Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Transl Psychiatry ; 13(1): 387, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38092781

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Humans , DNA Methylation , Alzheimer Disease/metabolism , Epigenome , Neurodegenerative Diseases/genetics , Biomarkers , CpG Islands , Tumor Suppressor Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics
2.
Dev Neurosci ; 2023 Jul 03.
Article in English | MEDLINE | ID: mdl-37399790

ABSTRACT

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.

3.
Algorithmica ; 85(4): 1067-1090, 2023.
Article in English | MEDLINE | ID: mdl-37008075

ABSTRACT

We consider two variants of orthogonal colouring games on graphs. In these games, two players alternate colouring uncoloured vertices (from a choice of m ∈ N colours) of a pair of isomorphic graphs while respecting the properness and the orthogonality of the partial colourings. In the normal play variant, the first player unable to move loses. In the scoring variant, each player aims to maximise their score, which is the number of coloured vertices in their copy of the graph. We prove that, given an instance with partial colourings, both the normal play and the scoring variant of the game are PSPACE-complete. An involution σ of a graph G is strictly matched if its fixed point set induces a clique and v σ ( v ) ∈ E ( G ) for any non-fixed point v ∈ V ( G ) . Andres et al. (Theor Comput Sci 795:312-325, 2019) gave a solution of the normal play variant played on graphs that admit a strictly matched involution. We prove that recognising graphs that admit a strictly matched involution is NP-complete.

5.
Endocrinology ; 163(11)2022 10 11.
Article in English | MEDLINE | ID: mdl-36130190

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Diabetes Mellitus, Type 2/metabolism , Female , Genome-Wide Association Study , Humans , Islets of Langerhans/metabolism , Male , Pancreas/metabolism , RNA/metabolism , Sex Characteristics
6.
Microsc Microanal ; 27(5): 1146-1153, 2021 10.
Article in English | MEDLINE | ID: mdl-35105417

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Plaque, Amyloid , Aging , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/diagnostic imaging , Brain/pathology , Humans , Plaque, Amyloid/pathology
7.
J Comp Neurol ; 527(2): 462-475, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30291623

ABSTRACT

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.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Hippocampus/metabolism , Hippocampus/pathology , Sex Characteristics , Aging/metabolism , Aging/pathology , Animals , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Proteome , RNA, Messenger/metabolism , Transcriptome
8.
Curr Biol ; 27(22): 3454-3467.e8, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29103939

ABSTRACT

Robustness in biology is the stability of phenotype under diverse genetic and/or environmental perturbations. The circadian clock has remarkable stability of period and phase that-unlike other biological oscillators-is maintained over a wide range of conditions. Here, we show that the high fidelity of the circadian system stems from robust degradation of the clock protein PERIOD. We show that PERIOD degradation is regulated by a balance between ubiquitination and deubiquitination, and that disruption of this balance can destabilize the clock. In mice with a loss-of-function mutation of the E3 ligase gene ß-Trcp2, the balance of PERIOD degradation is perturbed and the clock becomes dramatically unstable, presenting a unique behavioral phenotype unlike other circadian mutant animal models. We believe that our data provide a molecular explanation for how circadian phases, such as wake-sleep onset times, can become unstable in humans, and we present a unique mouse model to study human circadian disorders with unstable circadian rhythm phases.


Subject(s)
Circadian Rhythm/physiology , Period Circadian Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , CLOCK Proteins/genetics , Circadian Clocks , Circadian Rhythm/genetics , Mice , Models, Animal , Period Circadian Proteins/physiology , Proteolysis , Sleep/genetics , Sleep Disorders, Circadian Rhythm/genetics , Sleep Disorders, Circadian Rhythm/physiopathology , Ubiquitin-Protein Ligases/genetics , Ubiquitination , beta-Transducin Repeat-Containing Proteins/genetics
9.
Cell Rep ; 20(5): 1148-1160, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28768199

ABSTRACT

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.


Subject(s)
Cell Differentiation , G1 Phase/physiology , Neural Stem Cells/metabolism , Neurons/metabolism , S Phase/physiology , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Mice , Mice, Mutant Strains , Neural Stem Cells/cytology , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitin-Protein Ligases/genetics
10.
J Comp Neurol ; 525(15): 3360-3387, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28685836

ABSTRACT

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.


Subject(s)
Cerebellum/metabolism , Estrous Cycle/metabolism , Hippocampus/metabolism , Hypothalamus/metabolism , Neocortex/metabolism , Transcriptome/physiology , Animals , Female , Gene Expression/physiology , Gene Expression Profiling , Mice, Inbred C57BL , Sequence Analysis, RNA
11.
BMC Genomics ; 18(1): 237, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28302071

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Hippocampus/metabolism , Organogenesis/genetics , Proteome , Proteomics , Transcriptome , Animals , Computational Biology/methods , Female , Hippocampus/growth & development , Male , Mice , Mice, Transgenic , Proteomics/methods , Sex Factors
12.
Cell Biosci ; 6: 42, 2016.
Article in English | MEDLINE | ID: mdl-27293547

ABSTRACT

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.

13.
Electrophoresis ; 37(14): 2054-62, 2016 07.
Article in English | MEDLINE | ID: mdl-27129779

ABSTRACT

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.


Subject(s)
Chromatography, Liquid/methods , Isoelectric Focusing/methods , Proteome , Tandem Mass Spectrometry/methods , Animals , Mice , Mice, Inbred C57BL
14.
J Comp Neurol ; 524(13): 2696-710, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26917114

ABSTRACT

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.


Subject(s)
Gene Expression Profiling/methods , Hippocampus/physiology , Nervous System Diseases/genetics , Severity of Illness Index , Sex Characteristics , Animals , Female , Gene Regulatory Networks/genetics , Hippocampus/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Nervous System Diseases/pathology , Species Specificity
15.
G3 (Bethesda) ; 6(1): 221-33, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26596646

ABSTRACT

The developmental transition to motherhood requires gene expression changes that alter the brain to drive the female to perform maternal behaviors. We broadly examined the global transcriptional response in the mouse maternal brain, by examining four brain regions: hypothalamus, hippocampus, neocortex, and cerebellum, in virgin females, two pregnancy time points, and three postpartum time points. We find that overall there are hundreds of differentially expressed genes, but each brain region and time point shows a unique molecular signature, with only 49 genes differentially expressed in all four regions. Interestingly, a set of "early-response genes" is repressed in all brain regions during pregnancy and postpartum stages. Several genes previously implicated in underlying postpartum depression change expression. This study serves as an atlas of gene expression changes in the maternal brain, with the results demonstrating that pregnancy, parturition, and postpartum maternal experience substantially impact diverse brain regions.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Postpartum Period , Animals , Behavior, Animal , Cluster Analysis , Computational Biology/methods , Depression, Postpartum/genetics , Female , Gene Expression Profiling , Gene Ontology , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Phenotype , Pregnancy , Transcriptome
16.
Biomaterials ; 73: 231-42, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410789

ABSTRACT

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.


Subject(s)
Embryonic Stem Cells/cytology , Extracellular Matrix/metabolism , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Stem Cell Niche , Animals , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Cell Proliferation , Chromatography, Liquid , Embryoid Bodies/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Mice , Phenotype , Proteomics , Tandem Mass Spectrometry
17.
Front Neurosci ; 8: 257, 2014.
Article in English | MEDLINE | ID: mdl-25191221

ABSTRACT

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.

18.
Eur J Neurosci ; 35(12): 1879-86, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22708599

ABSTRACT

Therapeutic strategies are often based on two general principles: interference with the pathogenic process and repair of the damaged tissues. Recent studies, however, have suggested that several pathological conditions may result from the interplay between genetic susceptibility traits and environmental influences that, by modulating the epigenome, also affect disease onset and progression. Based on lessons from neural development, it is conceivable that new lines of preventive and possibly therapeutic intervention might be developed to modulate disease onset or decrease the severity of the symptoms. This review will discuss these concepts within the context of multiple sclerosis, the most common demyelinating disease of the central nervous system, and the leading cause of progressive neurological disability in young adults.


Subject(s)
Multiple Sclerosis/therapy , Adult , Animals , Disease Models, Animal , Early Medical Intervention , Gene-Environment Interaction , Genetic Predisposition to Disease/genetics , Humans , Mice , Molecular Targeted Therapy , Multiple Sclerosis/diagnosis , Multiple Sclerosis/genetics , Multiple Sclerosis/prevention & control , Precision Medicine , Young Adult
19.
Trends Cell Biol ; 21(8): 470-80, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21632253

ABSTRACT

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.


Subject(s)
Centrosome/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microcephaly/metabolism , Nerve Tissue Proteins/metabolism , Animals , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Microcephaly/genetics , Microcephaly/pathology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Spindle Apparatus/pathology
20.
Eur J Neurosci ; 32(4): 523-37, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20718853

ABSTRACT

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.


Subject(s)
Brain , Cell Movement/physiology , Cell Proliferation , Chromosomes, Mammalian , Quantitative Trait Loci , Animals , Brain/cytology , Brain/physiology , Cell Cycle/physiology , Chromosome Mapping , Mice , Mice, Inbred C57BL/anatomy & histology , Mice, Inbred C57BL/genetics , Mice, Inbred Strains/anatomy & histology , Mice, Inbred Strains/genetics , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...