Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 109(2): 328-344, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35077668

RESUMEN

Progress in earlier detection and clinical management has increased life expectancy and quality of life in people with Down syndrome (DS). However, no drug has been approved to help individuals with DS live independently and fully. Although rat models could support more robust physiological, behavioral, and toxicology analysis than mouse models during preclinical validation, no DS rat model is available as a result of technical challenges. We developed a transchromosomic rat model of DS, TcHSA21rat, which contains a freely segregating, EGFP-inserted, human chromosome 21 (HSA21) with >93% of its protein-coding genes. RNA-seq of neonatal forebrains demonstrates that TcHSA21rat expresses HSA21 genes and has an imbalance in global gene expression. Using EGFP as a marker for trisomic cells, flow cytometry analyses of peripheral blood cells from 361 adult TcHSA21rat animals show that 81% of animals retain HSA21 in >80% of cells, the criterion for a "Down syndrome karyotype" in people. TcHSA21rat exhibits learning and memory deficits and shows increased anxiety and hyperactivity. TcHSA21rat recapitulates well-characterized DS brain morphology, including smaller brain volume and reduced cerebellar size. In addition, the rat model shows reduced cerebellar foliation, which is not observed in DS mouse models. Moreover, TcHSA21rat exhibits anomalies in craniofacial morphology, heart development, husbandry, and stature. TcHSA21rat is a robust DS animal model that can facilitate DS basic research and provide a unique tool for preclinical validation to accelerate DS drug development.


Asunto(s)
Ansiedad/genética , Cromosomas Humanos Par 21 , Síndrome de Down/genética , Efecto Fundador , Hipercinesia/genética , Animales , Ansiedad/metabolismo , Ansiedad/patología , Cerebelo/metabolismo , Cerebelo/patología , Modelos Animales de Enfermedad , Síndrome de Down/metabolismo , Síndrome de Down/patología , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hipercinesia/metabolismo , Hipercinesia/patología , Cariotipo , Aprendizaje , Masculino , Mutagénesis Insercional , Tamaño de los Órganos , Postura , Prosencéfalo/metabolismo , Prosencéfalo/patología , Ratas , Ratas Transgénicas
2.
Mol Genet Genomics ; 298(1): 293-313, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36447056

RESUMEN

The aim of the present work was to explore the intriguing association of maternal folate regulator gene polymorphisms and mutations with the incidence of chromosome 21 nondisjunction and Down syndrome birth. We tested polymorphisms/mutations of DNMT3B and RFC1 genes for their association with meiotic errors in oocyte among the 1215 Down syndrome child-bearing women and 900 controls. We observed that 23 out of 31 variants of DNMT3B and RFC1 exhibited an association with meiosis II nondisjunction in maternal age-independent manner. Additionally, we have reported 17 novel mutations and 1 novel polymorphic variant that are unique to the Indian Bengali speaking cohort and increased odds in favour of meiosis II nondisjunction. We hypothesize that the risk variants and mutations of DNMT3B and RFC1 genes may cause reduction in two or more recombination events and also cause peri-centromeric single exchange that increases the risk of nondisjunction at any age of women. In silico analyses predicted the probable damages of the transcripts or proteins from the respective genes owing to the said polymorphisms. These findings from the largest population sample tested ever revealed that mutations/polymorphisms of the genes DNMT3B and RFC1 impair recombination that leads to chromosome 21 nondisjunction in the oocyte at meiosis II stage and bring us a significant step closer towards understanding the aetiology of chromosome 21 nondisjunction and birth of a child with Down syndrome to women at any age.


Asunto(s)
Síndrome de Down , Femenino , Humanos , Síndrome de Down/genética , Síndrome de Down/epidemiología , Edad Materna , Meiosis/genética , No Disyunción Genética , Oocitos , Polimorfismo Genético , ADN Metiltransferasa 3B
3.
Alzheimers Dement ; 18(8): 1498-1510, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34812584

RESUMEN

INTRODUCTION: Intellectual disability, accelerated aging, and early-onset Alzheimer-like neurodegeneration are key brain pathological features of Down syndrome (DS). Although growing research aims at the identification of molecular pathways underlying the aging trajectory of DS population, data on infants and adolescents with DS are missing. METHODS: Neuronal-derived extracellular vesicles (nEVs) were isolated form healthy donors (HDs, n = 17) and DS children (n = 18) from 2 to 17 years of age and nEV content was interrogated for markers of insulin/mTOR pathways. RESULTS: nEVs isolated from DS children were characterized by a significant increase in pIRS1Ser636 , a marker of insulin resistance, and the hyperactivation of the Akt/mTOR/p70S6K axis downstream from IRS1, likely driven by the higher inhibition of Phosphatase and tensin homolog (PTEN). High levels of pGSK3ßSer9 were also found. CONCLUSIONS: The alteration of the insulin-signaling/mTOR pathways represents an early event in DS brain and likely contributes to the cerebral dysfunction and intellectual disability observed in this unique population.


Asunto(s)
Enfermedad de Alzheimer , Síndrome de Down , Vesículas Extracelulares , Discapacidad Intelectual , Adolescente , Enfermedad de Alzheimer/patología , Niño , Síndrome de Down/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Lactante , Insulina , Serina-Treonina Quinasas TOR/metabolismo
4.
Alzheimers Dement (Amst) ; 16(2): e12580, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38623383

RESUMEN

Down syndrome (DS) is caused by a third copy of chromosome 21. Alzheimer's disease (AD) is a neurodegenerative condition characterized by the deposition of amyloid-beta (Aß) plaques and neurofibrillary tangles in the brain. Both disorders have elevated Aß, tau, dysregulated immune response, and inflammation. In people with DS, Hsa21 genes like APP and DYRK1A are overexpressed, causing an accumulation of amyloid and neurofibrillary tangles, and potentially contributing to an increased risk of AD. As a result, people with DS are a key demographic for research into AD therapeutics and prevention. The molecular links between DS and AD shed insights into the underlying causes of both diseases and highlight potential therapeutic targets. Also, using biomarkers for early diagnosis and treatment monitoring is an active area of research, and genetic screening for high-risk individuals may enable earlier intervention. Finally, the fundamental mechanistic parallels between DS and AD emphasize the necessity for continued research into effective treatments and prevention measures for DS patients at risk for AD. Genetic screening with customized therapy approaches may help the DS population in current clinical studies and future biomarkers.

5.
Neurobiol Dis ; 58: 92-101, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23643842

RESUMEN

In addition to intellectual disability, individuals with Down syndrome (DS) exhibit dementia by the third or fourth decade of life, due to the early onset of neuropathological changes typical of Alzheimer's disease (AD). Deficient ontogenetic neurogenesis contributes to the brain hypoplasia and hypocellularity evident in fetuses and children with DS. A murine model of DS and AD (the Ts65Dn mouse) exhibits key features of these disorders, notably deficient ontogenetic neurogenesis, degeneration of basal forebrain cholinergic neurons (BFCNs), and cognitive deficits. Adult hippocampal (HP) neurogenesis is also deficient in Ts65Dn mice and may contribute to the observed cognitive dysfunction. Herein, we demonstrate that supplementing the maternal diet with additional choline (approximately 4.5 times the amount in normal rodent chow) dramatically improved the performance of the adult trisomic offspring in a radial arm water maze task. Ts65Dn offspring of choline-supplemented dams performed significantly better than unsupplemented Ts65Dn mice. Furthermore, adult hippocampal neurogenesis was partially normalized in the maternal choline supplemented (MCS) trisomic offspring relative to their unsupplemented counterparts. A significant correlation was observed between adult hippocampal neurogenesis and performance in the water maze, suggesting that the increased neurogenesis seen in the supplemented trisomic mice contributed functionally to their improved spatial cognition. These findings suggest that supplementing the maternal diet with additional choline has significant translational potential for DS.


Asunto(s)
Colina/administración & dosificación , Síndrome de Down/patología , Hipocampo/patología , Discapacidades para el Aprendizaje/prevención & control , Neurogénesis/genética , Fenómenos Fisiologicos de la Nutrición Prenatal/efectos de los fármacos , Percepción Espacial/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Peso Corporal/genética , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Síndrome de Down/complicaciones , Síndrome de Down/genética , Femenino , Discapacidades para el Aprendizaje/etiología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neurogénesis/fisiología , Neuropéptidos/metabolismo , Embarazo/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal
6.
Mol Genet Metab ; 110(3): 371-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23920041

RESUMEN

BACKGROUND AND AIMS: Down syndrome is caused by trisomy of all or part of human chromosome 21. Individuals with Down syndrome present some metabolic abnormalities involving lipoproteins, notably lower high-density lipoprotein levels associated with altered lecithin:cholesterol acyltransferase activity and apolipoprotein A-I levels. DYRK1A is a kinase overexpressed in Down syndrome that can activate the STAT3 pathway, which is involved in lecithin:cholesterol acyltransferase expression. Therefore, we characterized the role of DYRK1A overexpression on lecithin:cholesterol acyltransferase activity and expression in mouse models. METHODS: Effects of Dyrk1a overexpression were examined in mice overexpressing Dyrk1a by ELISA, chemical analyses and Western blotting. RESULTS: Overexpression of DYRK1A decreased plasma lecithin:cholesterol acyltransferase activity and hepatic STAT3 activation, which was associated with activation of SHP2, a tyrosine phosphatase. Although hepatic apolipoprotein E and D levels were increased in mice overexpressing DYRK1A, decreased plasma lecithin:cholesterol acyltransferase activity was associated with decreased hepatic and plasma apolipoprotein A-I levels. High-density lipoprotein-cholesterol levels were also decreased in plasma despite similar total cholesterol and non-high-density lipoprotein-cholesterol levels. CONCLUSIONS: We identified the role of DYRK1A overexpression on altered lipoprotein metabolism.


Asunto(s)
Apolipoproteína A-I/sangre , Expresión Génica , Fosfatidilcolina-Esterol O-Aciltransferasa/sangre , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Animales , Apolipoproteínas D/metabolismo , Apolipoproteínas E/metabolismo , HDL-Colesterol/sangre , Activación Enzimática , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Quinasas DyrK
7.
Aging (Albany NY) ; 15(23): 14086-14108, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38095646

RESUMEN

Trisomy 21, or Down syndrome (DS), is the most frequent human autosomal chromosome aneuploidy, which leads to multiple developmental disorders, especially mental retardation in individuals. The presence of an additional human chromosome 21 (HSA21) could account for the pathological manifestations in DS. In this study, we analyzed the mRNA gene expression profile of DS-derived amniocytes compared with normal amniocytes, aiming to evaluate the relationship between candidate dysregulated HSA21 genes and DS developmental phenotypes. Differentially expressed genes (DEGs) included 1794 upregulated genes and 1411 downregulated genes, which are mainly involved in cell adhesion, inflammation, cell proliferation and thus may play an important role in inducing multiple dysplasia during DS fetal development. Furthermore, STRING protein network studies demonstrated 7 candidate HSA21 genes participated Gene Ontology (GO) terms: cell adhesion and extracellular matrix remodeling (COL6A1, COL6A2, COL18A1, ADAMTS5, JAM2, and POFUT2), inflammation and virus infection response (MX1 and MX2), histone modification and chromatin remodeling (NRIP1), glycerolipid and glycerophospholipid metabolism (AGPAT3), mitochondrial function (ATP5PF and ATP5PO), synaptic vesicle endocytosis (ITSN1 and SYNJ1) and amyloid metabolism (APP). Meanwhile, GSEA enrichment identified several transcription factors and miRNAs, which may target gene expression in the DS group. Our study established connections between dysregulated genes, especially HSA21 genes, and DS-associated phenotypes. The alteration of multiple pathways and biological processes may contribute to DS developmental disorders, providing potential pathogenesis and therapeutic targets for DS.


Asunto(s)
Síndrome de Down , MicroARNs , Humanos , Síndrome de Down/metabolismo , Transcriptoma , MicroARNs/metabolismo , Factores de Transcripción/genética , Inflamación
8.
Biol Psychiatry ; 94(1): 84-97, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074246

RESUMEN

BACKGROUND: Despite successful preclinical treatment studies to improve neurocognition in the Ts65Dn mouse model of Down syndrome, translation to humans has failed. This raises questions about the appropriateness of the Ts65Dn mouse as the gold standard. We used the novel Ts66Yah mouse that carries an extra chromosome and the identical segmental Mmu16 trisomy as Ts65Dn without the Mmu17 non-Hsa21 orthologous region. METHODS: Forebrains from embryonic day 18.5 Ts66Yah and Ts65Dn mice, along with euploid littermate controls, were used for gene expression and pathway analyses. Behavioral experiments were performed in neonatal and adult mice. Because male Ts66Yah mice are fertile, parent-of-origin transmission of the extra chromosome was studied. RESULTS: Forty-five protein-coding genes mapped to the Ts65Dn Mmu17 non-Hsa21 orthologous region; 71%-82% are expressed during forebrain development. Several of these genes are uniquely overexpressed in Ts65Dn embryonic forebrain, producing major differences in dysregulated genes and pathways. Despite these differences, the primary Mmu16 trisomic effects were highly conserved in both models, resulting in commonly dysregulated disomic genes and pathways. Delays in motor development, communication, and olfactory spatial memory were present in Ts66Yah but more pronounced in Ts65Dn neonates. Adult Ts66Yah mice showed milder working memory deficits and sex-specific effects in exploratory behavior and spatial hippocampal memory, while long-term memory was preserved. CONCLUSIONS: Our findings suggest that triplication of the non-Hsa21 orthologous Mmu17 genes significantly contributes to the phenotype of the Ts65Dn mouse and may explain why preclinical trials that used this model have unsuccessfully translated to human therapies.


Asunto(s)
Síndrome de Down , Femenino , Ratones , Masculino , Humanos , Animales , Síndrome de Down/genética , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/metabolismo , Trisomía/genética , Hipocampo/metabolismo , Modelos Animales de Enfermedad
9.
Stem Cell Res ; 72: 103204, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37734318

RESUMEN

Down syndrome (DS) is caused by trisomy of Homo sapiens chromosome 21 (HSA21) and is by far the most common chromosomal disorder accompanied by neurodevelopmental disorders and congenital heart disease. Here, we generated two induced pluripotent stem cell (iPSC) lines from two patients with DS. These two lines exhibited normal morphology, trisomy 21 karyotype, pluripotency and differentiation capability into derivatives of three germ layers. The patient-specific iPSC lines arean invaluable resource in research to model DS-related cellular and molecular pathologies and test possible therapeutic strategies for DS.


Asunto(s)
Síndrome de Down , Células Madre Pluripotentes Inducidas , Humanos , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/patología , Células Madre Pluripotentes Inducidas/metabolismo , Trisomía/patología , Diferenciación Celular/genética
10.
Elife ; 92020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32597754

RESUMEN

Animal models of Down syndrome (DS), trisomic for human chromosome 21 (HSA21) genes or orthologs, provide insights into better understanding and treatment options. The only existing transchromosomic (Tc) mouse DS model, Tc1, carries a HSA21 with over 50 protein coding genes (PCGs) disrupted. Tc1 is mosaic, compromising interpretation of results. Here, we "clone" the 34 MB long arm of HSA21 (HSA21q) as a mouse artificial chromosome (MAC). Through multiple steps of microcell-mediated chromosome transfer, we created a new Tc DS mouse model, Tc(HSA21q;MAC)1Yakaz ("TcMAC21"). TcMAC21 is not mosaic and contains 93% of HSA21q PCGs that are expressed and regulatable. TcMAC21 recapitulates many DS phenotypes including anomalies in heart, craniofacial skeleton and brain, molecular/cellular pathologies, and impairments in learning, memory and synaptic plasticity. TcMAC21 is the most complete genetic mouse model of DS extant and has potential for supporting a wide range of basic and preclinical research.


Asunto(s)
Cromosomas Humanos Par 21/genética , Síndrome de Down/genética , Ratones Transgénicos/genética , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Cardiopatías Congénitas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Trisomía/genética , Secuenciación Completa del Genoma
11.
Fertil Steril ; 111(5): 991-1001.e2, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30922649

RESUMEN

OBJECTIVE: To analyze how chromosome 21 (HSA21) ploidy affects global gene expression of early human blastocysts. DESIGN: Prospective study. SETTING: University-affiliated in vitro fertilization clinic. PATIENT(S): A total of 26 high-quality donated embryos from in vitro fertilization (IVF) patients: trisomy 21 (n = 8), monosomy 21 (n = 10), and euploid (n = 8) blastocysts. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Blastocyst transcriptome changes and its associated functions. RESULT(S): Trisomy 21, monosomy 21, and euploid blastocysts were classified by comparative genomic hybridization. The global transcriptome of whole blastocysts was analyzed with small cell number RNA sequencing, and they were compared to understand the gene expression behavior at early development and its implications for embryo implantation. We identified 1,232 differentially expressed genes (false discovery rate <0.05) in monosomy 21 compared with euploid blastocysts associated with dysregulated functions in embryo development as the Rap1 signaling pathway. Curiously, Down syndrome in early development revealed fewer transcriptomic changes than expected. In addition, Down syndrome gene expression in neonates, children, and adults revealed that the number of deregulated genes increases across life stages from blastocysts to adults, suggesting a potential dosage-compensation mechanism for human chromosome 21. CONCLUSION(S): At the transcriptomic level, early development in Down syndrome is mainly dosage compensated. However, monosomy 21 is strongly transcriptionally affected because early development involving main functions is associated with embryo implantation.


Asunto(s)
Aneuploidia , Técnicas de Cultivo de Embriones/métodos , Desarrollo Embrionario/genética , Estudios de Asociación Genética/métodos , Monosomía/genética , Transcriptoma/genética , Adulto , Cromosomas Humanos Par 21/genética , Femenino , Humanos , Embarazo , Estudios Prospectivos , Técnicas Reproductivas Asistidas
12.
G3 (Bethesda) ; 8(7): 2215-2223, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29760202

RESUMEN

Trisomy for human chromosome 21 (Hsa21) results in Down syndrome (DS), one of the most genetically complex conditions compatible with human survival. Assessment of the physiological consequences of dosage-driven overexpression of individual Hsa21 genes during early embryogenesis and the resulting contributions to DS pathology in mammals are not tractable in a systematic way. A recent study looked at loss-of-function of a subset of Caenorhabditis elegans orthologs of Hsa21 genes and identified ten candidates with behavioral phenotypes, but the equivalent over-expression experiment has not been done. We turned to zebrafish as a developmental model and, using a number of surrogate phenotypes, we screened Hsa21 genes for effects on early embyrogenesis. We prepared a library of 164 cDNAs of conserved protein coding genes, injected mRNA into early embryos and evaluated up to 5 days post-fertilization (dpf). Twenty-four genes produced a gross morphological phenotype, 11 of which could be reproduced reliably. Seven of these gave a phenotype consistent with down regulation of the sonic hedgehog (Shh) pathway; two showed defects indicative of defective neural crest migration; one resulted consistently in pericardial edema; and one was embryonic lethal. Combinatorial injections of multiple Hsa21 genes revealed both additive and compensatory effects, supporting the notion that complex genetic relationships underlie end phenotypes of trisomy that produce DS. Together, our data suggest that this system is useful in the genetic dissection of dosage-sensitive gene effects on early development and can inform the contribution of both individual loci and their combinatorial effects to phenotypes relevant to the etiopathology of DS.


Asunto(s)
Cromosomas Humanos Par 21 , Regulación del Desarrollo de la Expresión Génica , Pez Cebra/embriología , Pez Cebra/genética , Animales , Modelos Animales de Enfermedad , Síndrome de Down/genética , Dosificación de Gen , Biblioteca de Genes , Estudios de Asociación Genética , Prueba de Complementación Genética , Humanos , Fenotipo
13.
Stem Cell Reports ; 11(1): 32-42, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29861166

RESUMEN

Early-onset Alzheimer disease (AD)-like pathology in Down syndrome is commonly attributed to an increased dosage of the amyloid precursor protein (APP) gene. To test this in an isogenic human model, we deleted the supernumerary copy of the APP gene in trisomic Down syndrome induced pluripotent stem cells or upregulated APP expression in euploid human pluripotent stem cells using CRISPRa. Cortical neuronal differentiation shows that an increased APP gene dosage is responsible for increased ß-amyloid production, altered Aß42/40 ratio, and deposition of the pyroglutamate (E3)-containing amyloid aggregates, but not for several tau-related AD phenotypes or increased apoptosis. Transcriptome comparisons demonstrate that APP has a widespread and temporally modulated impact on neuronal gene expression. Collectively, these data reveal an important role for APP in the amyloidogenic aspects of AD but challenge the idea that increased APP levels are solely responsible for increasing specific phosphorylated forms of tau or enhanced neuronal cell death in Down syndrome-associated AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/etiología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Síndrome de Down/etiología , Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Diferenciación Celular , Células Cultivadas , Susceptibilidad a Enfermedades , Síndrome de Down/metabolismo , Síndrome de Down/patología , Dosificación de Gen , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Agregado de Proteínas , Agregación Patológica de Proteínas , Transcriptoma
14.
Drug Des Devel Ther ; 9: 103-25, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25552901

RESUMEN

Down syndrome (DS), also known as trisomy 21, is the most common genetic cause of intellectual disability (ID). Although ID can be mild, the average intelligence quotient is in the range of 40-50. All individuals with DS will also develop the neuropathology of Alzheimer's disease (AD) by the age of 30-40 years, and approximately half will display an AD-like dementia by the age of 60 years. DS is caused by an extra copy of the long arm of human chromosome 21 (Hsa21) and the consequent elevated levels of expression, due to dosage, of trisomic genes. Despite a worldwide incidence of one in 700-1,000 live births, there are currently no pharmacological treatments available for ID or AD in DS. However, over the last several years, very promising results have been obtained with a mouse model of DS, the Ts65Dn. A diverse array of drugs has been shown to rescue, or partially rescue, DS-relevant deficits in learning and memory and abnormalities in cellular and electrophysiological features seen in the Ts65Dn. These results suggest that some level of amelioration or prevention of cognitive deficits in people with DS may be possible. Here, we review information from the preclinical evaluations in the Ts65Dn, how drugs were selected, how efficacy was judged, and how outcomes differ, or not, among studies. We also summarize the current state of human clinical trials for ID and AD in DS. Lastly, we describe the genetic limitations of the Ts65Dn as a model of DS, and in the preclinical testing of pharmacotherapeutics, and suggest additional targets to be considered for potential pharmacotherapies.


Asunto(s)
Trastornos del Conocimiento/tratamiento farmacológico , Síndrome de Down/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Trastornos del Conocimiento/genética , Síndrome de Down/genética , Humanos
15.
Gene ; 530(2): 278-86, 2013 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-23933415

RESUMEN

Given the important roles of miRNAs in post-transcriptional regulation and its implications for the development of immune tissues and cells, characterization of miRNAs promotes us to uncover the molecular mechanisms underlying the pathway of trisomic chromosome 21 that disrupts the disomic genes expression and immunological defects related to Down syndrome (DS). In the present study, we analyzed global changes and chromosome distribution characteristics of miRNAs expression in lymphocytes from children with trisomy 21 by means of the Illumina high-throughput sequencing technology. Two small libraries were constructed using pool RNA of normal and DS children. The results have been further validated by stem-loop quantitative RT-PCR. Comparison between DS and normal profiles revealed that most of identified miRNAs were expressed at similar levels. The chromosome 21 that contributes to the abundantly expressed miRNAs was small, and not all Hsa21-derived miRNAs were over-expressed with ratios significantly ≥ 1.5 in Down syndrome children lymphocytes. Based on the deep sequencing technology, 108 novel candidate miRNAs have been identified, and 2 of them were derived from human chromosome 21. For the 114 significantly differentially expressed miRNAs, function annotation of target genes indicated that a set of highly abundantly and significantly differentially expressed miRNAs were involved in hematopoietic or lymphoid organ development, thymus development, and T/B cell differentiation and activation. Our results indicated that these abnormally expressed miRNAs might be associated with the mechanisms that trisomy 21 results in dysregulation of disomic genes and involved in the immunological defects seen in DS.


Asunto(s)
Linfocitos B/metabolismo , Cromosomas Humanos Par 21/genética , Síndrome de Down/genética , MicroARNs/genética , Linfocitos T/metabolismo , Linfocitos B/inmunología , Linfocitos B/patología , Secuencia de Bases , Estudios de Casos y Controles , Diferenciación Celular , Niño , Preescolar , Mapeo Cromosómico , Cromosomas Humanos Par 21/inmunología , Síndrome de Down/inmunología , Síndrome de Down/patología , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunidad Innata/genética , MicroARNs/inmunología , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Linfocitos T/inmunología , Linfocitos T/patología
16.
Gene ; 532(1): 1-12, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-23246696

RESUMEN

MicroRNAs (miRNAs), a family of small nonprotein-coding RNAs, play a critical role in posttranscriptional gene regulation by acting as adaptors for the miRNA-induced silencing complex to inhibit gene expression by targeting mRNAs for translational repression and/or cleavage. miR-155-5p and miR-155-3p are processed from the B-cell Integration Cluster (BIC) gene (now designated, MIR155 host gene or MIR155HG). MiR-155-5p is highly expressed in both activated B- and T-cells and in monocytes/macrophages. MiR-155-5p is one of the best characterized miRNAs and recent data indicate that miR-155-5p plays a critical role in various physiological and pathological processes such as hematopoietic lineage differentiation, immunity, inflammation, viral infections, cancer, cardiovascular disease, and Down syndrome. In this review we summarize the mechanisms by which MIR155HG expression can be regulated. Given that the pathologies mediated by miR-155-5p result from the over-expression of this miRNA it may be possible to therapeutically attenuate miR-155-5p levels in the treatment of several pathological processes.


Asunto(s)
Enfermedades Cardiovasculares/genética , Regulación de la Expresión Génica , Inflamación/genética , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/genética , Animales , Linfocitos B/fisiología , Diferenciación Celular/genética , Síndrome de Down/genética , Femenino , Humanos , Macrófagos/fisiología , Familia de Multigenes , FN-kappa B/genética , FN-kappa B/metabolismo , Linfocitos T/fisiología , Factor de Transcripción AP-1/metabolismo
17.
Braz. j. biol ; 68(2): 447-452, May 2008. tab
Artículo en Inglés | LILACS | ID: lil-486775

RESUMEN

Even though the molecular mechanisms underlying the Down syndrome (DS) phenotypes remain obscure, the characterization of the genes and conserved non-genic sequences of HSA21 together with large-scale gene expression studies in DS tissues are enhancing our understanding of this complex disorder. Also, mouse models of DS provide invaluable tools to correlate genes or chromosome segments to specific phenotypes. Here we discuss the possible contribution of HSA21 genes to DS and data from global gene expression studies of trisomic samples.


Embora os mecanismos moleculares que causam a síndrome de Down (SD) não sejam totalmente conhecidos, a caracterização de genes e seqüências não gênicas conservadas do HSA21 e os estudos de expressão em grande escala em amostras de pacientes com SD estão aumentando o entendimento da síndrome. Por outro lado, os modelos murinos da SD provêm ferramentas valiosas para correlacionar genes ou segmentos cromossômicos a características fenotípicas específicas. Nesta revisão, são discutidas as possíveis contribuições dos genes do HSA21 à SD e os dados de estudos de expressão gênica global de amostras trissômicas.


Asunto(s)
Animales , Humanos , Ratones , /genética , Síndrome de Down/genética , Perfilación de la Expresión Génica , Modelos Animales de Enfermedad , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA