3801
|
Genomic-anatomic evidence for distinct functional domains in hippocampal field CA1. Proc Natl Acad Sci U S A 2009; 106:11794-9. [PMID: 19561297 DOI: 10.1073/pnas.0812608106] [Citation(s) in RCA: 274] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Functional heterogeneity has been investigated for decades in the hippocampal region of the mammalian cerebral cortex, and evidence for vaguely defined "dorsal" and "ventral" regions is emerging. Direct evidence that hippocampal field CA1 displays clear regional, laminar, and pyramidal neuron differentiation is presented here, based on a systematic high-resolution analysis of a publicly accessible, genome-wide expression digital library (Allen Brain Atlas) [Lein et al. (2007) Genome-wide atlas of gene expression in the adult mouse brain. Nature 445:168-176]. First, genetic markers reveal distinct spatial expression domains and subdomains along the longitudinal (dorsal/septal/posterior to ventral/temporal/anterior) axis of field CA1. Second, genetic markers divide field CA1 pyramidal neurons into multiple subtypes with characteristic laminar distributions. And third, subcortical brain regions receiving axonal projections from molecularly distinct spatial domains of field CA1 display distinct global gene expression patterns, suggesting that field CA1 spatial domains may be genetically wired independently to form distinct functional networks related to cognition and emotion. Insights emerging from this genomic-anatomic approach provide a starting point for a detailed analysis of differential hippocampal structure-function organization.
Collapse
|
3802
|
Wu Q, Boyle MP, Palmiter RD. Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation. Cell 2009; 137:1225-34. [PMID: 19563755 PMCID: PMC2729323 DOI: 10.1016/j.cell.2009.04.022] [Citation(s) in RCA: 355] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 02/09/2009] [Accepted: 04/07/2009] [Indexed: 10/20/2022]
Abstract
Neurons in the arcuate nucleus that produce AgRP, NPY, and GABA (AgRP neurons) promote feeding. Ablation of AgRP neurons in adult mice results in Fos activation in postsynaptic neurons and starvation. Loss of GABA is implicated in starvation because chronic subcutaneous delivery of bretazenil (a GABA(A) receptor partial agonist) suppresses Fos activation and maintains feeding during ablation of AgRP neurons. Moreover, under these conditions, direct delivery of bretazenil into the parabrachial nucleus (PBN), a direct target of AgRP neurons that also relays gustatory and visceral sensory information, is sufficient to maintain feeding. Conversely, inactivation of GABA biosynthesis in the ARC or blockade of GABA(A) receptors in the PBN of mice promote anorexia. We suggest that activation of the PBN by AgRP neuron ablation or gastrointestinal malaise inhibits feeding. Chronic delivery of bretazenil during loss of AgRP neurons provides time to establish compensatory mechanisms that eventually allow mice to eat.
Collapse
Affiliation(s)
- Qi Wu
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | | |
Collapse
|
3803
|
The Treasury of the Commons: Making Use of Public Gene Expression Resources to Better Characterize the Molecular Diversity of Inhibitory Interneurons in the Cerebellar Cortex. THE CEREBELLUM 2009; 8:477-89. [DOI: 10.1007/s12311-009-0124-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 06/09/2009] [Indexed: 11/25/2022]
|
3804
|
Clough RL, Dermentzaki G, Stefanis L. Functional dissection of the alpha-synuclein promoter: transcriptional regulation by ZSCAN21 and ZNF219. J Neurochem 2009; 110:1479-90. [PMID: 19549071 DOI: 10.1111/j.1471-4159.2009.06250.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alpha-synuclein (SNCA) is an abundant neuronal protein involved in synaptic neurotransmission. SNCA expression levels have been strongly implicated in Parkinson's disease pathogenesis. We have previously demonstrated that in the PC12 cell line elements in intron 1 may mediate SNCA transcriptional regulation in response to neurotrophins. We have now identified transcription factor (TF) binding sites in intron 1 and the 5'-promoter of SNCA. A binding site for the TF zinc finger and SCAN domain containing (ZSCAN)21 in the 5'-region of intron 1 is required for intron 1 transcriptional activity. Small interfering RNA against ZSCAN21 inhibits activation in the luciferase assay and diminishes SNCA protein levels in naïve and neurotrophin-treated PC12 cells and in primary cultured cortical neurons, demonstrating that ZSCAN21 is a novel transcriptional regulator of SNCA in neuronal cells. The 5'-promoter of SNCA has a complex architecture, including multiple binding sites for the TF zinc finger protein (ZNF)219, which functions as both an activator and a repressor. Targeting ZSCAN21 or other TFs controlling SNCA transcriptional activity may provide novel therapeutic avenues not only for Parkinson's disease but also for other synucleopathies.
Collapse
Affiliation(s)
- Richard Lee Clough
- Division of Basic Neuroscience, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece.
| | | | | |
Collapse
|
3805
|
Chikina MD, Huttenhower C, Murphy CT, Troyanskaya OG. Global prediction of tissue-specific gene expression and context-dependent gene networks in Caenorhabditis elegans. PLoS Comput Biol 2009; 5:e1000417. [PMID: 19543383 PMCID: PMC2692103 DOI: 10.1371/journal.pcbi.1000417] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 05/14/2009] [Indexed: 01/09/2023] Open
Abstract
Tissue-specific gene expression plays a fundamental role in metazoan biology and is an important aspect of many complex diseases. Nevertheless, an organism-wide map of tissue-specific expression remains elusive due to difficulty in obtaining these data experimentally. Here, we leveraged existing whole-animal Caenorhabditis elegans microarray data representing diverse conditions and developmental stages to generate accurate predictions of tissue-specific gene expression and experimentally validated these predictions. These patterns of tissue-specific expression are more accurate than existing high-throughput experimental studies for nearly all tissues; they also complement existing experiments by addressing tissue-specific expression present at particular developmental stages and in small tissues. We used these predictions to address several experimentally challenging questions, including the identification of tissue-specific transcriptional motifs and the discovery of potential miRNA regulation specific to particular tissues. We also investigate the role of tissue context in gene function through tissue-specific functional interaction networks. To our knowledge, this is the first study producing high-accuracy predictions of tissue-specific expression and interactions for a metazoan organism based on whole-animal data. In animals, a crucial facet of any gene's function is the tissue or cell type in which that gene is expressed and the proteins that it interacts with in that cell. However, genome-wide identification of expression across the multitude of tissues of varying size and complexity is difficult to achieve experimentally. In this paper, we show that microararray data collected from whole animals can be analyzed to yield high-quality predictions of tissue-specific expression. These predictions are of better or comparable accuracy to tissue-specific expression determined from high-throughput experiments. Our results provide a global view of tissue-specific expression in Caenorhabditis elegans, allowing us to address the question of how expression patterns are regulated and to analyze how the functions of genes that are expressed in several tissues are influenced by the cellular context.
Collapse
Affiliation(s)
- Maria D Chikina
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | | | | | | |
Collapse
|
3806
|
Marowsky A, Burgener J, Falck JR, Fritschy JM, Arand M. Distribution of soluble and microsomal epoxide hydrolase in the mouse brain and its contribution to cerebral epoxyeicosatrienoic acid metabolism. Neuroscience 2009; 163:646-61. [PMID: 19540314 DOI: 10.1016/j.neuroscience.2009.06.033] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 06/05/2009] [Accepted: 06/07/2009] [Indexed: 12/17/2022]
Abstract
Epoxide hydrolases comprise a family of enzymes important in detoxification and conversion of lipid signaling molecules, namely epoxyeicosatrienoic acids (EETs), to their supposedly less active form, dihydroxyeicosatrienoic acids (DHETs). EETs control cerebral blood flow, exert analgesic, anti-inflammatory and angiogenic effects and protect against ischemia. Although the role of soluble epoxide hydrolase (sEH) in EET metabolism is well established, knowledge on its detailed distribution in rodent brain is rather limited. Here, we analyzed the expression pattern of sEH and of another important member of the EH family, microsomal epoxide hydrolase (mEH), in mouse brain by immunohistochemistry. To investigate the functional relevance of these enzymes in brain, we explored their individual contribution to EET metabolism in acutely isolated brain cells from respective EH -/- mice and wild type littermates by mass spectrometry. We find sEH immunoreactivity almost exclusively in astrocytes throughout the brain, except in the central amygdala, where neurons are also positive for sEH. mEH immunoreactivity is abundant in brain vascular cells (endothelial and smooth muscle cells) and in choroid plexus epithelial cells. In addition, mEH immunoreactivity is present in specific neuronal populations of the hippocampus, striatum, amygdala, and cerebellum, as well as in a fraction of astrocytes. In freshly isolated cells from hippocampus, where both enzymes are expressed, sEH mediates the bulk of EET metabolism. Yet we observe a significant contribution of mEH, pointing to a novel role of this enzyme in the regulation of physiological processes. Furthermore, our findings indicate the presence of additional, hitherto unknown cerebral epoxide hydrolases. Taken together, cerebral EET metabolism is driven by several epoxide hydrolases, a fact important in view of the present targeting of sEH as a potential therapeutic target. Our findings suggest that these different enzymes have individual, possibly quite distinct roles in brain function and cerebral EET metabolism.
Collapse
Affiliation(s)
- A Marowsky
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstr. 190, CH-8057 Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
3807
|
Chakrabarti O, Hegde RS. Functional depletion of mahogunin by cytosolically exposed prion protein contributes to neurodegeneration. Cell 2009; 137:1136-47. [PMID: 19524515 PMCID: PMC2709807 DOI: 10.1016/j.cell.2009.03.042] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 01/13/2009] [Accepted: 03/20/2009] [Indexed: 01/14/2023]
Abstract
The pathways leading from aberrant Prion protein (PrP) metabolism to neurodegeneration are poorly understood. Some familial PrP mutants generate increased (Ctm)PrP, a transmembrane isoform associated with disease. In other disease situations, a potentially toxic cytosolic form (termed cyPrP) might be produced. However, the mechanisms by which (Ctm)PrP or cyPrP cause selective neuronal dysfunction are unknown. Here, we show that both (Ctm)PrP and cyPrP can interact with and disrupt the function of Mahogunin (Mgrn), a cytosolic ubiquitin ligase whose loss causes spongiform neurodegeneration. Cultured cells and transgenic mice expressing either (Ctm)PrP-producing mutants or cyPrP partially phenocopy Mgrn depletion, displaying aberrant lysosomal morphology and loss of Mgrn in selected brain regions. These effects were rescued by either Mgrn overexpression, competition for PrP-binding sites, or prevention of cytosolic PrP exposure. Thus, transient or partial exposure of PrP to the cytosol leads to inappropriate Mgrn sequestration that contributes to neuronal dysfunction and disease.
Collapse
Affiliation(s)
- Oishee Chakrabarti
- Cell Biology and Metabolism Program, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ramanujan S. Hegde
- Cell Biology and Metabolism Program, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
3808
|
Shi Y, Lu W, Milstein AD, Nicoll RA. The stoichiometry of AMPA receptors and TARPs varies by neuronal cell type. Neuron 2009; 62:633-40. [PMID: 19524523 PMCID: PMC3119531 DOI: 10.1016/j.neuron.2009.05.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Revised: 04/13/2009] [Accepted: 05/15/2009] [Indexed: 11/25/2022]
Abstract
Synaptic AMPA receptors (AMPARs) are regulated by a family of auxiliary subunits known as transmembrane AMPA receptor regulatory proteins (TARPs). TARPs control the trafficking and gating of AMPARs. However, the number of TARP molecules that assemble within individual AMPAR channels is unknown. Here, we covalently link AMPARs to TARPs to investigate the properties of TARP/AMPAR complexes with known stoichiometry in HEK cells. We find that AMPARs are functional when associated with four, two, or no TARPs, and that the efficacy of the partial agonist kainate varies across these conditions, providing a sensitive assay for TARP/AMPAR stoichiometry. A comparison of these results with data obtained from hippocampal neurons demonstrates that native AMPARs associate with TARPs with a variable stoichiometry that depends on TARP expression level. Interestingly, AMPARs in hippocampal pyramidal neurons are saturated by TARP expression, while those in dentate gyrus granule neurons are not, indicating that variable TARP/AMPAR stoichiometry provides a mechanism for cell-type-specific regulation of AMPAR function.
Collapse
Affiliation(s)
- Yun Shi
- Departments of Cellular and Molecular Pharmacology and Physiology, University of California San Francisco, California 94143, USA
| | - Wei Lu
- Departments of Cellular and Molecular Pharmacology and Physiology, University of California San Francisco, California 94143, USA
| | - Aaron D. Milstein
- Departments of Cellular and Molecular Pharmacology and Physiology, University of California San Francisco, California 94143, USA
| | - Roger A. Nicoll
- Departments of Cellular and Molecular Pharmacology and Physiology, University of California San Francisco, California 94143, USA
| |
Collapse
|
3809
|
Giocomo LM, Hasselmo ME. Knock-out of HCN1 subunit flattens dorsal-ventral frequency gradient of medial entorhinal neurons in adult mice. J Neurosci 2009; 29:7625-30. [PMID: 19515931 PMCID: PMC2729850 DOI: 10.1523/jneurosci.0609-09.2009] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 04/15/2009] [Accepted: 05/05/2009] [Indexed: 11/21/2022] Open
Abstract
Layer II stellate cells at different locations along the dorsal to ventral axis of medial entorhinal cortex show differences in the frequency of intrinsic membrane potential oscillations and resonance (Giocomo et al., 2007). The frequency differences scale with differences in the size and spacing of grid-cell firing fields recorded in layer II of the medial entorhinal cortex in behaving animals. To determine the mechanism for this difference in intrinsic frequency, we analyzed oscillatory properties in adult control mice and adult mice with a global deletion of the HCN1 channel. Data from whole-cell patch recordings show that the oscillation frequency gradient along the dorsal-ventral axis previously shown in juvenile rats also appears in control adult mice, indicating that the dorsal-ventral gradient generalizes across age and species. Knock-out of the HCN1 channel flattens the dorsal-ventral gradient of the membrane potential oscillation frequency, the resonant frequency, the time constant of the "sag" potential and the amplitude of the sag potential. This supports a role of the HCN1 subunit in the mechanism of the frequency gradient in these neurons. These findings have important implications for models of grid cells and generate predictions for future in vivo work on entorhinal grid cells.
Collapse
Affiliation(s)
- Lisa M Giocomo
- Center for Memory and Brain, Boston University, Boston, Massachusetts 02215, USA.
| | | |
Collapse
|
3810
|
Cooper MA, Crockett DP, Nowakowski RS, Gale NW, Zhou R. Distribution of EphA5 receptor protein in the developing and adult mouse nervous system. J Comp Neurol 2009; 514:310-28. [PMID: 19326470 PMCID: PMC2724768 DOI: 10.1002/cne.22030] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
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.
Collapse
Affiliation(s)
- Margaret A. Cooper
- Laboratory for Cancer Research, College of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - David P. Crockett
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey, 08854
| | - Richard S. Nowakowski
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey, 08854
| | | | - Renping Zhou
- Laboratory for Cancer Research, College of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey, 08854
| |
Collapse
|
3811
|
Lara FA, Kahn SA, da Fonseca AC, Bahia CP, Pinho JP, Graca-Souza AV, Houzel JC, de Oliveira PL, Moura-Neto V, Oliveira MF. On the fate of extracellular hemoglobin and heme in brain. J Cereb Blood Flow Metab 2009; 29:1109-20. [PMID: 19337276 DOI: 10.1038/jcbfm.2009.34] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major cause of disability in adults worldwide. The pathophysiology of this syndrome is complex, involving both inflammatory and redox components triggered by the extravasation of blood into the cerebral parenchyma. Hemoglobin, heme, and iron released therein seem be important in the brain damage observed in ICH. However, there is a lack of information concerning hemoglobin traffic and metabolism in brain cells. Here, we investigated the fate of hemoglobin and heme in cultured neurons and astrocytes, as well as in the cortex of adult rats. Hemoglobin was made traceable by conjugation to Alexa 488, whereas a fluorescent heme analogue (tin-protoporphyrin IX) was prepared to allow heme tracking. Using fluorescence microscopy we observed that neurons were more efficient in uptake hemoglobin and heme than astrocytes. Exposure of cortical neurons to hemoglobin or heme resulted in an oxidative stress condition. Viability assays showed that neurons were more susceptible to both hemoglobin and heme toxicity than astrocytes. Together, these results show that neurons, rather than astrocytes, preferentially take up hemoglobin-derived products, indicating that these cells are actively involved in the ICH-associated brain damage.
Collapse
Affiliation(s)
- Flavio A Lara
- Laboratório de Morfogênese Celular, Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3812
|
Gibrat C, Saint-Pierre M, Bousquet M, Lévesque D, Rouillard C, Cicchetti F. Differences between subacute and chronic MPTP mice models: investigation of dopaminergic neuronal degeneration and α-synuclein inclusions. J Neurochem 2009; 109:1469-82. [DOI: 10.1111/j.1471-4159.2009.06072.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
3813
|
Park CC, Petyuk VA, Qian WJ, Smith RD, Smith DJ. Dual spatial maps of transcript and protein abundance in the mouse brain. Expert Rev Proteomics 2009; 6:243-9. [PMID: 19489697 PMCID: PMC2744401 DOI: 10.1586/epr.09.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Integrating quantitative proteomic and transcriptomic datasets promises valuable insights in unraveling the molecular mechanisms of the brain. We concentrate on recent studies using mass spectrometry and microarray data to investigate transcript and protein abundance in normal and diseased neural tissues. Highlighted are dual spatial maps of these molecules obtained using voxelation of the mouse brain. We demonstrate that the relationship between transcript and protein levels displays a specific anatomical distribution, with greatest fidelity in midline structures and the hypothalamus. Genes are also identified that have strong correlations between mRNA and protein abundance. In addition, transcriptomic and proteomic analysis of mouse models of Parkinson's disease are discussed.
Collapse
Affiliation(s)
- Christopher C Park
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
3814
|
Arimatsu Y, Nihonmatsu I, Hatanaka Y. Localization of latexin-immunoreactive neurons in the adult cat cerebral cortex and claustrum/endopiriform formation. Neuroscience 2009; 162:1398-410. [PMID: 19486926 DOI: 10.1016/j.neuroscience.2009.05.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 05/22/2009] [Accepted: 05/22/2009] [Indexed: 01/12/2023]
Abstract
The distribution of neurons that are immunoreactive to latexin, which is an endogenous inhibitor of the A/B subfamily of metallocarboxypeptidases, was investigated in the adult cat telencephalon. Latexin-immunoreactive neurons were distributed in the lower layers of the neocortex and adjacent ventral mesocortex, as well as in the claustrum/endopiriform formation. There were marked regional and laminar differences in density and distribution of latexin-immunoreactive neurons in the cerebral cortex. The density followed a roughly lateral-to-medial decreasing gradient: it was high in lateral cortical regions, which included the insular, second somatosensory, and anterior sylvian areas, and in the temporal auditory field; moderate in laterodorsal cortical regions, which included the primary and second auditory fields; and low in dorsal cortical regions, which included visual areas 18 and 19. Latexin-immunoreactive neurons were absent in medial cortical regions, which included the motor, premotor, prefrontal, prelimbic, cingulate, and retrosplenial areas. The lateral-to-medial gradient was apparent even within a single cytoarchitectonic area in certain cortical regions. The allocortex was devoid of latexin-immunoreactive neurons, with the exception of the anteroventral part of the dentate gyrus. The majority of cortical latexin-immunoreactive neurons were localized in layers V and VI and appeared to correspond to the "modified pyramidal cells in the infragranular layers." The remaining latexin-immunoreactive neurons were localized in layer IV, as well as in lower layer III and in the white matter. There were no latexin-immunoreactive neurons from layer I through upper layer III. Latexin-immunoreactive neurons were present in telencephalic structures outside the cerebral cortex, with particularly high density in the claustrum/endopiriform formation. All these features, with the exception of that detected in the archicortex, are compatible with the features observed previously in the rat telencephalon. The similar pattern of distribution of latexin-immunoreactive neurons in several mammalian species from different orders suggests that latexin plays an important role in a specific cortical network.
Collapse
Affiliation(s)
- Y Arimatsu
- Mitsubishi Kagaku Institute of Life Sciences, 11 Minamiooya, Machida-shi, Tokyo 194-8511, Japan.
| | | | | |
Collapse
|
3815
|
Attardo A, Fabel K, Krebs J, Haubensak W, Huttner WB, Kempermann G. Tis21 expression marks not only populations of neurogenic precursor cells but also new postmitotic neurons in adult hippocampal neurogenesis. ACTA ACUST UNITED AC 2009; 20:304-14. [PMID: 19482889 PMCID: PMC2803732 DOI: 10.1093/cercor/bhp100] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During embryonic cortical development, expression of Tis21 is associated with cell cycle lengthening and neurogenic divisions of progenitor cells. We here investigated if the expression pattern of Tis21 also correlates with the generation of new neurons in the adult hippocampus. We used Tis21 knock-in mice expressing green fluorescent protein (GFP) and studied Tis21-GFP expression together with markers of adult hippocampal neurogenesis in newly generated cells. We found that Tis21-GFP 1) was absent from the radial glia–like putative stem cells (type-1 cells), 2) first appeared in transient amplifying progenitor cells (type-2 and 3 cells), 3) did not colocalize with markers of early postmitotic maturation stage, 4) was expressed again in maturing neurons, and 5) finally decreased in mature granule cells. Our data show that, in the course of adult neurogenesis, Tis21 is expressed in a phase additional to the one of the embryonic neurogenesis. This additional phase of expression might be associated with a new and different function of Tis21 than during embryonic brain development, where no Tis21 is expressed in mature neurons. We hypothesize that this function is related to the final functional integration of the newborn neurons. Tis21 can thus serve as new marker for key stages of adult neurogenesis.
Collapse
Affiliation(s)
- Alessio Attardo
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
3816
|
Gene-network analysis identifies susceptibility genes related to glycobiology in autism. PLoS One 2009; 4:e5324. [PMID: 19492091 PMCID: PMC2683930 DOI: 10.1371/journal.pone.0005324] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Accepted: 03/20/2009] [Indexed: 12/08/2022] Open
Abstract
The recent identification of copy-number variation in the human genome has opened up new avenues for the discovery of positional candidate genes underlying complex genetic disorders, especially in the field of psychiatric disease. One major challenge that remains is pinpointing the susceptibility genes in the multitude of disease-associated loci. This challenge may be tackled by reconstruction of functional gene-networks from the genes residing in these loci. We applied this approach to autism spectrum disorder (ASD), and identified the copy-number changes in the DNA of 105 ASD patients and 267 healthy individuals with Illumina Humanhap300 Beadchips. Subsequently, we used a human reconstructed gene-network, Prioritizer, to rank candidate genes in the segmental gains and losses in our autism cohort. This analysis highlighted several candidate genes already known to be mutated in cognitive and neuropsychiatric disorders, including RAI1, BRD1, and LARGE. In addition, the LARGE gene was part of a sub-network of seven genes functioning in glycobiology, present in seven copy-number changes specifically identified in autism patients with limited co-morbidity. Three of these seven copy-number changes were de novo in the patients. In autism patients with a complex phenotype and healthy controls no such sub-network was identified. An independent systematic analysis of 13 published autism susceptibility loci supports the involvement of genes related to glycobiology as we also identified the same or similar genes from those loci. Our findings suggest that the occurrence of genomic gains and losses of genes associated with glycobiology are important contributors to the development of ASD.
Collapse
|
3817
|
A novel microdeletion syndrome involving 5q14.3-q15: clinical and molecular cytogenetic characterization of three patients. Eur J Hum Genet 2009; 17:1592-9. [PMID: 19471318 DOI: 10.1038/ejhg.2009.90] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Molecular karyotyping is being increasingly applied to delineate novel disease causing microaberrations and related syndromes in patients with mental retardation of unknown aetiology. We report on three unrelated patients with overlapping de novo interstitial microdeletions involving 5q14.3-q15. All three patients presented with severe psychomotor retardation, epilepsy or febrile seizures, muscular hypotonia and variable brain and minor anomalies. Molecular karyotyping revealed three overlapping microdeletions measuring 5.7, 3.9 and 3.6 Mb, respectively. The microdeletions were identified using single nucleotide polymorphism (SNP) arrays (Affymetrix 100K and Illumina 550K) and array comparative genomic hybridization (1 Mb Sanger array-CGH). Confirmation and segregation studies were performed using fluorescence in situ hybridization (FISH) and quantitative PCR. All three aberrations were confirmed and proven to have occurred de novo. The boundaries and sizes of the deletions in the three patients were different, but an overlapping region of around 1.6 Mb in 5q14.3 was defined. It included five genes: CETN3, AC093510.2, POLR3G, LYSMD3 and the proximal part of GPR98/MASS1, a known epilepsy gene. Haploinsufficiency of GPR98/MASS1 is probably responsible for the seizure phenotype in our patients. At least one other gene contained in the commonly deleted region, LYSMD3, shows a high level of central nervous expression during embryogenesis and is also, therefore, a good candidate gene for other central nervous system (CNS) symptoms, such as psychomotor retardation, brain anomalies and muscular hypotonia of the 5q14.3 microdeletion syndrome.
Collapse
|
3818
|
Torgersen JS, Takle H, Andersen O. Localization of mRNAs and proteins in methyl methacrylate-embedded tissues. J Histochem Cytochem 2009; 57:825-30. [PMID: 19435715 DOI: 10.1369/jhc.2009.953695] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Precise localization of proteins and mRNA in histological sections is necessary for evaluating spatial gene expression patterns. Here we report sensitive detection of the gene products in fish tissues by immunohistochemistry (IHC) and in situ hybridization (ISH) assays on sections of whole specimens and vertebra embedded in methyl methacrylate (MMA) resin. This plastic resin favors easy preparation of various specimen types and enables preparation of large sections with well-preserved cell morphology. IHC analysis of the muscle regulatory factor MyoD in transverse sections of juvenile cod revealed MyoD-positive cells in the dorsolateral parts of the adaxial muscle. ISH revealed less spatially restricted signals of the bone morphogenic protein bmp4 in muscle and brain. To assess the applicability of ISH on sections of bony tissue, col1a1 and col2a1 expression was investigated in non-decalcified vertebra sections of Atlantic salmon. The former was identified in both chondrocytes and osteoblasts, whereas the latter was mostly evident in chondrocytes. We conclude that MMA resin offers easy preparation of large and problematic tissues and the possibility of carrying out both IHC and ISH analyses using standard protocols.
Collapse
|
3819
|
Davis FP, Eddy SR. A tool for identification of genes expressed in patterns of interest using the Allen Brain Atlas. ACTA ACUST UNITED AC 2009; 25:1647-54. [PMID: 19414530 DOI: 10.1093/bioinformatics/btp288] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
MOTIVATION Gene expression patterns can be useful in understanding the structural organization of the brain and the regulatory logic that governs its myriad cell types. A particularly rich source of spatial expression data is the Allen Brain Atlas (ABA), a comprehensive genome-wide in situ hybridization study of the adult mouse brain. Here, we present an open-source program, ALLENMINER, that searches the ABA for genes that are expressed, enriched, patterned or graded in a user-specified region of interest. RESULTS Regionally enriched genes identified by ALLENMINER accurately reflect the in situ data (95-99% concordance with manual curation) and compare with regional microarray studies as expected from previous comparisons (61-80% concordance). We demonstrate the utility of ALLENMINER by identifying genes that exhibit patterned expression in the caudoputamen and neocortex. We discuss general characteristics of gene expression in the mouse brain and the potential application of ALLENMINER to design strategies for specific genetic access to brain regions and cell types. AVAILABILITY ALLENMINER is freely available on the Internet at http://research.janelia.org/davis/allenminer.
Collapse
Affiliation(s)
- Fred P Davis
- HHMI Janelia Farm Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA.
| | | |
Collapse
|
3820
|
Simmons DM, Swanson LW. Comparing histological data from different brains: sources of error and strategies for minimizing them. BRAIN RESEARCH REVIEWS 2009; 60:349-67. [PMID: 19248810 PMCID: PMC2680454 DOI: 10.1016/j.brainresrev.2009.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 02/12/2009] [Accepted: 02/13/2009] [Indexed: 10/21/2022]
Abstract
The recent development of brain atlases with computer graphics templates, and of huge databases of neurohistochemical data on the internet, has forced a systematic re-examination of errors associated with comparing histological features between adjacent sections of the same brain, between brains treated in the same way, and between brains from groups treated in different ways. The long-term goal is to compare as accurately as possible a broad array of data from experimental brains within the framework of reference atlases. Main sources of error, each of which ideally should be measured and minimized, include intrinsic biological variation, linear and nonlinear distortion of histological sections, plane of section differences between each brain, section alignment problems, and sampling errors. These variables are discussed, along with approaches to error estimation and minimization in terms of a specific example-the distribution of neuroendocrine neurons in the rat paraventricular nucleus. Based on the strategy developed here, the main conclusion is that the best long-term solution is a high-resolution 3D computer graphics model of the brain that can be sliced in any plane and used as the framework for quantitative neuroanatomy, databases, knowledge management systems, and structure-function modeling. However, any approach to the automatic annotation of neuroanatomical data-relating its spatial distribution to a reference atlas-should deal systematically with these sources of error, which reduce localization reliability.
Collapse
Affiliation(s)
- Donna M. Simmons
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | - Larry W. Swanson
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
3821
|
Larson SD, Martone ME. Ontologies for Neuroscience: What are they and What are they Good for? Front Neurosci 2009; 3:60-7. [PMID: 19753098 PMCID: PMC2695392 DOI: 10.3389/neuro.01.007.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 03/22/2009] [Indexed: 11/13/2022] Open
Abstract
Current information technology practices in neuroscience make it difficult to understand the organization of the brain across spatial scales. Subcellular junctional connectivity, cytoarchitectural local connectivity, and long-range topographical connectivity are just a few of the relevant data domains that must be synthesized in order to make sense of the brain. However, due to the heterogeneity of the data produced within these domains, the landscape of multiscale neuroscience data is fragmented. A standard framework for neuroscience data is needed to bridge existing digital data resources and to help in the conceptual unification of the multiple disciplines of neuroscience. Using our efforts in building ontologies for neuroscience as an example, we examine the benefits and limits of ontologies as a solution for this data integration problem. We provide several examples of their application to problems of image annotation, content-based retrieval of structural data, and integration of data across scales and researchers.
Collapse
Affiliation(s)
- Stephen D Larson
- Department of Neurosciences, University of California San Diego, CA, USA
| | | |
Collapse
|
3822
|
Badea A, Johnson GA, Williams RW. Genetic dissection of the mouse brain using high-field magnetic resonance microscopy. Neuroimage 2009; 45:1067-79. [PMID: 19349225 PMCID: PMC2667384 DOI: 10.1016/j.neuroimage.2009.01.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Revised: 12/05/2008] [Accepted: 01/12/2009] [Indexed: 10/21/2022] Open
Abstract
Magnetic resonance (MR) imaging has demonstrated that variation in brain structure is associated with differences in behavior and disease state. However, it has rarely been practical to prospectively test causal models that link anatomical and functional differences in humans. In the present study we have combined classical mouse genetics with high-field MR to systematically explore and test such structure-functional relations across multiple brain regions. We segmented 33 regions in two parental strains-C57BL/6J (B) and DBA/2J (D)-and in nine BXD recombinant inbred strains. All strains have been studied extensively for more than 20 years using a battery of genetic, functional, anatomical, and behavioral assays. We compared levels of variation within and between strains and sexes, by region, and by system. Average within-strain variation had a coefficient of variation (CV) of 1.6% for the whole brain; while the CV ranged from 2.3 to 3.6% for olfactory bulbs, cortex and cerebellum, and up to approximately 18% for septum and laterodorsal thalamic nucleus. Variation among strain averages ranged from 6.7% for cerebellum, 7.6% for whole brain, 9.0% for cortex, up to approximately 26% for the ventricles, laterodorsal thalamic nucleus, and the interpeduncular nucleus. Heritabilities averaged 0.60+/-0.18. Sex differences were not significant with the possible (and unexpected) exception of the pons ( approximately 20% larger in males). A correlation matrix of regional volumes revealed high correlations among functionally related parts of the CNS (e.g., components of the limbic system), and several high correlations between regions that are not anatomically connected, but that may nonetheless be functionally or genetically coupled.
Collapse
Affiliation(s)
- A Badea
- Center for In Vivo Microscopy, Box 3302 Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
3823
|
Wexler EM, Paucer A, Kornblum HI, Palmer TD, Plamer TD, Geschwind DH. Endogenous Wnt signaling maintains neural progenitor cell potency. Stem Cells 2009; 27:1130-41. [PMID: 19418460 PMCID: PMC2782960 DOI: 10.1002/stem.36] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Wnt signaling regulates neural stem cell (NSC) function and development throughout an individual's lifetime. Intriguingly, adult hippocampal progenitors (AHPs) produce several Wnts, and the intracellular machinery necessary to respond to them, creating the potential for an active autocrine-signaling loop within this stem cell niche. However, the standard luciferase-based Wnt assay failed to detect this signaling loop. This assay is inherently less temporally sensitive to activity among a population of unsynchronized proliferating cells because it relies on the rapidly degrading reporter luciferase. We circumvented this limitation using a promoter assay that employs green fluorescent protein (GFP), as a relatively long-lived reporter of canonical Wnt activity. We found that at baseline, AHPs secreted functional Wnt that self-stimulates low-level canonical Wnt signaling. Elimination baseline Wnt activity, via application of an extracellular Wnt antagonist promoted neurogenesis, based on a combination of unbiased gene expression analysis and cell-fate analysis. A detailed clonal analysis of progenitors transduced with specific intracellular antagonists of canonical signaling, either Axin or truncated cadherin (beta-catenin sequestering), revealed that loss of baseline signaling depletes the population of multipotent precursors, thereby driving an increasing fraction to assume a committed cell fate (i.e., unipotent progenitors). Similarly, baseline Wnt signaling repressed differentiation of human NSCs. Although the specific Wnts produced by neural precursors vary with age and between species, their effects remain remarkably consistent. In sum, this study establishes that autonomous Wnt signaling is a conserved feature of the neurogenic niche that preserves the delicate balance between NSC maintenance and differentiation.
Collapse
Affiliation(s)
- Eric M Wexler
- Department of Psychiatry, The Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA 90024-1759, USA.
| | | | | | | | | | | |
Collapse
|
3824
|
Fu Y, Tvrdik P, Makki N, Palombi O, Machold R, Paxinos G, Watson C. The precerebellar linear nucleus in the mouse defined by connections, immunohistochemistry, and gene expression. Brain Res 2009; 1271:49-59. [DOI: 10.1016/j.brainres.2009.02.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 02/24/2009] [Accepted: 02/25/2009] [Indexed: 01/29/2023]
|
3825
|
Dahlin A, Royall J, Hohmann JG, Wang J. Expression profiling of the solute carrier gene family in the mouse brain. J Pharmacol Exp Ther 2009; 329:558-70. [PMID: 19179540 PMCID: PMC2672879 DOI: 10.1124/jpet.108.149831] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2008] [Accepted: 01/28/2009] [Indexed: 01/19/2023] Open
Abstract
The solute carrier (Slc) superfamily is a major group of membrane transport proteins present in mammalian cells. Although Slc transporters play essential and diverse roles in the central nervous system, the localization and function of the vast majority of Slc genes in the mammalian brain are largely unknown. Using high-throughput in situ hybridization data generated by the Allen Brain Atlas, we systematically and quantitatively analyzed the spatial and cellular distribution of 307 Slc genes, which represent nearly 90% of presently known mouse Slc genes, in the adult C57BL/6J mouse brain. Our analysis showed that 252 (82%) of the 307 Slc genes are present in the brain, and a large proportion of these genes were detected at low to moderate expression levels. Evaluation of 20 anatomical brain subdivisions demonstrated a comparable level of Slc gene complexity but significant difference in transcript enrichment. The distribution of the expressed Slc genes was diverse, ranging from near-ubiquitous to highly localized. Functional annotation in 20 brain regions, including the blood-brain and blood-cerebral spinal fluid (CSF) barriers, suggests major roles of Slc transporters in supporting brain energy utilization, neurotransmission, nutrient supply, and CSF production. Furthermore, hierarchical cluster analysis revealed intricate Slc expression patterns associated with neuroanatomical organization. Our studies also revealed Slc genes present within defined brain microstructures and described the putative cell types expressing individual Slc genes. These results provide a useful resource for investigators to explore the roles of Slc genes in neurophysiological and pathological processes.
Collapse
Affiliation(s)
- Amber Dahlin
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
3826
|
Vidal RL, Valenzuela JI, Luján R, Couve A. Cellular and subcellular localization of Marlin-1 in the brain. BMC Neurosci 2009; 10:37. [PMID: 19386132 PMCID: PMC2685396 DOI: 10.1186/1471-2202-10-37] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Accepted: 04/22/2009] [Indexed: 02/07/2023] Open
Abstract
Background Marlin-1 is a microtubule binding protein that associates specifically with the GABAB1 subunit in neurons and with members of the Janus kinase family in lymphoid cells. In addition, it binds the molecular motor kinesin-I and nucleic acids, preferentially single stranded RNA. Marlin-1 is expressed mainly in the central nervous system but little is known regarding its cellular and subcellular distribution in the brain. Results Here we have studied the localization of Marlin-1 in the rodent brain and cultured neurons combining immunohistochemistry, immunofluorescence and pre-embedding electron microscopy. We demonstrate that Marlin-1 is enriched in restricted areas of the brain including olfactory bulb, cerebral cortex, hippocampus and cerebellum. Marlin-1 is abundant in dendrites and axons of GABAergic and non-GABAergic hippocampal neurons. At the ultrastructural level, Marlin-1 is present in the cytoplasm and the nucleus of CA1 neurons in the hippocampus. In the cytoplasm it associates to microtubules in the dendritic shaft and occasionally with the Golgi apparatus, the endoplasmic reticulum (ER) and dendritic spines. In the nucleus, clusters of Marlin-1 associate to euchromatin. Conclusion Our results demonstrate that Marlin-1 is expressed in discrete areas of the brain. They also confirm the microtubule association at the ultrastructural level in neurons. Together with the abundance of the protein in dendrites and axons they are consistent with the emerging role of Marlin-1 as an intracellular protein linking the cytoskeleton and transport. Our study constitutes the first detailed description of the cellular and subcellular distribution of Marlin-1 in the brain. As such, it will set the basis for future studies on the functional implications of Marlin-1 in protein trafficking.
Collapse
Affiliation(s)
- René L Vidal
- Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia, Santiago, Chile.
| | | | | | | |
Collapse
|
3827
|
Ji S, Li YX, Zhou ZH, Kumar S, Ye J. A bag-of-words approach for Drosophila gene expression pattern annotation. BMC Bioinformatics 2009; 10:119. [PMID: 19383139 PMCID: PMC2680406 DOI: 10.1186/1471-2105-10-119] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 04/21/2009] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Drosophila gene expression pattern images document the spatiotemporal dynamics of gene expression during embryogenesis. A comparative analysis of these images could provide a fundamentally important way for studying the regulatory networks governing development. To facilitate pattern comparison and searching, groups of images in the Berkeley Drosophila Genome Project (BDGP) high-throughput study were annotated with a variable number of anatomical terms manually using a controlled vocabulary. Considering that the number of available images is rapidly increasing, it is imperative to design computational methods to automate this task. RESULTS We present a computational method to annotate gene expression pattern images automatically. The proposed method uses the bag-of-words scheme to utilize the existing information on pattern annotation and annotates images using a model that exploits correlations among terms. The proposed method can annotate images individually or in groups (e.g., according to the developmental stage). In addition, the proposed method can integrate information from different two-dimensional views of embryos. Results on embryonic patterns from BDGP data demonstrate that our method significantly outperforms other methods. CONCLUSION The proposed bag-of-words scheme is effective in representing a set of annotations assigned to a group of images, and the model employed to annotate images successfully captures the correlations among different controlled vocabulary terms. The integration of existing annotation information from multiple embryonic views improves annotation performance.
Collapse
Affiliation(s)
- Shuiwang Ji
- Center for Evolutionary Functional Genomics, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- Department of Computer Science and Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Ying-Xin Li
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210093, PR China
| | - Zhi-Hua Zhou
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210093, PR China
| | - Sudhir Kumar
- Center for Evolutionary Functional Genomics, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Jieping Ye
- Center for Evolutionary Functional Genomics, The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- Department of Computer Science and Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
3828
|
Chan LF, Webb TR, Chung TT, Meimaridou E, Cooray SN, Guasti L, Chapple JP, Egertová M, Elphick MR, Cheetham ME, Metherell LA, Clark AJL. MRAP and MRAP2 are bidirectional regulators of the melanocortin receptor family. Proc Natl Acad Sci U S A 2009; 106:6146-51. [PMID: 19329486 PMCID: PMC2661846 DOI: 10.1073/pnas.0809918106] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Indexed: 01/06/2023] Open
Abstract
The melanocortin receptor (MCR) family consists of 5 G protein-coupled receptors (MC1R-MC5R) with diverse physiologic roles. MC2R is a critical component of the hypothalamic-pituitary-adrenal axis, whereas MC3R and MC4R have an essential role in energy homeostasis. Mutations in MC4R are the single most common cause of monogenic obesity. Investigating the way in which these receptors signal and traffic to the cell membrane is vital in understanding disease processes related to MCR dysfunction. MRAP is an MC2R accessory protein, responsible for adrenal MC2R trafficking and function. Here we identify MRAP2 as a unique homologue of MRAP, expressed in brain and the adrenal gland. We report that MRAP and MRAP2 can interact with all 5 MCRs. This interaction results in MC2R surface expression and signaling. In contrast, MRAP and MRAP2 can reduce MC1R, MC3R, MC4R, and MC5R responsiveness to [Nle4,D-Phe7]alpha-melanocyte-stimulating hormone (NDP-MSH). Collectively, our data identify MRAP and MRAP2 as unique bidirectional regulators of the MCR family.
Collapse
Affiliation(s)
- Li F. Chan
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Tom R. Webb
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Teng-Teng Chung
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Eirini Meimaridou
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Sadani N. Cooray
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Leonardo Guasti
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - J. Paul Chapple
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Michaela Egertová
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom; and
| | - Maurice R. Elphick
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, United Kingdom; and
| | - Michael E. Cheetham
- Division of Molecular and Cellular Neuroscience, University College London Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Louise A. Metherell
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| | - Adrian J. L. Clark
- Queen Mary University of London, Centre for Endocrinology, St. Bartholomew's and Royal London School of Medicine and Dentistry, London EC1M 6BQ, United Kingdom
| |
Collapse
|
3829
|
Gundlach AL, Ma S, Sang Q, Shen PJ, Piccenna L, Sedaghat K, Smith CM, Bathgate RAD, Lawrence AJ, Tregear GW, Wade JD, Finkelstein DI, Bonaventure P, Liu C, Lovenberg TW, Sutton SW. Relaxin Family Peptides and Receptors in Mammalian Brain. Ann N Y Acad Sci 2009; 1160:226-35. [DOI: 10.1111/j.1749-6632.2009.03956.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
3830
|
Jakobsson J, Cordero MI, Bisaz R, Groner AC, Busskamp V, Bensadoun JC, Cammas F, Losson R, Mansuy IM, Sandi C, Trono D. KAP1-mediated epigenetic repression in the forebrain modulates behavioral vulnerability to stress. Neuron 2009; 60:818-31. [PMID: 19081377 DOI: 10.1016/j.neuron.2008.09.036] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 05/06/2008] [Accepted: 09/29/2008] [Indexed: 02/07/2023]
Abstract
KAP1 is an essential cofactor of KRAB-zinc finger proteins, a family of vertebrate-specific epigenetic repressors of largely unknown functions encoded in the hundreds by the mouse and human genomes. Here, we report that KAP1 is expressed at high levels and necessary for KRAB-mediated repression in mature neurons of the mouse brain. Mice deleted for KAP1 in the adult forebrain exhibit heightened levels of anxiety-like and exploratory activity and stress-induced alterations in spatial learning and memory. In the hippocampus, a small number of genes are dysregulated, including some imprinted genes. Chromatin analyses of the promoters of two genes markedly upregulated in knockout mice reveal decreased histone 3 K9-trimethylation and increased histone 3 and histone 4 acetylation. We propose a model in which the tethering of KAP1-associated chromatin remodeling factors via KRAB-ZFPs epigenetically controls gene expression in the hippocampus, thereby conditioning responses to behavioral stress.
Collapse
Affiliation(s)
- Johan Jakobsson
- School of Life Sciences, National Center of Competence in Research, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3831
|
Mulder J, Björling E, Jonasson K, Wernérus H, Hober S, Hökfelt T, Uhlén M. Tissue profiling of the mammalian central nervous system using human antibody-based proteomics. Mol Cell Proteomics 2009; 8:1612-22. [PMID: 19351664 DOI: 10.1074/mcp.m800539-mcp200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A need exists for mapping the protein profiles in the human brain both during normal and disease conditions. Here we studied 800 antibodies generated toward human proteins as part of a Human Protein Atlas program and investigated their suitability for detailed analysis of various levels of a rat brain using immuno-based methods. In this way, the parallel, rather limited analysis of the human brain, restricted to four brain areas (cerebellum, cerebral cortex, hippocampus, and lateral subventricular zone), could be extended in the rat model to 25 selected areas of the brain. Approximately 100 antibodies (12%) revealed a distinct staining pattern and passed validation of specificity using Western blot analysis. These antibodies were applied to coronal sections of the rat brain at 0.7-mm intervals covering the entire brain. We have now produced detailed protein distribution profiles for these antibodies and acquired over 640 images that form the basis of a publicly available portal of an antibody-based Rodent Brain Protein Atlas database (www.proteinatlas.org/rodentbrain). Because of the systematic selection of target genes, the majority of antibodies included in this database are generated against proteins that have not been studied in the brain before. Furthermore optimized tissue processing and colchicine treatment allow a high quality, more extended annotation and detailed analysis of subcellular distributions and protein dynamics.
Collapse
Affiliation(s)
- Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
3832
|
Abstract
In mediating fast synaptic communication in the brain, AMPA receptors require TARP auxiliary proteins. It seems that another distinct class of proteins also bind to AMPA receptors and regulate their function.
Collapse
|
3833
|
Li X, Yankeelov TE, Rosen GD, Gore JC, Dawant BM. Enhancement of histological volumes through averaging and their use for the analysis of magnetic resonance images. Magn Reson Imaging 2009; 27:401-16. [PMID: 18786794 PMCID: PMC2690703 DOI: 10.1016/j.mri.2008.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 06/21/2008] [Accepted: 07/30/2008] [Indexed: 11/17/2022]
Abstract
Magnetic resonance imaging (MRI) of small animals is routinely performed in research centers. But despite its many advantages, MR still suffers from limited spatial resolution which makes the interpretation and quantitative analysis of the images difficult, particularly for small structures of interest within areas of significant heterogeneity. One possibility to address this issue is to complement the MR images with histological data, which requires reconstructing 3D volumes from a series of 2D images. A number of methods have been proposed recently in the literature to address this issue, but deformation or tearing during the slicing process often produces reconstructed volumes with visible artifacts and imperfections. In this paper, we show that a possible solution to this problem is to work with several histological volumes, reconstruct each of these separately and then compute an average. The resulting histological atlas shows structures and substructures more clearly than any individual volume. We also propose an original approach to normalize intensity values across slices, a required preprocessing step when reconstructing histological volumes. We show that the histological atlas we have created can be used to localize structures and substructures, which cannot be seen easily in MR images. We also create an MR atlas that is associated with the histological atlas. We show that using the histological volumes to create the MR atlas is better than using the MR volumes only. Finally, we validate our approach quantitatively on MR image volumes by comparing volumetric measurements obtained manually and obtained automatically with our atlases.
Collapse
Affiliation(s)
- Xia Li
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN 37240-1662, USA.
| | | | | | | | | |
Collapse
|
3834
|
Boonen RA, van Tijn P, Zivkovic D. Wnt signaling in Alzheimer's disease: up or down, that is the question. Ageing Res Rev 2009; 8:71-82. [PMID: 19101658 DOI: 10.1016/j.arr.2008.11.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 11/26/2008] [Accepted: 11/26/2008] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, neuropathologically characterized by amyloid-beta (Abeta) plaques and hyperphosphorylated tau accumulation. AD occurs sporadically (SAD), or is caused by hereditary missense mutations in the amyloid precursor protein (APP) or presenilin-1 and -2 (PSEN1 and PSEN2) genes, leading to early-onset familial AD (FAD). Accumulating evidence points towards a role for altered Wnt/beta-catenin-dependent signaling in the etiology of both forms of AD. Presenilins are involved in modulating beta-catenin stability; therefore FAD-linked PSEN-mediated effects can deregulate the Wnt pathway. Genetic variations in the low-density lipoprotein receptor-related protein 6 and apolipoprotein E in AD have been associated with reduced Wnt signaling. In addition, tau phosphorylation is mediated by glycogen synthase kinase-3 (GSK-3), a key antagonist of the Wnt pathway. In this review, we discuss Wnt/beta-catenin signaling in both SAD and FAD, and recapitulate which of its aberrant functions may be critical for (F)AD pathogenesis. We discuss the intriguing possibility that Abeta toxicity may downregulate the Wnt/beta-catenin pathway, thereby upregulating GSK-3 and consequent tau hyperphosphorylation, linking Abeta and tangle pathology. The currently available evidence implies that disruption of tightly regulated Wnt signaling may constitute a key pathological event in AD. In this context, drug targets aimed at rescuing Wnt signaling may prove to be a constructive therapeutic strategy for AD.
Collapse
|
3835
|
Leyris JP, Gondeau C, Charnet A, Delattre C, Rousset M, Cens T, Charnet P. RGK GTPase-dependent CaV2.1 Ca2+ channel inhibition is independent of CaVbeta-subunit-induced current potentiation. FASEB J 2009; 23:2627-38. [PMID: 19332647 DOI: 10.1096/fj.08-122135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
RGK (Rad-Gem-Rem) GTPases have been described as potent negative regulators of the Ca(2+) influx via high-threshold voltage-activated Ca(2+) channels. Recent work, mostly performed on Ca(V)1.2 Ca(2+) channels, has highlighted the crucial role played by the channel auxiliary Ca(V)beta subunits and identified several GTPase and beta-subunit protein domains involved in this regulation. We now extend these conclusions by producing the first complete characterization of the effects of Gem, Rem, and Rem2 on the neuronal Ca(V)2.1 Ca(2+) channels expressed with Ca(V)beta(1) or Ca(V)beta(2) subunits. Current inhibition is limited to a decrease in amplitude with no modification in the voltage dependence or kinetics of the current. We demonstrate that this inhibition can occur for Ca(V)beta constructs with impaired capacity to induce current potentiation, but that it is lost for Ca(V)beta constructs deleted for their beta-interaction domain. The RGK C-terminal last approximately 80 amino acids are sufficient to allow potent current inhibition and in vivo beta-subunit/Gem interaction. Interestingly, although Gem and Gem carboxy-terminus induce a completely different pattern of beta-subunit cellular localization, they both potently inhibit Ca(V)2.1 channels. These data therefore set the status of neuronal Ca(V)2.1 Ca(2+) channel inhibition by RGK GTPases, emphasizing the role of short amino acid sequences of both proteins in beta-subunit binding and channel inhibition and revealing a new mechanism for channel inhibition.
Collapse
Affiliation(s)
- J-P Leyris
- CRBM, CNRS UMR 5237, Université de Montpellier 1, 34293 Montpellier cedex, France
| | | | | | | | | | | | | |
Collapse
|
3836
|
Bohland JW, Wu C, Barbas H, Bokil H, Bota M, Breiter HC, Cline HT, Doyle JC, Freed PJ, Greenspan RJ, Haber SN, Hawrylycz M, Herrera DG, Hilgetag CC, Huang ZJ, Jones A, Jones EG, Karten HJ, Kleinfeld D, Kötter R, Lester HA, Lin JM, Mensh BD, Mikula S, Panksepp J, Price JL, Safdieh J, Saper CB, Schiff ND, Schmahmann JD, Stillman BW, Svoboda K, Swanson LW, Toga AW, Van Essen DC, Watson JD, Mitra PP. A proposal for a coordinated effort for the determination of brainwide neuroanatomical connectivity in model organisms at a mesoscopic scale. PLoS Comput Biol 2009; 5:e1000334. [PMID: 19325892 PMCID: PMC2655718 DOI: 10.1371/journal.pcbi.1000334] [Citation(s) in RCA: 201] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In this era of complete genomes, our knowledge of neuroanatomical circuitry remains surprisingly sparse. Such knowledge is critical, however, for both basic and clinical research into brain function. Here we advocate for a concerted effort to fill this gap, through systematic, experimental mapping of neural circuits at a mesoscopic scale of resolution suitable for comprehensive, brainwide coverage, using injections of tracers or viral vectors. We detail the scientific and medical rationale and briefly review existing knowledge and experimental techniques. We define a set of desiderata, including brainwide coverage; validated and extensible experimental techniques suitable for standardization and automation; centralized, open-access data repository; compatibility with existing resources; and tractability with current informatics technology. We discuss a hypothetical but tractable plan for mouse, additional efforts for the macaque, and technique development for human. We estimate that the mouse connectivity project could be completed within five years with a comparatively modest budget.
Collapse
Affiliation(s)
- Jason W Bohland
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3837
|
Hwang D, Lee IY, Yoo H, Gehlenborg N, Cho JH, Petritis B, Baxter D, Pitstick R, Young R, Spicer D, Price ND, Hohmann JG, Dearmond SJ, Carlson GA, Hood LE. A systems approach to prion disease. Mol Syst Biol 2009; 5:252. [PMID: 19308092 PMCID: PMC2671916 DOI: 10.1038/msb.2009.10] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 01/20/2009] [Indexed: 01/10/2023] Open
Abstract
Prions cause transmissible neurodegenerative diseases and replicate by conformational conversion of normal benign forms of prion protein (PrPC) to disease-causing PrPSc isoforms. A systems approach to disease postulates that disease arises from perturbation of biological networks in the relevant organ. We tracked global gene expression in the brains of eight distinct mouse strain–prion strain combinations throughout the progression of the disease to capture the effects of prion strain, host genetics, and PrP concentration on disease incubation time. Subtractive analyses exploiting various aspects of prion biology and infection identified a core of 333 differentially expressed genes (DEGs) that appeared central to prion disease. DEGs were mapped into functional pathways and networks reflecting defined neuropathological events and PrPSc replication and accumulation, enabling the identification of novel modules and modules that may be involved in genetic effects on incubation time and in prion strain specificity. Our systems analysis provides a comprehensive basis for developing models for prion replication and disease, and suggests some possible therapeutic approaches.
Collapse
Affiliation(s)
- Daehee Hwang
- Institute for Systems Biology, Seattle, WA 98103, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3838
|
Ruths T, Ruths D, Nakhleh L. GS2: an efficiently computable measure of GO-based similarity of gene sets. Bioinformatics 2009; 25:1178-84. [PMID: 19289444 PMCID: PMC2672633 DOI: 10.1093/bioinformatics/btp128] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Motivation: The growing availability of genome-scale datasets has attracted increasing attention to the development of computational methods for automated inference of functional similarities among genes and their products. One class of such methods measures the functional similarity of genes based on their distance in the Gene Ontology (GO). To measure the functional relatedness of a gene set, these measures consider every pair of genes in the set, and the average of all pairwise distances is calculated. However, as more data becomes available and gene sets used for analysis become larger, such pair-based calculation becomes prohibitive. Results: In this article, we propose GS2 (GO-based similarity of gene sets), a novel GO-based measure of gene set similarity that is computable in linear time in the size of the gene set. The measure quantifies the similarity of the GO annotations among a set of genes by averaging the contribution of each gene's GO terms and their ancestor terms with respect to the GO vocabulary graph. To study the performance of our method, we compared our measure with an established pair-based measure when run on gene sets with varying degrees of functional similarities. In addition to a significant speed improvement, our method produced comparable similarity scores to the established method. Our method is available as a web-based tool and an open-source Python library. Availability: The web-based tools and Python code are available at: http://bioserver.cs.rice.edu/gs2. Contact:troy.ruths@rice.edu
Collapse
Affiliation(s)
- Troy Ruths
- Department of Computer Science, Rice University, 6100 Main Street, MS 132, Houston, TX, USA.
| | | | | |
Collapse
|
3839
|
Xiong L, Catoire H, Dion P, Gaspar C, Lafrenière RG, Girard SL, Levchenko A, Rivière JB, Fiori L, St-Onge J, Bachand I, Thibodeau P, Allen R, Earley C, Turecki G, Montplaisir J, Rouleau GA. MEIS1 intronic risk haplotype associated with restless legs syndrome affects its mRNA and protein expression levels. Hum Mol Genet 2009; 18:1065-74. [PMID: 19126776 PMCID: PMC2722232 DOI: 10.1093/hmg/ddn443] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 12/12/2008] [Accepted: 12/30/2008] [Indexed: 11/13/2022] Open
Abstract
Restless legs syndrome (RLS) is a common neurological disorder characterized by an irresistible urge to move the legs at night, which is often accompanied by unpleasant sensations. A recent genomewide association study identified an association between RLS and intronic markers from the MEIS1 gene. Comparative genomic analysis indicates that MEIS1 is the only gene encompassed in this evolutionarily conserved chromosomal segment, i.e. a conservation synteny block, from mammals to fish. We carried out a series of experiments to delineate the role of MEIS1 in RLS pathogenesis and the underlying genetic mechanism. We sequenced all 13 MEIS1 exons and their splice junctions in 285 RLS probands with confirmed clinical diagnosis and did not identify any causative coding or exon-intron junction mutations. We found no evidence of structural variation or disease-associated haplotype differential splicing. However, sequencing of conserved regions of MEIS1 introns 8 and 9 identified a novel single nucleotide polymorphism (C13B_2) significantly associated with RLS (allelic association, P = 1.81E-07). We detected a significant decrease in MEIS1 mRNA expression by quantitative real-time polymerase chain reaction in lymphoblastoid cell lines (LCLs) and brain tissues from RLS patients homozygous for the intronic RLS risk haplotype, compared with those homozygous for the non-risk haplotype. Finally, we found significantly decreased MEIS1 protein levels in the same batch of LCLs and brain tissues from the homozygous carriers of the risk haplotype, compared with the homozygous non-carriers. Therefore, these data suggest that reduced expression of the MEIS1 gene, possibly through intronic cis-regulatory element(s), predisposes to RLS.
Collapse
Affiliation(s)
- Lan Xiong
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Hélène Catoire
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Patrick Dion
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Claudia Gaspar
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Ronald G. Lafrenière
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Simon L. Girard
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Anastasia Levchenko
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Jean-Baptiste Rivière
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Laura Fiori
- Research Center, Douglas Hospital, McGill University, Montréal, Québec, H4H 1R3, Canada
| | - Judith St-Onge
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Isabelle Bachand
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Pascale Thibodeau
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| | - Richard Allen
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Christopher Earley
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Gustavo Turecki
- Research Center, Douglas Hospital, McGill University, Montréal, Québec, H4H 1R3, Canada
| | - Jacques Montplaisir
- Centre d'Étude du Sommeil, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec, H4J 1C5, Canada
| | - Guy A. Rouleau
- Centre of Excellence in Neuromics of University of Montreal, CHUM Research Centerand Department of Medicine, University of Montreal, Montréal, Québec, H2L 4M1, Canada
| |
Collapse
|
3840
|
Ruttenberg A, Rees JA, Samwald M, Marshall MS. Life sciences on the Semantic Web: the Neurocommons and beyond. Brief Bioinform 2009; 10:193-204. [PMID: 19282504 DOI: 10.1093/bib/bbp004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Translational research, the effort to couple the results of basic research to clinical applications, depends on the ability to effectively answer questions using information that spans multiple disciplines. The Semantic Web, with its emphasis on combining information using standard representation languages, access to that information via standard web protocols, and technologies to leverage computation, such as in the form of inference and distributable query, offers a social and technological basis for assembling, integrating and making available biomedical knowledge at Web scale. In this article, we discuss the use of Semantic Web technology for assembling and querying biomedical knowledge from multiple sources and disciplines. We present the Neurocommons prototype knowledge base, a demonstration intended to show the feasibility and benefits of using these technologies. The prototype knowledge base can be used to experiment with and assess the scalability of current tools and methods for creating such a resource, and to elicit issues that will need to be addressed in order to expand the scope and use of it. We demonstrate the utility of the knowledge base by reviewing a few example queries that provide answers to precise questions relevant to the understanding of disease. All components of the knowledge base are freely available at http://neurocommons.org/, enabling readers to reconstruct the knowledge base and experiment with this new technology.
Collapse
Affiliation(s)
- Alan Ruttenberg
- Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
3841
|
Dahlberg CL, Nguyen EZ, Goodlett D, Kimelman D. Interactions between Casein kinase Iepsilon (CKIepsilon) and two substrates from disparate signaling pathways reveal mechanisms for substrate-kinase specificity. PLoS One 2009; 4:e4766. [PMID: 19274088 PMCID: PMC2651596 DOI: 10.1371/journal.pone.0004766] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 02/10/2009] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Members of the Casein Kinase I (CKI) family of serine/threonine kinases regulate diverse biological pathways. The seven mammalian CKI isoforms contain a highly conserved kinase domain and divergent amino- and carboxy-termini. Although they share a preferred target recognition sequence and have overlapping expression patterns, individual isoforms often have specific substrates. In an effort to determine how substrates recognize differences between CKI isoforms, we have examined the interaction between CKIepsilon and two substrates from different signaling pathways. METHODOLOGY/PRINCIPAL FINDINGS CKIepsilon, but not CKIalpha, binds to and phosphorylates two proteins: Period, a transcriptional regulator of the circadian rhythms pathway, and Disheveled, an activator of the planar cell polarity pathway. We use GST-pull-down assays data to show that two key residues in CKIalpha's kinase domain prevent Disheveled and Period from binding. We also show that the unique C-terminus of CKIepsilon does not determine Dishevelled's and Period's preference for CKIepsilon nor is it essential for binding, but instead plays an auxillary role in stabilizing the interactions of CKIepsilon with its substrates. We demonstrate that autophosphorylation of CKIepsilon's C-terminal tail prevents substrate binding, and use mass spectrometry and chemical crosslinking to reveal how a phosphorylation-dependent interaction between the C-terminal tail and the kinase domain prevents substrate phosphorylation and binding. CONCLUSIONS/SIGNIFICANCE The biochemical interactions between CKIepsilon and Disheveled, Period, and its own C-terminus lead to models that explain CKIepsilon's specificity and regulation.
Collapse
Affiliation(s)
- Caroline Lund Dahlberg
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
| | - Elizabeth Z. Nguyen
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, United States of America
| | - David Goodlett
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, United States of America
| | - David Kimelman
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
3842
|
Jørgensen JR, Thompson L, Fjord-Larsen L, Krabbe C, Torp M, Kalkkinen N, Hansen C, Wahlberg L. Characterization of Meteorin—An Evolutionary Conserved Neurotrophic Factor. J Mol Neurosci 2009; 39:104-16. [DOI: 10.1007/s12031-009-9189-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 02/16/2009] [Indexed: 01/05/2023]
|
3843
|
Manipulation of tissue contrast using contrast agents for enhanced MR microscopy in ex vivo mouse brain. Neuroimage 2009; 46:589-99. [PMID: 19264139 DOI: 10.1016/j.neuroimage.2009.02.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 12/22/2008] [Accepted: 02/16/2009] [Indexed: 11/20/2022] Open
Abstract
Detailed 3D mouse brain images may promote better understanding of phenotypical differences between normal and transgenic/mutant mouse models. Previously, a number of magnetic resonance microscopy (MRM) studies have successfully established brain atlases, revealing genotypic traits of several commonly used mouse strains. In such studies, MR contrast agents, mainly gadolinium (Gd) based, were often used to reduce acquisition time and improve signal-to-noise ratio (SNR). In this paper, we intended to extend the utility of contrast agents for MRM applications. Using Gd-DTPA and MnCl(2), we exploited the potential use of MR contrast agents to manipulate image contrast by drawing upon the multiple relaxation mechanisms and tissue-dependent staining properties characteristic of each contrast agent. We quantified r(1) and r(2) of Gd-DTPA and MnCl(2) in both aqueous solution and brain tissue and demonstrated the presence of divergent relaxation mechanisms between solution and tissue for each contrast agent. Further analyses using nuclear magnetic resonance dispersion (NMRD) of Mn(2+) in ex vivo tissue strongly suggested macromolecule binding of Mn(2+), leading to increased T(1) relaxation. Moreover, inductively coupled plasma (ICP) mass spectroscopy revealed that MnCl(2) had higher tissue affinity than Gd-DTPA. As a result, multiple regions of the brain stained by the two agents exhibited different image contrasts. Our results show that differential MRM staining can be achieved using multiple MR contrast agents, revealing detailed cytoarchitecture, and may ultimately offer a window for investigating new techniques by which to understand biophysical MR relaxation mechanisms and perhaps to visualize tissue anomalies even at the molecular level.
Collapse
|
3844
|
Greene JG, Borges K, Dingledine R. Quantitative transcriptional neuroanatomy of the rat hippocampus: evidence for wide-ranging, pathway-specific heterogeneity among three principal cell layers. Hippocampus 2009; 19:253-64. [PMID: 18830999 PMCID: PMC2649995 DOI: 10.1002/hipo.20502] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We have used laser-capture microdissection and microarray hybridization to characterize gene expression in the three principal neuron layers of rat hippocampus. Correlative and clustering analyses revealed all three layers to be easily differentiated from one another based on gene expression profile alone. A greater disparity in gene expression exists between dentate granule and pyramidal cell layers, reflecting phenotypic and ontological differences between those cell populations. Remarkably, the level of more than 45% of expressed transcripts was significantly different among the three neuron populations, with more than a third of those (>1,000 transcripts) being at least twofold different between layers. Even CA1 and CA3 pyramidal cell layers were dramatically different on a transcriptional level with a separate analysis indicating that nearly 20% of transcripts are differentially expressed between them. Only a small number of transcripts were specific for a given hippocampal cell layer, suggesting that functional differences are more likely secondary to wide-ranging expression differences of modest magnitude rather than very large disparities in a few genes. Categorical analysis of transcript abundance revealed concerted differences in gene expression among the three cell layers referable to specific cellular pathways. For instance, transcripts encoding proteins involved in glucose metabolism are most highly expressed in the CA3 pyramidal layer, which may reflect an underlying greater metabolic rate of these neurons and partially explain their exquisite vulnerability to seizure-induced damage. Conversely, transcripts related to MAP kinase signaling pathways and transcriptional regulator activity are prominent in the dentate granule cell layer, which could contribute to its resistance to damage following seizure activity by positioning these neurons to respond to external stimuli by altering transcription. Taken together, these data suggest that unique physiological characteristics of major cell layers, such as neuronal activity, neuronal plasticity, and vulnerability to neurodegeneration, are reflected in substantial transcriptional heterogeneity within the hippocampus.
Collapse
Affiliation(s)
- James G Greene
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.
| | | | | |
Collapse
|
3845
|
Kfoury N, Kapatos G. Identification of neuronal target genes for CCAAT/enhancer binding proteins. Mol Cell Neurosci 2009; 40:313-27. [PMID: 19103292 PMCID: PMC2703816 DOI: 10.1016/j.mcn.2008.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Revised: 11/04/2008] [Accepted: 11/05/2008] [Indexed: 01/19/2023] Open
Abstract
CCAAT/Enhancer Binding Proteins (C/EBPs) play pivotal roles in the development and plasticity of the nervous system. Identification of the physiological targets of C/EBPs (C/EBP target genes) should therefore provide insight into the underlying biology of these processes. We used unbiased genome-wide mapping to identify 115 C/EBPbeta target genes in PC12 cells that include transcription factors, neurotransmitter receptors, ion channels, protein kinases and synaptic vesicle proteins. C/EBPbeta binding sites were located primarily within introns, suggesting novel regulatory functions, and were associated with binding sites for other developmentally important transcription factors. Experiments using dominant negatives showed C/EBPbeta to repress transcription of a subset of target genes. Target genes in rat brain were subsequently found to preferentially bind C/EBPalpha, beta and delta. Analysis of the hippocampal transcriptome of C/EBPbeta knockout mice revealed dysregulation of a high percentage of transcripts identified as C/EBP target genes. These results support the hypothesis that C/EBPs play non-redundant roles in the brain.
Collapse
Affiliation(s)
- Najla Kfoury
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | | |
Collapse
|
3846
|
Wfs1-deficient mice display impaired behavioural adaptation in stressful environment. Behav Brain Res 2009; 198:334-45. [DOI: 10.1016/j.bbr.2008.11.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 10/28/2008] [Accepted: 11/04/2008] [Indexed: 11/19/2022]
|
3847
|
Global implications of mRNA localization pathways in cellular organization. Curr Opin Cell Biol 2009; 21:409-15. [PMID: 19249199 DOI: 10.1016/j.ceb.2009.01.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 01/27/2009] [Accepted: 01/29/2009] [Indexed: 12/21/2022]
Abstract
Genome expression profiling has led to the important realization that RNA molecules are more numerous and diverse than previously expected. One aspect of RNA biology that is just beginning to be fully appreciated is the extent to which mRNAs are transported to specific subcellular destinations before being translated, an exquisite mechanism for targeting proteins where they are required in the cell. While several excellent reviews have discussed the subject of mRNA localization, it is only in recent years that high-throughput technologies have been applied to address issues such as the extent and diversity of RNA localization events and mechanisms. This review focuses on these recent functional genomic approaches, their implications, and the new tools and methods that will be needed to further elucidate mRNA localization pathways.
Collapse
|
3848
|
Nagarajan RP, Costello JF. Epigenetic mechanisms in glioblastoma multiforme. Semin Cancer Biol 2009; 19:188-97. [PMID: 19429483 DOI: 10.1016/j.semcancer.2009.02.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 02/11/2009] [Indexed: 11/26/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive and lethal cancer, accounting for the majority of primary brain tumors in adults. GBMs are characterized by genetic alterations large and small, affecting genes that control cell growth, apoptosis, angiogenesis, and invasion. Epigenetic alterations also affect the expression of cancer genes alone, or in combination with genetic mechanisms. For example, in each GBM, hundreds of genes are subject to DNA hypermethylation at their CpG island promoters. A subset of GBMs is also characterized by locus-specific and genome-wide decrease in DNA methylation, or DNA hypomethylation. Other epigenetic alterations, such as changes in the position of histone variants and changes in histone modifications are also likely important in the molecular pathology of GBM, but somewhat surprisingly there are very limited data about these in GBM. Alterations in histone modifications are especially important to understand, given that histone deacetylases are targets for drugs that are in clinical trial for GBMs. The technological wave of next-generation sequencing will accelerate GBM epigenome profiling, allowing the direct integration of DNA methylation, histone modification and gene expression profiles. Ultimately, genomic and epigenomic data should provide new predictive markers of response and lead to more effective therapies for GBM.
Collapse
Affiliation(s)
- Raman P Nagarajan
- Brain Tumor Research Center, Department of Neurosurgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
3849
|
Bifari F, Decimo I, Chiamulera C, Bersan E, Malpeli G, Johansson J, Lisi V, Bonetti B, Fumagalli G, Pizzolo G, Krampera M. Novel stem/progenitor cells with neuronal differentiation potential reside in the leptomeningeal niche. J Cell Mol Med 2009; 13:3195-208. [PMID: 19228261 PMCID: PMC4516477 DOI: 10.1111/j.1582-4934.2009.00706.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Stem cells capable of generating neural differentiated cells are recognized by the expression of nestin and reside in specific regions of the brain, namely, hippocampus, subventricular zone and olfactory bulb. For other brain structures, such as leptomeninges, which contribute to the correct cortex development and functions, there is no evidence so far that they may contain stem/precursor cells. In this work, we show for the first time that nestin-positive cells are present in rat leptomeninges during development up to adulthood. The newly identified nestin-positive cells can be extracted and expanded in vitro both as neurospheres, displaying high similarity with subventricular zone-derived neural stem cells, and as homogeneous cell population with stem cell features. In vitro expanded stem cell population can differentiate with high efficiency into excitable cells with neuronal phenotype and morphology. Once injected into the adult brain, these cells survive and differentiate into neurons, thus showing that their neuronal differentiation potential is operational also in vivo. In conclusion, our data provide evidence that a specific population of immature cells endowed of neuronal differentiation potential is resident in the leptomeninges throughout the life. As leptomeninges cover the entire central nervous system, these findings could have relevant implications for studies on cortical development and for regenerative medicine applied to neurological disorders.
Collapse
Affiliation(s)
- Francesco Bifari
- Department of Clinical and Experimental Medicine, Stem Cell Research Laboratory, Section of Hematology, University of Verona, Verona, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3850
|
Watakabe A. Comparative molecular neuroanatomy of mammalian neocortex: what can gene expression tell us about areas and layers? Dev Growth Differ 2009; 51:343-54. [PMID: 19222526 DOI: 10.1111/j.1440-169x.2008.01085.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is over 100 years since Brodmann proposed the homology of layer and area structure of the cerebral cortex across species. His proposal was based on the extensive comparative analyses of various mammalian brains. Although such homology is now well accepted, the recent data in our laboratory showed striking variations of gene expression patterns across areas and species. Are cortical layers and areas really homologous? If they are, to what extent and how are they similar or different? We are trying to answer these questions by identifying the homologous neuronal types common to various areas and species. Toward this goal, we started to classify the cortical pyramidal neurons by expression of particular sets of genes. By using fluorescent double in situ hybridization combined with retrograde tracers, we are characterizing the gene expression phenotypes and projection specificity of cortical excitatory neuron types. In this review, I discuss the recent findings in our laboratory in light of the past and present knowledge about cortical cell types, which provides insight to the homology (and lack thereof) of the mammalian neocortical organization.
Collapse
Affiliation(s)
- Akiya Watakabe
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki 444-8585, Japan.
| |
Collapse
|