease, DS and Pick's disease have increased DYRK1A immunoreactivity suggesting a possible association of DYRK1A with neurofibrillary tangle pathology. Epigallocatechin-3-gallate (EGCG) displays appreciable inhibition of DYRK1A activity and, contrary to all other published inhibitors, EGCG is a non-competitive inhibitor of DYRK1A. Prenatal exposure to green tea polyphenols containing EGCG protects from brain defects induced by overexpression of DYRK1A. In order to produce more robust and possibly more active analogues of the natural compound EGCG, here we synthetized new EGCG-like molecules with several structural modifications to the EGCG skeleton. We replaced the ester boun of EGCG with a more resistant amide bond. We also replaced the oxygen ring by a methylene group. And finally, we positioned a nitrogen atom within this ring. The selected compound was shown to maintain the non-competitive property of EGCG and to correct biochemical and behavioral defects present in a DS mouse model. In addition it showed high stability and specificity.


Subject(s)
Catechin/analogs & derivatives , Down Syndrome , Humans , Female , Pregnancy , Mice , Animals , Down Syndrome/drug therapy , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Mice, Transgenic , Cognition
2.
Transl Psychiatry ; 13(1): 111, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37015911

ABSTRACT

Early markers are needed for more effective prevention of Alzheimer's disease. We previously showed that individuals with Alzheimer's disease have decreased plasma DYRK1A levels compared to controls. We assessed DYRK1A in the plasma of cognitively healthy elderly volunteers, individuals with either Alzheimer's disease (AD), tauopathies or Down syndrome (DS), and in lymphoblastoids from individuals with DS. DYRK1A levels were inversely correlated with brain amyloid ß burden in asymptomatic elderly individuals and AD patients. Low DYRK1A levels were also detected in patients with tauopathies. Individuals with DS had higher DYRK1A levels than controls, although levels were lower in individuals with DS and with dementia. These data suggest that plasma DYRK1A levels could be used for early detection of at risk individuals of AD and for early detection of AD. We hypothesize that lack of increase of DYRK1A at middle age (40-50 years) could be a warning before the cognitive decline, reflecting increased risk for AD.


Subject(s)
Alzheimer Disease , Down Syndrome , Neurodegenerative Diseases , Tauopathies , Middle Aged , Humans , Aged , Adult , Alzheimer Disease/prevention & control , Alzheimer Disease/diagnosis , Amyloid beta-Peptides , Aging
3.
Alzheimers Dement (N Y) ; 6(1): e12046, 2020.
Article in English | MEDLINE | ID: mdl-32642550

ABSTRACT

INTRODUCTION: An effective therapy has not yet been developed for Alzheimer's disease (AD), in part because pathological changes occur years before clinical symptoms manifest. We recently showed that decreased plasma DYRK1A identifies individuals with mild cognitive impairment (MCI) or AD, and that aged mice have higher DYRK1A levels. METHODS: We assessed DYRK1A in plasma in young/aged controls and in elderly cognitive complainers with low (L) and high (H) brain amyloid load. RESULTS: DYRK1A level increases with age in humans. However, plasma from elderly individuals reporting cognitive complaints showed that the H group had the same DYRK1A level as young adults, suggesting that the age-associated DYRK1A increase is blocked in this group. L and H groups had similar levels of clusterin. DISCUSSION: These results are reflective of early changes in the brain. These observations suggest that plasma DYRK1A and not clusterin could be used to classify elderly memory complainers for risk for amyloid beta pathology.

4.
Pharmacol Ther ; 194: 199-221, 2019 02.
Article in English | MEDLINE | ID: mdl-30268771

ABSTRACT

The dosage of the serine threonine kinase DYRK1A is critical in the central nervous system (CNS) during development and aging. This review analyzes the functions of this kinase by considering its interacting partners and pathways. The role of DYRK1A in controlling the differentiation of prenatal newly formed neurons is presented separately from its role at the pre- and post-synaptic levels in the adult CNS; its effects on synaptic plasticity are also discussed. Because this kinase is positioned at the crossroads of many important processes, genetic dosage errors in this protein produce devastating effects arising from DYRK1A deficiency, such as in MRD7, an autism spectrum disorder, or from DYRK1A excess, such as in Down syndrome. Effects of these errors have been shown in various animal models including Drosophila, zebrafish, and mice. Dysregulation of DYRK1A levels also occurs in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Finally, this review describes inhibitors that have been assessed in vivo. Accurate targeting of DYRK1A levels in the brain, with either inhibitors or activators, is a future research challenge.


Subject(s)
Cognition , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Animals , Autism Spectrum Disorder/metabolism , Down Syndrome/metabolism , Humans , Neurodegenerative Diseases/metabolism , Neurogenesis , Neurons/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Synaptic Transmission , Dyrk Kinases
5.
Sci Rep ; 8(1): 2859, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29434250

ABSTRACT

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.


Subject(s)
Down Syndrome/psychology , Maze Learning/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/administration & dosage , Animals , Cognition/drug effects , Disease Models, Animal , Down Syndrome/enzymology , Humans , Injections, Intraperitoneal , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Mice , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Dyrk Kinases
6.
Dis Model Mech ; 10(10): 1165-1186, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28993310

ABSTRACT

Down syndrome is caused by trisomy of chromosome 21. To date, a multiplicity of mouse models with Down-syndrome-related features has been developed to understand this complex human chromosomal disorder. These mouse models have been important for determining genotype-phenotype relationships and identification of dosage-sensitive genes involved in the pathophysiology of the condition, and in exploring the impact of the additional chromosome on the whole genome. Mouse models of Down syndrome have also been used to test therapeutic strategies. Here, we provide an overview of research in the last 15 years dedicated to the development and application of rodent models for Down syndrome. We also speculate on possible and probable future directions of research in this fast-moving field. As our understanding of the syndrome improves and genome engineering technologies evolve, it is necessary to coordinate efforts to make all Down syndrome models available to the community, to test therapeutics in models that replicate the whole trisomy and design new animal models to promote further discovery of potential therapeutic targets.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Chromosomes, Mammalian/genetics , Down Syndrome/genetics , Trisomy , Animals , Behavior, Animal , Cognition , Disease Models, Animal , Down Syndrome/psychology , Down Syndrome/therapy , Gene Dosage , Genetic Predisposition to Disease , Humans , Mice , Mice, Transgenic , Phenotype
7.
Front Behav Neurosci ; 9: 267, 2015.
Article in English | MEDLINE | ID: mdl-26539088

ABSTRACT

Cognitive impairment in Down syndrome (DS) has been linked to increased synaptic inhibition. The underlying mechanisms remain unknown, but memory deficits are rescued in DS mouse models by drugs targeting GABA receptors. Similarly, administration of epigallocatechin gallate (EGCG)-containing extracts rescues cognitive phenotypes in Ts65Dn mice, potentially through GABA pathway. Some developmental and cognitive alterations have been traced to increased expression of the serine-threonine kinase DYRK1A on Hsa21. To better understand excitation/inhibition balance in DS, we investigated the consequences of long-term (1-month) treatment with EGCG-containing extracts in adult mBACtgDyrk1a mice that overexpress Dyrk1a. Administration of POL60 rescued components of GABAergic and glutamatergic pathways in cortex and hippocampus but not cerebellum. An intermediate dose (60 mg/kg) of decaffeinated green tea extract (MGTE) acted on components of both GABAergic and glutamatergic pathways and rescued behavioral deficits as demonstrated on the alternating paradigm, but did not rescue protein level of GABA-synthesizing GAD67. These results indicate that excessive synaptic inhibition in people with DS may be attributable, in large part, to increased DYRK1A dosage. Thus, controlling the level of active DYRK1A is a clear issue for DS therapy. This study also defines a panel of synaptic markers for further characterization of DS treatments in murine models.

8.
EBioMedicine ; 2(2): 120-34, 2015.
Article in English | MEDLINE | ID: mdl-26137553

ABSTRACT

Alterations in cerebral cortex connectivity lead to intellectual disability and in Down syndrome, this is associated with a deficit in cortical neurons that arises during prenatal development. However, the pathogenic mechanisms that cause this deficit have not yet been defined. Here we show that the human DYRK1A kinase on chromosome 21 tightly regulates the nuclear levels of Cyclin D1 in embryonic cortical stem (radial glia) cells, and that a modest increase in DYRK1A protein in transgenic embryos lengthens the G1 phase in these progenitors. These alterations promote asymmetric proliferative divisions at the expense of neurogenic divisions, producing a deficit in cortical projection neurons that persists in postnatal stages. Moreover, radial glial progenitors in the Ts65Dn mouse model of Down syndrome have less Cyclin D1, and Dyrk1a is the triplicated gene that causes both early cortical neurogenic defects and decreased nuclear Cyclin D1 levels in this model. These data provide insights into the mechanisms that couple cell cycle regulation and neuron production in cortical neural stem cells, emphasizing that the deleterious effect of DYRK1A triplication in the formation of the cerebral cortex begins at the onset of neurogenesis, which is relevant to the search for early therapeutic interventions in Down syndrome.


Subject(s)
Cyclin D1/metabolism , Down Syndrome/genetics , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Animals , Cerebral Cortex/pathology , Disease Models, Animal , G1 Phase/genetics , Gene Dosage/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Telencephalon/cytology , Trisomy/genetics , Dyrk Kinases
9.
J Med Chem ; 56(23): 9569-85, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24188002

ABSTRACT

A series of 3,5-diaryl-1H-pyrrolo[2,3-b]pyridines were synthesized and evaluated for inhibition of DYRKIA kinase in vitro. Derivatives having hydroxy groups on the aryl moieties (2c, 2j-l) demonstrated high inhibitory potencies with Kis in the low nanomolar range. Their methoxy analogues were up to 100 times less active. Docking studies at the ATP binding site suggested that these compounds bind tightly to this site via a network of multiple H-bonds with the peptide backbone. None of the active compounds were cytotoxic to KB cells at 10(-6) M. Kinase profiling revealed that compound 2j showed 2-fold selectivity for DYRK1A with respect to DYRK2 and DYRK3.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Indoles/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Bonding , Indoles/pharmacology , KB Cells , Molecular Docking Simulation , Pyridines/pharmacology , Dyrk Kinases
10.
Genome Res ; 23(9): 1410-21, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23783273

ABSTRACT

Congenital heart defect (CHD) occurs in 40% of Down syndrome (DS) cases. While carrying three copies of chromosome 21 increases the risk for CHD, trisomy 21 itself is not sufficient to cause CHD. Thus, additional genetic variation and/or environmental factors could contribute to the CHD risk. Here we report genomic variations that in concert with trisomy 21, determine the risk for CHD in DS. This case-control GWAS includes 187 DS with CHD (AVSD = 69, ASD = 53, VSD = 65) as cases, and 151 DS without CHD as controls. Chromosome 21-specific association studies revealed rs2832616 and rs1943950 as CHD risk alleles (adjusted genotypic P-values <0.05). These signals were confirmed in a replication cohort of 92 DS-CHD cases and 80 DS-without CHD (nominal P-value 0.0022). Furthermore, CNV analyses using a customized chromosome 21 aCGH of 135K probes in 55 DS-AVSD and 53 DS-without CHD revealed three CNV regions associated with AVSD risk (FDR ≤ 0.05). Two of these regions that are located within the previously identified CHD region on chromosome 21 were further confirmed in a replication study of 49 DS-AVSD and 45 DS- without CHD (FDR ≤ 0.05). One of these CNVs maps near the RIPK4 gene, and the second includes the ZBTB21 (previously ZNF295) gene, highlighting the potential role of these genes in the pathogenesis of CHD in DS. We propose that the genetic architecture of the CHD risk of DS is complex and includes trisomy 21, and SNP and CNV variations in chromosome 21. In addition, a yet-unidentified genetic variation in the rest of the genome may contribute to this complex genetic architecture.


Subject(s)
DNA Copy Number Variations , Down Syndrome/diagnosis , Heart Defects, Congenital/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Chromosomes, Human, Pair 21/genetics , DNA-Binding Proteins/genetics , Down Syndrome/complications , Genetic Predisposition to Disease , Genome-Wide Association Study , Heart Defects, Congenital/etiology , Humans , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics
11.
Hum Mol Genet ; 22(14): 2775-84, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23512985

ABSTRACT

Down syndrome (DS) results from the triplication of approximately 300 human chromosome 21 (Hsa21) genes and affects almost all body organs. Children with DS have defects in visual processing that may have a negative impact on their daily life and cognitive development. However, there is little known about the genes and pathogenesis underlying these defects. Here, we show morphometric in vivo data indicating that the neural retina is thicker in DS individuals than in the normal population. A similar thickening specifically affecting the inner part of the retina was also observed in a trisomic model of DS, the Ts65Dn mouse. Increased retinal size and cellularity in this model correlated with abnormal retinal function and resulted from an impaired caspase-9-mediated apoptosis during development. Moreover, we show that mice bearing only one additional copy of Dyrk1a have the same retinal phenotype as Ts65Dn mice and normalization of Dyrk1a gene copy number in Ts65Dn mice completely rescues both, morphological and functional phenotypes. Thus, triplication of Dyrk1a is necessary and sufficient to cause the retinal phenotype described in the trisomic model. Our data demonstrate for the first time the implication of DYRK1A overexpression in a developmental alteration of the central nervous system associated with DS, thereby providing insights into the aetiology of neurosensorial dysfunction in a complex disease.


Subject(s)
Down Syndrome/enzymology , Down Syndrome/genetics , Gene Dosage , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Retina/anatomy & histology , Adult , Animals , Apoptosis , Caspase 9/genetics , Caspase 9/metabolism , Disease Models, Animal , Down Syndrome/physiopathology , Female , Gene Amplification , Humans , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Retina/cytology , Retina/enzymology , Young Adult , Dyrk Kinases
12.
Hum Mol Genet ; 21(14): 3156-72, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22511594

ABSTRACT

Enlarged early endosomes have been observed in neurons and fibroblasts in Down syndrome (DS). These endosome abnormalities have been implicated in the early development of Alzheimer's disease (AD) pathology in these subjects. Here, we show the presence of enlarged endosomes in blood mononuclear cells and lymphoblastoid cell lines (LCLs) from individuals with DS using immunofluorescence and confocal microscopy. Genotype-phenotype correlations in LCLs carrying partial trisomies 21 revealed that triplication of a 2.56 Mb locus in 21q22.11 is associated with the endosomal abnormalities. This locus contains the gene encoding the phosphoinositide phosphatase synaptojanin 1 (SYNJ1), a key regulator of the signalling phospholipid phosphatidylinositol-4,5-biphosphate that has been shown to regulate clathrin-mediated endocytosis. We found that SYNJ1 transcripts are increased in LCLs from individuals with DS and that overexpression of SYNJ1 in a neuroblastoma cell line as well as in transgenic mice leads to enlarged endosomes. Moreover, the proportion of enlarged endosomes in fibroblasts from an individual with DS was reduced after silencing SYNJ1 expression with RNA interference. In LCLs carrying amyloid precursor protein (APP) microduplications causing autosomal dominant early-onset AD, enlarged endosomes were absent, suggesting that APP overexpression alone is not involved in the modification of early endosomes in this cell type. These findings provide new insights into the contribution of SYNJ1 overexpression to the endosomal changes observed in DS and suggest an attractive new target for rescuing endocytic dysfunction and lipid metabolism in DS and in AD.


Subject(s)
Down Syndrome/enzymology , Endosomes/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Trisomy , Animals , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 21/enzymology , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Endosomes/metabolism , Humans , Mice , Mice, Transgenic
13.
Neurobiol Dis ; 46(1): 190-203, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22293606

ABSTRACT

Copy number variation in a small region of chromosome 21 containing DYRK1A produces morphological and cognitive alterations in human. In mouse models, haploinsufficiency results in microcephaly, and a human DYRK1A gain-of-function model (three alleles) exhibits increased brain volume. To investigate these developmental aspects, we used a murine BAC clone containing the entire gene to construct an overexpression model driven by endogenous regulatory sequences. We compared this new model to two other mouse models with three copies of Dyrk1a, YACtgDyrk1a and Ts65Dn, as well as the loss-of-function model with one copy (Dyrk1a(+/-)). Growth, viability, brain weight, and brain volume depended strongly upon gene copy number. Brain region-specific variations observed in gain-of-function models mirror their counterparts in the loss-of-function model. Some variations, such as increased volume of the superior colliculus and ventricles, were observed in both the BAC transgenic and Ts65Dn mice. Using unbiased stereology we found that, in the cortex, neuron density is inversely related to Dyrk1a copy number but, in thalamic nuclei, neuron density is directly related to copy number. In addition, six genes involved either in cell division (Ccnd1 and pAkt) or in neuronal machinery (Gap43, Map2, Syp, Snap25) were regulated by Dyrk1a throughout development, from birth to adult. These results imply that Dyrk1a expression alters different cellular processes during brain development. Dyrk1a, then, has two roles in the development process: shaping the brain and controlling the structure of neuronal components.


Subject(s)
Brain/embryology , Brain/physiology , Gene Expression Regulation, Developmental/physiology , Microcephaly/genetics , Neurons/physiology , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Animals , Apoptosis/physiology , Brain/cytology , Cell Proliferation , DNA Copy Number Variations/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcephaly/pathology , Microcephaly/physiopathology , Neurons/cytology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Dyrk Kinases
14.
PLoS One ; 7(1): e29056, 2012.
Article in English | MEDLINE | ID: mdl-22253703

ABSTRACT

BACKGROUND: The cystathionine ß-synthase (CBS) gene, located on human chromosome 21q22.3, is a good candidate for playing a role in the Down Syndrome (DS) cognitive profile: it is overexpressed in the brain of individuals with DS, and it encodes a key enzyme of sulfur-containing amino acid (SAA) metabolism, a pathway important for several brain physiological processes. METHODOLOGY/PRINCIPAL FINDINGS: Here, we have studied the neural consequences of CBS overexpression in a transgenic mouse line (60.4P102D1) expressing the human CBS gene under the control of its endogenous regulatory regions. These mice displayed a ∼2-fold increase in total CBS proteins in different brain areas and a ∼1.3-fold increase in CBS activity in the cerebellum and the hippocampus. No major disturbance of SAA metabolism was observed, and the transgenic mice showed normal behavior in the rotarod and passive avoidance tests. However, we found that hippocampal synaptic plasticity is facilitated in the 60.4P102D1 line. CONCLUSION/SIGNIFICANCE: We demonstrate that CBS overexpression has functional consequences on hippocampal neuronal networks. These results shed new light on the function of the CBS gene, and raise the interesting possibility that CBS overexpression might have an advantageous effect on some cognitive functions in DS.


Subject(s)
Brain/physiology , Cystathionine beta-Synthase/metabolism , Amino Acids, Sulfur/metabolism , Animals , Behavior, Animal/physiology , Blotting, Western , Gene Dosage , Humans , Long-Term Potentiation/physiology , Metabolic Networks and Pathways , Metabolome , Mice , Mice, Transgenic , Organ Specificity , Phenotype , Synapses/metabolism , Synaptic Transmission/physiology , Transgenes/genetics
15.
PLoS One ; 4(2): e4606, 2009.
Article in English | MEDLINE | ID: mdl-19242551

ABSTRACT

Individuals with partial HSA21 trisomies and mice with partial MMU16 trisomies containing an extra copy of the DYRK1A gene present various alterations in brain morphogenesis. They present also learning impairments modeling those encountered in Down syndrome. Previous MRI and histological analyses of a transgenic mice generated using a human YAC construct that contains five genes including DYRK1A reveal that DYRK1A is involved, during development, in the control of brain volume and cell density of specific brain regions. Gene dosage correction induces a rescue of the brain volume alterations. DYRK1A is also involved in the control of synaptic plasticity and memory consolidation. Increased gene dosage results in brain morphogenesis defects, low BDNF levels and mnemonic deficits in these mice. Epigallocatechin gallate (EGCG) - a member of a natural polyphenols family, found in great amount in green tea leaves - is a specific and safe DYRK1A inhibitor. We maintained control and transgenic mice overexpressing DYRK1A on two different polyphenol-based diets, from gestation to adulthood. The major features of the transgenic phenotype were rescued in these mice.


Subject(s)
Catechin/analogs & derivatives , Flavonoids/pharmacology , Phenols/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Animals , Antioxidants/pharmacology , Brain/growth & development , Catechin/pharmacology , Gene Dosage , Humans , Memory , Mice , Mice, Transgenic , Neuronal Plasticity , Polyphenols , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Tea , Dyrk Kinases
16.
Eur J Hum Genet ; 17(4): 454-66, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19002211

ABSTRACT

Down syndrome (DS) is one of the most frequent congenital birth defects, and the most common genetic cause of mental retardation. In most cases, DS results from the presence of an extra copy of chromosome 21. DS has a complex phenotype, and a major goal of DS research is to identify genotype-phenotype correlations. Cases of partial trisomy 21 and other HSA21 rearrangements associated with DS features could identify genomic regions associated with specific phenotypes. We have developed a BAC array spanning HSA21q and used array comparative genome hybridization (aCGH) to enable high-resolution mapping of pathogenic partial aneuploidies and unbalanced translocations involving HSA21. We report the identification and mapping of 30 pathogenic chromosomal aberrations of HSA21 consisting of 19 partial trisomies and 11 partial monosomies for different segments of HSA21. The breakpoints have been mapped to within approximately 85 kb. The majority of the breakpoints (26 of 30) for the partial aneuploidies map within a 10-Mb region. Our data argue against a single DS critical region. We identify susceptibility regions for 25 phenotypes for DS and 27 regions for monosomy 21. However, most of these regions are still broad, and more cases are needed to narrow down the phenotypic maps to a reasonable number of candidate genomic elements per phenotype.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Phenotype , Trisomy/genetics , Abnormalities, Multiple/genetics , Comparative Genomic Hybridization , Genotype , Humans
17.
Biochim Biophys Acta ; 1782(7-8): 482-8, 2008.
Article in English | MEDLINE | ID: mdl-18541157

ABSTRACT

Cystathionine beta synthase deficiency induces hyperhomocysteinemia which is considered as a risk factor for vascular diseases. Studies underlined the importance of altered cellular redox reactions in hyperhomocysteinemia-induced vascular pathologies. Nevertheless, hyperhomocysteinemia also induces hepatic dysfunction which may accelerate the development of vascular pathologies by modifying cholesterol homeostasis. The aim of the present study was to analyze the modifications of redox state in the liver of heterozygous cystathionine beta synthase-deficient mice, a murine model of hyperhomocysteinemia. In this purpose, we quantified levels of reactive oxygen and nitrogen species and we assayed activities of main antioxidant enzymes. We found that cystathionine beta synthase deficiency induced NADPH oxidase activation. However, there was no accumulation of reactive oxygen (superoxide anion, hydrogen peroxide) and nitrogen (nitrite, peroxynitrite) species. On the contrary, hepatic hydrogen peroxide level was decreased independently of an activation of glutathione-dependent mechanisms. In fact, cystathionine beta synthase deficiency had no effect on glutathione peroxidase, glutathione reductase and glutathione S-transferase activities. However, we found a 50% increase in hepatic catalase activity without any variation of expression. These findings demonstrate that cystathionine beta synthase deficiency initiates redox disequilibrium in the liver. However, the activation of catalase attenuates oxidative impairments.


Subject(s)
Catalase/metabolism , Cystathionine beta-Synthase/deficiency , Hydrogen Peroxide/metabolism , Hyperhomocysteinemia/metabolism , Liver/metabolism , Animals , Base Sequence , Catalase/genetics , Cystathionine beta-Synthase/genetics , DNA Primers/genetics , Disease Models, Animal , Female , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Heterozygote , Homocystinuria/genetics , Homocystinuria/metabolism , Humans , Hyperhomocysteinemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NADPH Oxidases/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL
...