Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Transl Psychiatry ; 13(1): 387, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38092781

RESUMO

Alzheimer disease (AD) is a common neurodegenerative disease with a late onset. It is critical to identify novel blood-based DNA methylation biomarkers to better understand the extent of the molecular pathways affected in AD. Two sets of blood DNA methylation genetic prediction models developed using different reference panels and modelling strategies were leveraged to evaluate associations of genetically predicted DNA methylation levels with AD risk in 111,326 (46,828 proxy) cases and 677,663 controls. A total of 1,168 cytosine-phosphate-guanine (CpG) sites showed a significant association with AD risk at a false discovery rate (FDR) < 0.05. Methylation levels of 196 CpG sites were correlated with expression levels of 130 adjacent genes in blood. Overall, 52 CpG sites of 32 genes showed consistent association directions for the methylation-gene expression-AD risk, including nine genes (CNIH4, THUMPD3, SERPINB9, MTUS1, CISD1, FRAT2, CCDC88B, FES, and SSH2) firstly reported as AD risk genes. Nine of 32 genes were enriched in dementia and AD disease categories (P values ranged from 1.85 × 10-4 to 7.46 × 10-6), and 19 genes in a neurological disease network (score = 54) were also observed. Our findings improve the understanding of genetics and etiology for AD.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Metilação de DNA , Doença de Alzheimer/metabolismo , Epigenoma , Doenças Neurodegenerativas/genética , Biomarcadores , Ilhas de CpG , Proteínas Supressoras de Tumor/genética , Receptores Citoplasmáticos e Nucleares/genética
2.
Dev Neurosci ; 2023 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-37399790

RESUMO

The author met Verne Caviness in 1972 at Harvard Medical School, Boston, MA when he was a graduate student and Verne was a Fellow in Neurology. They came to know each other well and eventually started a long and successful collaboration. This is a story about Verne and some of our colleagues over a period of about 40 years.

4.
Endocrinology ; 163(11)2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36130190

RESUMO

Epidemiology studies demonstrate that women are at a significantly lower risk of developing type 2 diabetes (T2D) compared to men. However, the molecular basis of this risk difference is not well understood. In this study, we examined the sex differences in the genetic programs of pancreatic endocrine cells. We combined pancreas perifusion data and single-cell genomic data from our laboratory and from publicly available data sets to investigate multiple axes of the sex differences in the human pancreas at the single-cell type and single-cell level. We systematically compared female and male islet secretion function, gene expression program, and regulatory principles of pancreatic endocrine cells. The perifusion data indicate that female endocrine cells have a higher secretion capacity than male endocrine cells. Single-cell RNA-sequencing analysis suggests that endocrine cells in male controls have molecular signatures that resemble T2D. In addition, we identified genomic elements associated with genome-wide association study T2D loci to have differential accessibility between female and male delta cells. These genomic elements may play a sex-specific causal role in the pathogenesis of T2D. We provide molecular mechanisms that explain the differential risk of T2D between women and men. Knowledge gained from our study will accelerate the development of diagnostics and therapeutics in sex-aware precision medicine for diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Ilhotas Pancreáticas , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Humanos , Ilhotas Pancreáticas/metabolismo , Masculino , Pâncreas/metabolismo , RNA/metabolismo , Caracteres Sexuais
5.
Microsc Microanal ; 27(5): 1146-1153, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-35105417

RESUMO

Amyloid plaques, one of the main hallmarks of Alzheimer's disease (AD), are classified into diffuse (associated with cognitive impairment) and dense-core types (a common finding in brains of people without Alzheimer's disease (non-AD) and without impaired cognitive function) based on their morphology. We tried to determine the usability of gray-level co-occurrence matrix (GLCM) texture parameters of homogeneity and heterogeneity for the differentiation of amyloid plaque images obtained from AD and non-AD individuals. Images of amyloid-ß (Aß) immunostained brain tissue samples were obtained from the Aging, Dementia and Traumatic Brain Injury Project. A total of 1,039 plaques were isolated from different brain regions of 69 AD and non-AD individuals and used for further GLCM analysis. Images of Aß stained plaques show higher values of heterogeneity parameters and lower values of homogeneity parameters in AD patients, and vice versa in non-AD patients. Additionally, GLCM analysis shows differences in Aß plaque texture between different brain regions in non-AD patients and correlates with variables that characterize patient's dementia status. The present study shows that GLCM texture analysis is an efficient method to discriminate between different types of amyloid plaques based on their morphology and thus can prove as a valuable tool in the neuropathological investigation of dementia.


Assuntos
Doença de Alzheimer , Placa Amiloide , Envelhecimento , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Humanos , Placa Amiloide/patologia
6.
J Comp Neurol ; 527(2): 462-475, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30291623

RESUMO

Alzheimer's disease is a progressive neurodegenerative disorder and the most common form of dementia. Like many neurological disorders, Alzheimer's disease has a sex-biased epidemiological profile, affecting approximately twice as many women as men. The cause of this sex difference has yet to be elucidated. To identify molecular correlates of this sex bias, we investigated molecular pathology in females and males using the 5XFamilial Alzheimer's disease mutations (5XFAD) genetic mouse model of Alzheimer's disease. We profiled the transcriptome and proteome of the mouse hippocampus during early stages of disease development (1, 2, and 4 months of age). Our analysis reveals 42 genes that are differentially expressed between disease and wild-type animals at 2 months of age, prior to observable plaque deposition. In 4-month-old animals, we detect 1,316 differentially expressed transcripts between transgenic and control 5XFAD mice, many of which are associated with immune function. Additionally, we find that some of these transcriptional perturbations are correlated with altered protein levels in 4-month-old transgenic animals. Importantly, our data indicate that female 5XFAD mouse exhibit more profound pathology than their male counterparts as measured by differences in gene expression. We also find that the 5XFAD transgenes are more highly expressed in female 5XFAD mice than their male counterparts, which could partially account for the sex-biased molecular pathology observed in this dataset.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Caracteres Sexuais , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Proteoma , RNA Mensageiro/metabolismo , Transcriptoma
7.
Cell Rep ; 20(5): 1148-1160, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768199

RESUMO

Cells initiate fate decisions during G1 phase by converting extracellular signals into distinctive cell cycle kinetics. The DNA replication timing is determined in G1 phase; lengthened G1 and hastened S phases correlate with increased neurogenic propensity of neural progenitor cells (NPCs), although the underlying molecular control remains elusive. Here, we report that proper G1 phase completion in NPCs requires Brap, a Ras-Erk signaling modulator with ubiquitin E3 ligase activity. We identified Skp2 and Skp2-associated SCF ubiquitin ligase as a key target of Brap-mediated polyubiquitination. Loss of Brap resulted in elevated Skp2, which increased p27Kip1 destruction, leading to G1 phase truncation and premature S phase entry. The aberrantly executed G1 in Brap-mutant NPCs, followed by hindered S phase progression and increased G2 phase arrest, which together prolonged the cell cycle, impeded neuronal differentiation and culminated in microcephaly. These findings demonstrate that neuronal differentiation is potentiated during G1 phase by Brap-directed cascade of events in cell signaling and protein turnover.


Assuntos
Diferenciação Celular , Fase G1/fisiologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Fase S/fisiologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Animais , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Camundongos , Camundongos Mutantes , Células-Tronco Neurais/citologia , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Ubiquitina-Proteína Ligases/genética
8.
J Comp Neurol ; 525(15): 3360-3387, 2017 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-28685836

RESUMO

We analyzed the transcriptome of the C57BL/6J mouse hypothalamus, hippocampus, neocortex, and cerebellum to determine estrous cycle-specific changes in these four brain regions. We found almost 16,000 genes are present in one or more of the brain areas but only 210 genes, ∼1.3%, are significantly changed as a result of the estrous cycle. The hippocampus has the largest number of differentially expressed genes (DEGs) (82), followed by the neocortex (76), hypothalamus (63), and cerebellum (26). Most of these DEGs (186/210) are differentially expressed in only one of the four brain regions. A key finding is the unique expression pattern of growth hormone (Gh) and prolactin (Prl). Gh and Prl are the only DEGs to be expressed during only one stage of the estrous cycle (metestrus). To gain insight into the function of the DEGs, we examined gene ontology and phenotype enrichment and found significant enrichment for genes associated with myelination, hormone stimulus, and abnormal hormone levels. Additionally, 61 of the 210 DEGs are known to change in response to estrogen in the brain. 50 of the 210 genes differentially expressed as a result of the estrous cycle are related to myelin and oligodendrocytes and 12 of the 63 DEGs in the hypothalamus are oligodendrocyte- and myelin-specific genes. This transcriptomic analysis reveals that gene expression in the female mouse brain is remarkably stable during the estrous cycle and demonstrates that the genes that do fluctuate are functionally related.


Assuntos
Cerebelo/metabolismo , Ciclo Estral/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Neocórtex/metabolismo , Transcriptoma/fisiologia , Animais , Feminino , Expressão Gênica/fisiologia , Perfilação da Expressão Gênica , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA
9.
BMC Genomics ; 18(1): 237, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28302071

RESUMO

BACKGROUND: A variety of neurological disorders, including Alzheimer's disease, Parkinson's disease, major depressive disorder, dyslexia and autism, are differentially prevalent between females and males. To better understand the possible molecular basis for the sex-biased nature of neurological disorders, we used a developmental series of female and male mice at 1, 2, and 4 months of age to assess both mRNA and protein in the hippocampus with RNA-sequencing and mass-spectrometry, respectively. RESULTS: The transcriptomic analysis identifies 2699 genes that are differentially expressed between animals of different ages. The bulk of these differentially expressed genes are changed in both sexes at one or more ages, but a total of 198 transcripts are differentially expressed between females and males at one or more ages. The number of transcripts that are differentially expressed between females and males is greater in adult animals than in younger animals. Additionally, we identify 69 transcripts that show complex and sex-specific patterns of temporal regulation through postnatal development, 8 of which are heat-shock proteins. We also find a modest correlation between levels of mRNA and protein in the mouse hippocampus (Rho = 0.53). CONCLUSION: This study adds to the substantial body of evidence for transcriptomic regulation in the hippocampus during postnatal development. Additionally, this analysis reveals sex differences in the transcriptome of the developing mouse hippocampus, and further clarifies the need to include both female and male mice in longitudinal studies involving molecular changes in the hippocampus.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/metabolismo , Organogênese/genética , Proteoma , Proteômica , Transcriptoma , Animais , Biologia Computacional/métodos , Feminino , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Transgênicos , Proteômica/métodos , Fatores Sexuais
10.
Cell Biosci ; 6: 42, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27293547

RESUMO

BACKGROUND: The single-stranded RNA Flavivirus, Zika virus (ZIKV), has recently re-emerged and spread rapidly across the western hemisphere's equatorial countries, primarily through Aedes mosquito transmission. While symptoms in adult infections appear to be self-limiting and mild, severe birth defects, such as microcephaly, have been linked to infection during early pregnancy. Recently, Tang et al. (Cell Stem Cell 2016, doi: 10.1016/j.stem.2016.02.016) demonstrated that ZIKV efficiently infects induced pluripotent stem cell (iPSC) derived human neural progenitor cells (hNPCs), resulting in cell cycle abnormalities and apoptosis. Consequently, hNPCs are a suggested ZIKV target. METHODS: We analyzed the transcriptomic sequencing (RNA-seq) data (GEO: GSE78711) of ZIKV (Strain: MR766) infected hNPCs. For comparison to the ZIKV-infected hNPCs, the expression data from hNPCs infected with human cytomegalovirus (CMV) (Strain: AD169) was used (GEO: GSE35295). Utilizing a combination of Gene Ontology, database of human diseases, and pathway analysis, we generated a putative systemic model of infection supported by known molecular pathways of other highly related viruses. RESULTS: We analyzed RNA-sequencing data for transcript expression alterations in ZIKV-infected hNPCs, and then compared them to expression patterns of iPSC-derived hNPCs infected with CMV, a virus that can also induce severe congenital neurological defects in developing fetuses. We demonstrate for the first time that many of cellular pathways correlate with clinical pathologies following ZIKV infection such as microcephaly, congenital nervous system disorders and epilepsy. Furthermore, ZIKV activates several inflammatory signals within infected hNPCs that are implicated in innate and acquired immune responses, while CMV-infected hNPCs showed limited representation of these pathways. Moreover, several genes related to pathogen responses are significantly upregulated upon ZIKV infection, but not perturbed in CMV-infected hNPCs. CONCLUSION: The presented study is the first to report enrichment of numerous pro-inflammatory pathways in ZIKV-infected hNPCs, indicating that hNPCs are capable of signaling through canonical pro-inflammatory pathways following viral infection. By defining gene expression profiles, new factors in the pathogenesis of ZIKV were identified which could help develop new therapeutic strategies.

11.
Electrophoresis ; 37(14): 2054-62, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27129779

RESUMO

An assessment of fractionated mouse hippocampal peptides was conducted. Protein extract from a single mouse hippocampus was enzymatically digested and fractionated by IEF. Aliquots of fractions were pooled into fewer, more complex samples. The unfractionated lysate, fractions, and pooled fractions were subjected to LC-MS/MS analysis. Samples consisting of many individual fractions had more protein identifications, greater protein sequence coverage, and quantified proteins with more spectral counts than protein extract that was unfractionated or pooled into fewer LC-MS/MS samples. Additionally, prefractionation reduced the median CV for spectral counts as much as 33%. However, the relative gain in proteome resolution was found to saturate with increasing fractionation extent. This study demonstrates how prefractionation by offline IEF can improve the resolution of proteomic investigations of the mouse hippocampus, and that a data-driven pooling methodology can reduce excessive and cost-ineffective fractionation.


Assuntos
Cromatografia Líquida/métodos , Focalização Isoelétrica/métodos , Proteoma , Espectrometria de Massas em Tandem/métodos , Animais , Camundongos , Camundongos Endogâmicos C57BL
12.
J Comp Neurol ; 524(13): 2696-710, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-26917114

RESUMO

Identifying sex differences in gene expression within the brain is critical for determining why multiple neurological and behavioral disorders differentially affect males and females. Several disorders are more common or severe in males (e.g., autism and schizophrenia) or in females (e.g., Alzheimer's disease and depression). We analyzed transcriptomic data from the mouse hippocampus of six inbred strains (129S1/SvImJ, A/J, C57BL/6J, DBA/1J, DBA/2J, and PWD/Ph) to provide a perspective on differences between male and female gene expression. Our data show that 1) gene expression differences in males vs. females varies substantially across the strains, 2) only a few genes are differentially expressed across all of the strains (termed core genes), and 3) >2,600 genes differ in the individual strain comparisons (termed noncore genes). We found that DBA/2J uniquely has a substantial majority (89%) of differentially expressed genes (DEGs) that are more highly expressed in females than in males (female-biased); 129/SvImJ has a majority (69%) of DEGs that are more highly expressed in males. To gain insight into the function of the DEGs, we examined gene ontology and pathway and phenotype enrichment and found significant enrichment in phenotypes related to abnormal nervous system morphology and physiology, among others. In addition, several pathways are enriched significantly, including Alzheimer's disease (AD), with 32 genes implicated in AD, eight of which are male-biased. Three of the male-biased genes have been implicated in a neuroprotective role in AD. Our transcriptomic data provide new insight into the possible genetic bases for sex-specific susceptibility and severity of brain disorders. J. Comp. Neurol. 524:2696-2710, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Perfilação da Expressão Gênica/métodos , Hipocampo/fisiologia , Doenças do Sistema Nervoso/genética , Índice de Gravidade de Doença , Caracteres Sexuais , Animais , Feminino , Redes Reguladoras de Genes/genética , Hipocampo/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Doenças do Sistema Nervoso/patologia , Especificidade da Espécie
13.
Biomaterials ; 73: 231-42, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26410789

RESUMO

Extracellular matrices (ECM) derived from pluripotent stem cells (PSCs) provide a unique tissue microenvironment that can direct cellular differentiation and tissue regeneration, and rejuvenate aged progenitor cells. The unlimited growth capacity of PSCs allows for the scalable generation of PSC-secreted ECMs. Therefore, the derivation and characterization of PSC-derived ECMs is of critical importance in drug screening, disease modeling and tissue regeneration. In this study, 3-D ECMs were generated from decellularized undifferentiated embryonic stem cell (ESC) aggregates (AGG), spontaneously differentiated embryoid bodies (EB), and ESC-derived neural progenitor cell (NPC) aggregates. The capacities of different ECMs to direct proliferation and neural differentiation of the reseeded mouse ESCs and human induced pluripotent stem cells (iPSCs) were characterized. Proteomic analysis by liquid chromatography-tandem mass spectrometry (LC-MS/MS) revealed protein expression profiles that reflected distinct niche properties for each tested ECM group. The reseeded mouse ESCs and human iPSCs responded to different types of ECMs with different cellular phenotypes. Cells grown on the AGG-ECM displayed high levels of pluripotent markers Oct-4 and Nanog, while the cells grown on the NPC-ECM showed increased expression of neural marker ß-tubulin III. The expression levels of ß-catenin were high for cells grown on the AGG-ECM and the EB-ECM, but reduced in cells grown on the NPC-ECM, indicating a possible role of Wnt/ß-catenin signaling in the cell-matrix interactions. This study demonstrates that PSC-derived ECMs can influence stem cell fate decisions by providing a spectrum of stem cell niche microenvironments during tissue development.


Assuntos
Células-Tronco Embrionárias/citologia , Matriz Extracelular/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Nicho de Células-Tronco , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Cromatografia Líquida , Corpos Embrioides/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Camundongos , Fenótipo , Proteômica , Espectrometria de Massas em Tandem
14.
Front Neurosci ; 8: 257, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25191221

RESUMO

Neurons of the mammalian neocortex are produced by proliferating cells located in the ventricular zone (VZ) lining the lateral ventricles. This is a complex and sequential process, requiring precise control of cell cycle progression, fate commitment and differentiation. We have analyzed publicly available databases from mouse and human to identify candidate genes that are potentially involved in regulating early neocortical development and neurogenesis. We used a mouse in situ hybridization dataset (The Allen Institute for Brain Science) to identify 13 genes (Cdon, Celsr1, Dbi, E2f5, Eomes, Hmgn2, Neurog2, Notch1, Pcnt, Sox3, Ssrp1, Tead2, Tgif2) with high correlation of expression in the proliferating cells of the VZ of the neocortex at early stages of development (E15.5). We generated a similar human brain network using microarray and RNA-seq data (BrainSpan Atlas) and identified 407 genes with high expression in the developing human VZ and subventricular zone (SVZ) at 8-9 post-conception weeks. Seven of the human genes were also present in the mouse VZ network. The human and mouse networks were extended using available genetic and proteomic datasets through GeneMANIA. A gene ontology search of the mouse and human networks indicated that many of the genes are involved in the cell cycle, DNA replication, mitosis and transcriptional regulation. The reported involvement of Cdon, Celsr1, Dbi, Eomes, Neurog2, Notch1, Pcnt, Sox3, Tead2, and Tgif2 in neural development or diseases resulting from the disruption of neurogenesis validates these candidate genes. Taken together, our knowledge-based discovery method has validated the involvement of many genes already known to be involved in neocortical development and extended the potential number of genes by 100's, many of which are involved in functions related to cell proliferation but others of which are potential candidates for involvement in the regulation of neocortical development.

15.
Trends Cell Biol ; 21(8): 470-80, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21632253

RESUMO

Autosomal recessive primary microcephaly (MCPH) is characterized by small brain size as a result of deficient neuron production in the developing cerebral cortex. Although MCPH is a rare disease, the questions surrounding its etiology strike at the core of stem cell biology. The seven genes implicated in MCPH all encode centrosomal proteins and disruption of the MCPH gene Cdk5rap2 in mice revealed its role in neural progenitor proliferation and in maintaining normal centriole replication control. We discuss here the impact that centrosome regulation has upon neural progenitors in the developing brain. We integrate the impact of centriole replication defects with the functions of Cdk5rap2 and other MCPH proteins, propose mechanisms for progenitor loss in MCPH, and discuss links to two other microcephaly syndromes.


Assuntos
Centrossomo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microcefalia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Microcefalia/genética , Microcefalia/patologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Fuso Acromático/patologia
16.
Eur J Neurosci ; 32(4): 523-37, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20718853

RESUMO

Neuron production takes place continuously in the rostral migratory stream (RMS) of the adult mammalian brain. The molecular mechanisms that regulate progenitor cell division and differentiation in the RMS remain largely unknown. Here, we surveyed the mouse genome in an unbiased manner to identify candidate gene loci that regulate proliferation in the adult RMS. We quantified neurogenesis in adult C57BL/6J and A/J mice, and 27 recombinant inbred lines derived from those parental strains. We showed that the A/J RMS had greater numbers of bromodeoxyuridine-labeled cells than that of C57BL/6J mice with similar cell cycle parameters, indicating that the differences in the number of bromodeoxyuridine-positive cells reflected the number of proliferating cells between the strains. AXB and BXA recombinant inbred strains demonstrated even greater variation in the numbers of proliferating cells. Genome-wide mapping of this trait revealed that chromosome 11 harbors a significant quantitative trait locus at 116.75 +/- 0.75 Mb that affects cell proliferation in the adult RMS. The genomic regions that influence RMS proliferation did not overlap with genomic regions regulating proliferation in the adult subgranular zone of the hippocampal dentate gyrus. On the contrary, a different, suggestive locus that modulates cell proliferation in the subgranular zone was mapped to chromosome 3 at 102 +/- 7 Mb. A subset of genes in the chromosome 11 quantitative trait locus region is associated with neurogenesis and cell proliferation. Our findings provide new insights into the genetic control of neural proliferation and an excellent starting point to identify genes critical to this process.


Assuntos
Encéfalo , Movimento Celular/fisiologia , Proliferação de Células , Cromossomos de Mamíferos , Locos de Características Quantitativas , Animais , Encéfalo/citologia , Encéfalo/fisiologia , Ciclo Celular/fisiologia , Mapeamento Cromossômico , Camundongos , Camundongos Endogâmicos C57BL/anatomia & histologia , Camundongos Endogâmicos C57BL/genética , Camundongos Endogâmicos/anatomia & histologia , Camundongos Endogâmicos/genética , Dados de Sequência Molecular
17.
Trends Neurosci ; 32(8): 443-50, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19635637

RESUMO

Each of the five cellular layers of the cerebral neocortex is composed of a specific number of a single predominant 'class' of projection neuron. The projection neuron class is defined by its unique morphology and axonal projections to other areas of the brain. Precursor cell populations lining the embryonic lateral ventricles produce the projection neurons. The mechanisms regulating precursor cell proliferation also regulate total numbers of neurons produced at specific developmental periods and destined to a specific neocortical layer. Because the newborn neurons migrate relatively long distances to reach their final layer destinations, it is often assumed that the mechanisms governing acquisition of neuronal-class-specific characteristics, many of which become evident after neuron production, are independent of the mechanisms governing neuron production. We review evidence that suggests that the two mechanisms might be linked via operations of Notch1 and p27(Kip1), molecules known to regulate precursor cell proliferation and neuron production.


Assuntos
Movimento Celular/fisiologia , Neocórtex/citologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células-Tronco Embrionárias/fisiologia , Humanos , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais/fisiologia
18.
OMICS ; 13(3): 219-37, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19445647

RESUMO

In this paper we explore the concept of consensus clustering to identify, within a set of differentially expressed genes, a subset of genes that are either highly coexpressed or highly noncoexpressed based on the hypothesis that this subset would serve as a better starting point for further analyses. A number of core clustering methods form the basis for the assertion of an agreement matrix (AM) characterizing the level of coexpression between any two probesets. In order to overcome the limitations of using a single distance metric, we explore different metrics and examine the sensitivity of the AM as a function of the input number of clusters to find a suggestive number of clusters that best describes a particular dataset. The result of this level of analysis is a systematic framework for eliminating probesets that cannot be clearly characterized as either coexpressed or noncoexpressed with others, thus eliminating a number of probesets from further analysis. Subsequently, an agglomerative hierarchical clustering approach is applied to cluster the selected subset using the agreement metric information as the similarity measure. Thus, the goal of the proposed methodology is twofold: (1) we opt to identify a more "clusterable" subset of the original set; and (2) we aim at further refining the subset in order to identify a core of genes that contains genes that are either coexpressed or noncoexpressed within a certain confidence level. The approach is tested with a number of data sets, both synthetic and real, and it is demonstrated that it is successful in identifying more clusterable, also hypothesized to be more biologically relevant, subsets of genes and expression profiles.


Assuntos
Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Algoritmos , Bases de Dados Genéticas , Humanos
19.
J Comp Neurol ; 514(4): 310-28, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19326470

RESUMO

The EphA5 receptor tyrosine kinase plays key roles in axon guidance during development. However, the presence of EphA5 protein in the nervous system has not been fully characterized. To examine EphA5 localization better, mutant mice, in which the EphA5 cytoplasmic domain was replaced with beta-galactosidase, were analyzed for both temporal and regional changes in the distribution of EphA5 protein in the developing and adult nervous system. During embryonic development, high levels of EphA5 protein were found in the retina, olfactory bulb, cerebral neocortex, hippocampus, pretectum, tectum, cranial nerve nuclei, and spinal cord. Variations in intensity were observed as development proceeded. Staining of pretectal nuclei, tectal nuclei, and other areas of the mesencephalon became more diffuse after maturity, whereas the cerebral neocortex gained more robust intensity. In the adult, receptor protein continued to be detected in many areas including the olfactory nuclei, neocortex, piriform cortex, induseum griseum, hippocampus, thalamus, amygdala, hypothalamus, and septum. In addition, EphA5 protein was found in the claustrum, stria terminalis, barrel cortex, and striatal patches, and along discrete axon tracts within the corpus callosum of the adult. We conclude that EphA5 function is not limited to the developing mouse brain and may play a role in synaptic plasticity in the adult.


Assuntos
Encéfalo/metabolismo , Receptor EphA5/metabolismo , Medula Espinal/metabolismo , Animais , Western Blotting , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Galactosídeos , Hibridização In Situ , Indóis , Camundongos , Camundongos Transgênicos , Receptor EphA5/genética , Retina/embriologia , Retina/crescimento & desenvolvimento , Retina/metabolismo , Medula Espinal/embriologia , Medula Espinal/crescimento & desenvolvimento , beta-Galactosidase/genética
20.
Front Neurosci ; 3: 55, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20582282

RESUMO

Differences in gene expression in the CNS influence behavior and disease susceptibility. To systematically explore the role of normal variation in expression on hippocampal structure and function, we generated an online microarray database for a diverse panel of strains of mice, including most common inbred strains and numerous recombinant inbred lines (www.genenetwork.org). Using this resource, coexpression networks for families of genes can be generated rapidly to test causal models related to function. The data set is optimized for quantitative trait locus (QTL) mapping and was used to identify over 5500 QTLs that modulate mRNA levels. We describe a wide variety of analyses and novel synthetic approaches that take advantage of this resource, and demonstrate how both the data and associated tools can be applied to the study of gene regulation in the hippocampus and relations to structure and function.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA