Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
2.
Am J Hum Genet ; 86(5): 749-64, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20466091

ABSTRACT

Chromosomal microarray (CMA) is increasingly utilized for genetic testing of individuals with unexplained developmental delay/intellectual disability (DD/ID), autism spectrum disorders (ASD), or multiple congenital anomalies (MCA). Performing CMA and G-banded karyotyping on every patient substantially increases the total cost of genetic testing. The International Standard Cytogenomic Array (ISCA) Consortium held two international workshops and conducted a literature review of 33 studies, including 21,698 patients tested by CMA. We provide an evidence-based summary of clinical cytogenetic testing comparing CMA to G-banded karyotyping with respect to technical advantages and limitations, diagnostic yield for various types of chromosomal aberrations, and issues that affect test interpretation. CMA offers a much higher diagnostic yield (15%-20%) for genetic testing of individuals with unexplained DD/ID, ASD, or MCA than a G-banded karyotype ( approximately 3%, excluding Down syndrome and other recognizable chromosomal syndromes), primarily because of its higher sensitivity for submicroscopic deletions and duplications. Truly balanced rearrangements and low-level mosaicism are generally not detectable by arrays, but these are relatively infrequent causes of abnormal phenotypes in this population (<1%). Available evidence strongly supports the use of CMA in place of G-banded karyotyping as the first-tier cytogenetic diagnostic test for patients with DD/ID, ASD, or MCA. G-banded karyotype analysis should be reserved for patients with obvious chromosomal syndromes (e.g., Down syndrome), a family history of chromosomal rearrangement, or a history of multiple miscarriages.


Subject(s)
Chromosome Disorders/genetics , Congenital Abnormalities/genetics , Developmental Disabilities/genetics , Child , Chromosome Banding , Humans , Karyotyping
3.
Free Radic Biol Med ; 48(11): 1501-12, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20188820

ABSTRACT

Mn superoxide dismutase (MnSOD) is an important mitochondrial antioxidant enzyme, and elevated MnSOD levels have been shown to reduce tumor growth in part by suppressing cell proliferation. Studies with fibroblasts have shown that increased MnSOD expression prolongs cell cycle transition time in G1/S and favors entrance into the quiescent state. To determine if the same effect occurs during tissue regeneration in vivo, we used a transgenic mouse system with liver-specific MnSOD expression and a partial hepatectomy paradigm to induce synchronized in vivo cell proliferation during liver regeneration. We show in this experimental system that a 2.6-fold increase in MnSOD activity leads to delayed entry into S phase, as measured by reduction in bromodeoxyuridine (BrdU) incorporation and decreased expression of proliferative cell nuclear antigen (PCNA). Thus, compared to control mice with baseline MnSOD levels, transgenic mice with increased MnSOD expression in the liver have 23% fewer BrdU-positive cells and a marked attenuation of PCNA expression. The increase in MnSOD activity also leads to an increase in the mitochondrial form of thioredoxin (thioredoxin 2), but not in several other peroxidases examined, suggesting the importance of thioredoxin 2 in maintaining redox balance in mitochondria with elevated levels of MnSOD.


Subject(s)
Cell Cycle/genetics , Mitochondria/enzymology , Superoxide Dismutase/genetics , Thioredoxins/biosynthesis , Aconitate Hydratase/metabolism , Animals , Antioxidants/metabolism , Female , Liver Regeneration/physiology , Male , Mice , Mice, Transgenic , Mitochondria/genetics , Proliferating Cell Nuclear Antigen/biosynthesis , Sex Factors , Signal Transduction/physiology , Superoxide Dismutase/biosynthesis , Thioredoxins/genetics , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis , Up-Regulation
4.
Cereb Cortex ; 20(5): 1131-43, 2010 May.
Article in English | MEDLINE | ID: mdl-19710359

ABSTRACT

Down syndrome (DS) is the most common cause of mental retardation. Although structural and neurogenic abnormalities have been shown in the brains of DS patients, the molecular etiology is still unknown. To define it, we have performed structural and histological examinations of the brains of Ts1Cje and Ts2Cje, 2 mouse models for DS. These mice carry different length of trisomic segments of mouse chromosome 16 that are orthologous to human chromosome 21. At 3 months of age, ventricular enlargements were observed in both Ts1Cje and Ts2Cje brains at a similar degree. Both mice also showed decreases of the number of doublecortin-positive neuroblasts and thymidine-analog BrdU-labeled proliferating cells in the subventricular zone of the lateral ventricles (LVs) and in the hippocampal dentate gyrus at a similar degree, suggesting impaired adult neurogenesis. Additionally, at embryonic day 14.5, both strains of mice, when compared with diploid littermates, had smaller brains and decreased cortical neurogenesis that could possibly contribute to the ventricular enlargements observed in adulthood. Our findings suggest that the trisomic segment of the Ts1Cje mouse, which is shared with Ts2Cje, contains the genes that are responsible for these abnormal phenotypes and could be relevant to the mental retardation associated with DS.


Subject(s)
Cerebral Ventricles/pathology , Chromosomes, Mammalian/genetics , Down Syndrome/genetics , Neurogenesis , Trisomy/genetics , Trisomy/physiopathology , Animals , Cell Proliferation , Cerebral Ventricles/embryology , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Chromosomes, Human, Pair 21 , Disease Models, Animal , Doublecortin Domain Proteins , Down Syndrome/pathology , Down Syndrome/physiopathology , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Pregnancy
5.
J Neurochem ; 110(6): 1965-76, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19645748

ABSTRACT

Elevated oxidative stress has been suggested to be associated with the features of Down's syndrome (DS). We previously reported increased oxidative stress in cultured cells from the embryonic brain of Ts1Cje, a mouse genetic DS model. However, since in vivo evidence for increased oxidative stress is lacking, we here examined lipid peroxidation, a typical marker of oxidative stress, in the brains of Ts1Cje and another DS mouse model Ts2Cje with an overlapping but larger trisomic segment. Accumulations of proteins modified with the lipid peroxidation-derived products, 13-hydroperoxy-9Z,11E-octadecadienoic acid and 4-hydroxy-2-nonenal were markedly increased in Ts1Cje and Ts2Cje brains. Analysis with oxidation-sensitive fluorescent probe also showed that reactive oxygen species themselves were increased in Ts1Cje brain. However, electron spin resonance analysis of microdialysate from the hippocampus of Ts1Cje showed that antioxidant activity remained unaffected, suggesting that the reactive oxygen species production was accelerated in Ts1Cje. Proteomics approaches with mass spectrometry identified the proteins modified with 13-hydroperoxy-9Z,11E-octadecadienoic acid and/or 4-hydroxy-2-nonenal to be involved in either ATP generation, the neuronal cytoskeleton or antioxidant activity. Structural or functional impairments of these proteins by such modifications may contribute to the DS features such as cognitive impairment that are present in the Ts1Cje mouse.


Subject(s)
Brain/metabolism , Down Syndrome/metabolism , Down Syndrome/physiopathology , Lipid Peroxidation/physiology , Age Factors , Aldehydes/metabolism , Animals , Brain/pathology , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/pathology , Electrophoresis, Gel, Two-Dimensional/methods , Female , Gene Expression Regulation/genetics , Humans , Linoleic Acids/metabolism , Lipid Peroxides/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis , Reactive Oxygen Species/metabolism , Trisomy/genetics
6.
J Gerontol A Biol Sci Med Sci ; 64(11): 1114-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19633237

ABSTRACT

Genetic manipulations of Mn superoxide dismutase (MnSOD), SOD2 expression have demonstrated that altering the level of MnSOD activity is critical for cellular function and life span in invertebrates. In mammals, Sod2 homozygous knockout mice die shortly after birth, and alterations of MnSOD levels are correlated with changes in oxidative damage and in the generation of mitochondrial reactive oxygen species. In this study, we directly tested the effects of overexpressing MnSOD in young (4-6 months) and old (26-28 months) mice on mitochondrial function, levels of oxidative damage or stress, life span, and end-of-life pathology. Our data show that an approximately twofold overexpression of MnSOD throughout life in mice resulted in decreased lipid peroxidation, increased resistance against paraquat-induced oxidative stress, and decreased age-related decline in mitochondrial ATP production. However, this change in MnSOD expression did not alter either life span or age-related pathology.


Subject(s)
Aging/metabolism , Superoxide Dismutase/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Muscle/physiology , Muscle, Skeletal/pathology , Organ Size , Oxidative Stress , Reactive Oxygen Species/metabolism
7.
Aging Cell ; 8(1): 73-5, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19077044

ABSTRACT

We evaluated the effect of overexpressing antioxidant enzymes on the lifespans of transgenic mice that overexpress copper zinc superoxide dismutase (CuZnSOD), catalase, or combinations of either CuZnSOD and catalase or CuZnSOD and manganese superoxide dismutase (MnSOD). Our results show that the overexpression of these major antioxidant enzymes, which are known to scavenge superoxide and hydrogen peroxide in the cytosolic and mitochondrial compartments, is insufficient to extend lifespan in mice.


Subject(s)
Catalase/biosynthesis , Longevity/physiology , Superoxide Dismutase/biosynthesis , Animals , Catalase/genetics , Disease Models, Animal , Humans , Hydrogen Peroxide/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics
8.
Toxicol Appl Pharmacol ; 234(3): 326-38, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19063909

ABSTRACT

Alcohol consumption increases reactive oxygen species formation and lipid peroxidation, whose products can damage mitochondrial DNA (mtDNA) and alter mitochondrial function. A possible role of manganese superoxide dismutase (MnSOD) on these effects has not been investigated. To test whether MnSOD overexpression modulates alcohol-induced mitochondrial alterations, we added ethanol to the drinking water of transgenic MnSOD-overexpressing (TgMnSOD) mice and their wild type (WT) littermates for 7 weeks. In TgMnSOD mice, alcohol administration further increased the activity of MnSOD, but decreased cytosolic glutathione as well as cytosolic glutathione peroxidase activity and peroxisomal catalase activity. Whereas ethanol increased cytochrome P-450 2E1 and mitochondrial ROS generation in both WT and TgMnSOD mice, hepatic iron, lipid peroxidation products and respiratory complex I protein carbonyls were only increased in ethanol-treated TgMnSOD mice but not in WT mice. In ethanol-fed TgMnSOD mice, but not ethanol-fed WT mice, mtDNA was depleted, and mtDNA lesions blocked the progress of polymerases. The iron chelator, DFO prevented hepatic iron accumulation, lipid peroxidation, protein carbonyl formation and mtDNA depletion in alcohol-treated TgMnSOD mice. Alcohol markedly decreased the activities of complexes I, IV and V of the respiratory chain in TgMnSOD, with absent or lesser effects in WT mice. There was no inflammation, apoptosis or necrosis, and steatosis was similar in ethanol-treated WT and TgMnSOD mice. In conclusion, prolonged alcohol administration selectively triggers iron accumulation, lipid peroxidation, respiratory complex I protein carbonylation, mtDNA lesions blocking the progress of polymerases, mtDNA depletion and respiratory complex dysfunction in TgMnSOD mice but not in WT mice.


Subject(s)
Alcohol Drinking/adverse effects , DNA Damage , DNA, Mitochondrial/metabolism , Ethanol/toxicity , Liver/drug effects , Mitochondria, Liver/drug effects , Superoxide Dismutase/metabolism , Animals , Body Weight , Caspase 3/metabolism , Catalase/metabolism , Cytochrome P-450 CYP2E1/metabolism , DNA-Binding Proteins/metabolism , Deferoxamine/pharmacology , Down-Regulation , Electron Transport Complex I/metabolism , Ethanol/blood , Ethanol/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , High Mobility Group Proteins/metabolism , Iron/metabolism , Iron Chelating Agents/pharmacology , Lipid Peroxidation/drug effects , Liver/enzymology , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Liver/enzymology , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Thiobarbituric Acid Reactive Substances/metabolism , Transcription Factors/metabolism , Up-Regulation
9.
Am J Hum Genet ; 83(3): 388-400, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18771760

ABSTRACT

Down syndrome (DS) is the most common cause of mental retardation. Many neural phenotypes are shared between DS individuals and DS mouse models; however, the common underlying molecular pathogenetic mechanisms remain unclear. Using a transchromosomic model of DS, we show that a 30%-60% reduced expression of Nrsf/Rest (a key regulator of pluripotency and neuronal differentiation) is an alteration that persists in trisomy 21 from undifferentiated embryonic stem (ES) cells to adult brain and is reproducible across several DS models. Using partially trisomic ES cells, we map this effect to a three-gene segment of HSA21, containing DYRK1A. We independently identify the same locus as the most significant eQTL controlling REST expression in the human genome. We show that specifically silencing the third copy of DYRK1A rescues Rest levels, and we demonstrate altered Rest expression in response to inhibition of DYRK1A expression or kinase activity, and in a transgenic Dyrk1A mouse. We reveal that undifferentiated trisomy 21 ES cells show DYRK1A-dose-sensitive reductions in levels of some pluripotency regulators, causing premature expression of transcription factors driving early endodermal and mesodermal differentiation, partially overlapping recently reported downstream effects of Rest +/-. They produce embryoid bodies with elevated levels of the primitive endoderm progenitor marker Gata4 and a strongly reduced neuroectodermal progenitor compartment. Our results suggest that DYRK1A-mediated deregulation of REST is a very early pathological consequence of trisomy 21 with potential to disturb the development of all embryonic lineages, warranting closer research into its contribution to DS pathology and new rationales for therapeutic approaches.


Subject(s)
Down Syndrome/metabolism , Embryonic Stem Cells/pathology , Gene Dosage , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Repressor Proteins/physiology , Animals , Cell Differentiation , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/pathology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Transgenic , Pluripotent Stem Cells/pathology , Pluripotent Stem Cells/physiology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Quantitative Trait Loci , Repressor Proteins/genetics , Dyrk Kinases
10.
J Gen Virol ; 89(Pt 7): 1777-1788, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18559949

ABSTRACT

Prion diseases are caused by conversion of a normally folded, non-pathogenic isoform of the prion protein (PrP(C)) to a misfolded, pathogenic isoform (PrP(Sc)). Prion inoculation experiments in mice expressing homologous PrP(C) molecules on different genetic backgrounds displayed different incubation times, indicating that the conversion reaction may be influenced by other gene products. To identify genes that contribute to prion pathogenesis, we analysed incubation times of prions in mice in which the gene product was inactivated, knocked out or overexpressed. We tested 20 candidate genes, because their products either colocalize with PrP, are associated with Alzheimer's disease, are elevated during prion disease, or function in PrP-mediated signalling, PrP glycosylation, or protein maintenance. Whereas some of the candidates tested may have a role in the normal function of PrP(C), our data show that many genes previously implicated in prion replication have no discernible effect on the pathogenesis of prion disease. While most genes tested did not significantly affect survival times, ablation of the amyloid beta (A4) precursor protein (App) or interleukin-1 receptor, type I (Il1r1), and transgenic overexpression of human superoxide dismutase 1 (SOD1) prolonged incubation times by 13, 16 and 19 %, respectively.


Subject(s)
Prion Diseases/genetics , Prions/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Gene Dosage , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prions/genetics , Receptors, Interleukin-1 Type I/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Analysis
11.
Physiol Genomics ; 34(1): 112-26, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18445702

ABSTRACT

How higher organisms respond to elevated oxidative stress in vivo is poorly understood. Therefore, we measured oxidative stress parameters and gene expression alterations (Affymetrix arrays) in the liver caused by elevated reactive oxygen species induced in vivo by diquat or by genetic ablation of the major antioxidant enzymes CuZn-superoxide dismutase (Sod1) and glutathione peroxidase-1 (Gpx1). Diquat (50 mg/kg) treatment resulted in a significant increase in oxidative damage within 3-6 h in wild-type mice without any lethality. In contrast, treatment of Sod1(-/-) or Gpx1(-/-) mice with a similar concentration of diquat resulted in a significant increase in oxidative damage within an hour of treatment and was lethal, i.e., these mice are extremely sensitive to the oxidative stress generated by diquat. The expression response to elevated oxidative stress in vivo does not involve an upregulation of classic antioxidant genes, although long-term oxidative stress in Sod1(-/-) mice leads to a significant upregulation of thiol antioxidants (e.g., Mt1, Srxn1, Gclc, Txnrd1), which appears to be mediated by the redox-sensitive transcription factor Nrf2. The main finding of our study is that the common response to elevated oxidative stress with diquat treatment in wild-type, Gpx1(-/-), and Sod1(-/-) mice and in untreated Sod1(-/-) mice is an upregulation of p53 target genes (p21, Gdf15, Plk3, Atf3, Trp53inp1, Ddit4, Gadd45a, Btg2, Ndrg1). A retrospective comparison with previous studies shows that induction of these p53 target genes is a conserved expression response to oxidative stress, in vivo and in vitro, in different species and different cells/organs.


Subject(s)
Gene Expression Profiling , Oxidative Stress/genetics , Animals , Antioxidants/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA/metabolism , Diquat/toxicity , Gene Expression Regulation/drug effects , Glutathione Peroxidase/deficiency , Lipid Peroxidation/drug effects , Liver Diseases/enzymology , Liver Diseases/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Superoxide Dismutase/deficiency , Tumor Suppressor Protein p53/metabolism , Glutathione Peroxidase GPX1
12.
J Comp Neurol ; 504(4): 329-45, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17663443

ABSTRACT

Down syndrome (DS) is caused by trisomy of human chromosome 21. Because Ts65Dn and Ts1Cje mice are segmentally trisomic for a region of mouse chromosome 16, they genetically model DS and are used to study pathogenic mechanisms. Previously, we provided evidence for changes in both the structure and function of pre- and postsynaptic elements in the Ts65Dn mouse. Striking changes were evident in the size of the dendritic spines and in the ability to induce long-term potentiation (LTP) in the fascia dentata (FD). To explore the genetic basis for these changes, we examined Ts1Cje mice, which are trisomic for a completely overlapping but smaller segment of mouse chromosome 16. As in the Ts65Dn mouse, there was a regionally selective decrease in the density of dendritic spines ( approximately 12%), an increase in the size of spine heads ( approximately 26%), a decrease in the length of spine necks ( approximately 26%), and reorganization of inhibitory inputs with a relative decrease in inputs to dendrite shafts and spine heads and a significant increase to the necks of spines (6.4%). Thus, all of the Ts65Dn phenotypes were present, but they were significantly less severe. In contrast, and just as was the case for the Ts65Dn mouse, LTP could not be induced unless the selective gamma-aminobutyric acid (GABA)(A) receptor antagonist picrotoxin was applied. Therefore, there was conservation of important synaptic phenotypes in the Ts1Cje mice. The analysis of data from this and earlier studies points to genotype-phenotype linkages in DS whose complexity ranges from relatively simple to quite complex.


Subject(s)
Cognition Disorders/physiopathology , Dendritic Spines/pathology , Down Syndrome/physiopathology , Synapses/pathology , Trisomy/physiopathology , Animals , Brain/pathology , Brain/physiopathology , Cognition Disorders/complications , Cognition Disorders/genetics , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Disease Models, Animal , Down Syndrome/complications , Down Syndrome/genetics , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry , Long-Term Potentiation , Male , Maze Learning , Mice , Mice, Mutant Strains , Neural Inhibition/genetics , Neural Pathways/pathology , Neural Pathways/physiopathology , Organ Size , Phenotype , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Synapses/genetics , Trisomy/genetics
14.
Hum Mol Genet ; 15(18): 2752-62, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16891409

ABSTRACT

Trisomy 21 or Down syndrome (DS) is the most common genetic birth defect associated with mental retardation. The over-expression of genes on chromosome 21, including SOD1 (Cu/Zn superoxide dismutase) and APP (amyloid-beta precursor protein) is believed to underlie the increased oxidative stress and neurodegeneration commonly described in DS. However, a segmental trisomy 16 mouse model for DS, Ts1Cje, has a subset of triplicated human chromosome 21 gene orthologs that exclude APP and SOD1. Here, we report that Ts1Cje brain shows decreases of mitochondrial membrane potential and ATP production, increases of reactive oxygen species, hyperphosphorylation of tau without NFT formation, increase of GSK3beta and JNK/SAPK activities and unaltered AbetaPP metabolism. Our findings suggest that genes on the trisomic Ts1Cje segment other than APP and SOD1 can cause oxidative stress, mitochondrial dysfunction and hyperphosphorylation of tau, all of which may play critical roles in the pathogenesis of mental retardation in DS.


Subject(s)
Down Syndrome/genetics , Down Syndrome/metabolism , Mitochondria/metabolism , tau Proteins/metabolism , Adenosine Triphosphate/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Down Syndrome/complications , Down Syndrome/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Humans , Intellectual Disability/etiology , Intellectual Disability/genetics , Intellectual Disability/metabolism , MAP Kinase Signaling System , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neurofibrillary Tangles/pathology , Oxidative Stress , Phosphorylation , Trisomy , tau Proteins/chemistry
15.
Trends Mol Med ; 12(10): 473-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16935027

ABSTRACT

Down syndrome (DS) has been recognized as a clinical entity for about 150 years, but it is only recently that there has been hope for the possibility to understand its pathogenesis and to use this information to devise approaches for the prevention and treatment of its numerous features. The earlier pessimism was due to several reasons, including: (i) the nature of the genetic defect that leads to the syndrome; (ii) the multiplicity of systems involved; and (iii) the high degree of variability of the phenotype. However, science has now caught up with the problem, and recent developments, especially in genetics, genomics, developmental biology and neuroscience, suggest that these potential impediments might not be as arduous as once appeared. As a result, basic research on DS is now rapidly accelerating, and there is hope that the findings will be translatable into benefit for people with DS.


Subject(s)
Down Syndrome/physiopathology , Multigene Family , Animals , Chromosomes, Human, Pair 21/genetics , Disease Models, Animal , Down Syndrome/genetics , Genetic Variation , Humans
16.
Neuron ; 51(1): 29-42, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16815330

ABSTRACT

Degeneration of basal forebrain cholinergic neurons (BFCNs) contributes to cognitive dysfunction in Alzheimer's disease (AD) and Down's syndrome (DS). We used Ts65Dn and Ts1Cje mouse models of DS to show that the increased dose of the amyloid precursor protein gene, App, acts to markedly decrease NGF retrograde transport and cause degeneration of BFCNs. NGF transport was also decreased in mice expressing wild-type human APP or a familial AD-linked mutant APP; while significant, the decreases were less marked and there was no evident degeneration of BFCNs. Because of evidence suggesting that the NGF transport defect was intra-axonal, we explored within cholinergic axons the status of early endosomes (EEs). NGF-containing EEs were enlarged in Ts65Dn mice and their App content was increased. Our study thus provides evidence for a pathogenic mechanism for DS in which increased expression of App, in the context of trisomy, causes abnormal transport of NGF and cholinergic neurodegeneration.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Cholinergic Fibers/pathology , Down Syndrome/physiopathology , Nerve Degeneration/metabolism , Nerve Growth Factor/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Axonal Transport/genetics , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/pathology , Basal Nucleus of Meynert/physiopathology , Cholinergic Fibers/metabolism , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Endosomes/genetics , Endosomes/metabolism , Endosomes/pathology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Nerve Growth Factor/genetics , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Transport/genetics , Up-Regulation/genetics
17.
Free Radic Biol Med ; 41(4): 590-600, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16863992

ABSTRACT

Atm-deficient mice, a cancer-prone model of the human disease ataxia-telangiectasia, display increased levels of oxidative stress and damage. Chronic treatment of these mice with the nitroxide antioxidant and superoxide dismutase (SOD) mimetic Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl) resulted in an increased latency to tumorigenesis. We initially hypothesized that the chemopreventative effect of Tempol was due to its SOD mimetic activity reducing cellular oxidative stress and damage. However, it is also possible that the chemopreventative effect of Tempol results from mechanisms other than directly reducing superoxide radical-induced oxidative stress and damage. To help distinguish between these possibilities, we attempted to genetically increase oxidative stress in Atm-deficient mice by either removing cytosolic Sod1 or reducing mitochondrial Sod2, or we attempted to decrease oxidative stress by treatment of Atm-deficient mice with alpha-tocopherol. Surprisingly, we found that reducing both Atm and Sod1 or Atm and Sod2 did not shorten latency to tumorigenesis or significantly affect life span. Furthermore, continuous administration of alpha-tocopherol did not affect latency to thymic lymphomas. Thus, genetically reducing Sod in Atm-deficient mice or treatment with alpha-tocopherol had no effect on survival or tumorigenesis, suggesting that the chemopreventative effect of Tempol may be at least partially independent of its effects on reducing oxidative damage and stress.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Isoenzymes/metabolism , Protein Serine-Threonine Kinases/physiology , Superoxide Dismutase/metabolism , Tumor Suppressor Proteins/physiology , alpha-Tocopherol/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Membrane Potentials , Mice , Mice, Knockout , Oxidation-Reduction , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics
19.
Free Radic Biol Med ; 40(11): 1993-2004, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16716900

ABSTRACT

We describe a novel phenotype in mice lacking the major antioxidant enzyme, CuZn-superoxide dismutase (Sod1(-/-) mice), namely a dramatic acceleration of age-related loss of skeletal muscle mass. Sod1(-/-) mice are 17 to 20% smaller and have a significantly lower muscle mass than wild-type mice as early as 3 to 4 months of age. Muscle mass in the Sod1(-/-) mice is further reduced with age and by 20 months, the hind-limb muscle mass in Sod1(-/-) mice is nearly 50% lower than in age-matched wild-type mice. Skeletal muscle tissue from young Sod1(-/-) mice has elevated oxidative damage to proteins, lipids, and DNA compared to muscle from young wild-type mice. The reduction in muscle mass and elevated oxidative damage are accompanied by a 40% decrease in voluntary wheel running by 6 months of age and decreased performance on the Rota-rod test at 13 months of age, but are not associated with a decline in overall spontaneous activity. In some of the old Sod1(-/-) mice, the loss in muscle mass is also associated with the presence of tremors and gait disturbances. Thus, the absence of CuZnSOD imposes elevated oxidative stress, loss of muscle mass, and physiological consequences that resemble an acceleration of normal age-related sarcopenia.


Subject(s)
Aging/pathology , Muscle, Skeletal/pathology , Muscular Atrophy , Oxidative Stress , Superoxide Dismutase/metabolism , Body Weight , Catalase/metabolism , Glutathione Peroxidase/metabolism , Motor Activity
20.
Hum Mol Genet ; 15(7): 1187-94, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16497723

ABSTRACT

Sod2-/- mice, which are deficient in the mitochondrial form of superoxide dismutase (MnSOD), have a short survival time that is strongly affected by genetic background. This suggests the existence of genetic modifiers that are capable of modulating the degree of mitochondrial oxidative damage caused by the MnSOD deficiency, thereby altering longevity. To identify these modifier(s), we generated recombinant congenic mice with quantitative trait loci (QTL) containing the putative genetic modifiers on the short-lived C57BL/6J genetic background. MnSOD deficient C57BL/6J mice with a QTL from the distal region of chromosome 13 from DBA/2J were able to survive for as long as those generated on the long-lived DBA/2J background. Within this region, the gene encoding nicotinamide nucleotide transhydrogenase (Nnt) was found to be defective in C57BL/6J mice, and no mature NNT protein could be detected. The forward reaction of NNT, a nuclear-encoded mitochondrial inner membrane protein, couples the generation of NADPH to proton transport and provides NADPH for the regeneration of two important antioxidant compounds, glutathione and thioredoxin, in the mitochondria. This action of NNT could explain its putative protective role in MnSOD-deficient mice.


Subject(s)
Mitochondria/metabolism , Phenotype , Superoxide Dismutase/genetics , Alleles , Animals , Cell Nucleus/metabolism , Female , Linkage Disequilibrium , Male , Mice , Mice, Congenic/genetics , Mice, Congenic/metabolism , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mitochondria/genetics , Models, Biological , Models, Genetic , Mutation , NADP Transhydrogenases/genetics , NADP Transhydrogenases/metabolism , Quantitative Trait Loci , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...