Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neuron ; 109(8): 1283-1301.e6, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33675684

ABSTRACT

Loss-of-function TREM2 mutations strongly increase Alzheimer's disease (AD) risk. Trem2 deletion has revealed protective Trem2 functions in preclinical models of ß-amyloidosis, a prominent feature of pre-diagnosis AD stages. How TREM2 influences later AD stages characterized by tau-mediated neurodegeneration is unclear. To understand Trem2 function in the context of both ß-amyloid and tau pathologies, we examined Trem2 deficiency in the pR5-183 mouse model expressing mutant tau alone or in TauPS2APP mice, in which ß-amyloid pathology exacerbates tau pathology and neurodegeneration. Single-cell RNA sequencing in these models revealed robust disease-associated microglia (DAM) activation in TauPS2APP mice that was amyloid-dependent and Trem2-dependent. In the presence of ß-amyloid pathology, Trem2 deletion further exacerbated tau accumulation and spreading and promoted brain atrophy. Without ß-amyloid pathology, Trem2 deletion did not affect these processes. Therefore, TREM2 may slow AD progression and reduce tau-driven neurodegeneration by restricting the degree to which ß-amyloid facilitates the spreading of pathogenic tau.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Brain/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Brain/pathology , Disease Models, Animal , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Receptors, Immunologic/genetics , tau Proteins/genetics
2.
Cell Rep ; 31(13): 107843, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610143

ABSTRACT

Damage-associated microglia (DAM) profiles observed in Alzheimer's disease (AD)-related mouse models reflect an activation state that could modulate AD risk or progression. To learn whether human AD microglia (HAM) display a similar profile, we develop a method for purifying cell types from frozen cerebrocortical tissues for RNA-seq analysis, allowing better transcriptome coverage than typical single-nucleus RNA-seq approaches. The HAM profile we observe bears little resemblance to the DAM profile. Instead, HAM display an enhanced human aging profile, in addition to other disease-related changes such as APOE upregulation. Analyses of whole-tissue RNA-seq and single-cell/nucleus RNA-seq datasets corroborate our findings and suggest that the lack of DAM response in human microglia occurs specifically in AD tissues, not other neurodegenerative settings. These results, which can be browsed at http://research-pub.gene.com/BrainMyeloidLandscape, provide a genome-wide picture of microglial activation in human AD and highlight considerable differences between mouse models and human disease.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cellular Senescence/genetics , Microglia/metabolism , Microglia/pathology , Transcriptional Activation/genetics , Aged , Aged, 80 and over , Animals , Databases, Genetic , Female , Frontal Lobe/pathology , Frozen Sections , Gene Expression Profiling , Genetic Predisposition to Disease , Heterografts , Humans , Male , Mice , Monocytes/metabolism , Multiple Sclerosis/pathology , Phenotype , Reproducibility of Results , Risk Factors , Temporal Lobe/pathology
3.
J Neurosci ; 40(5): 958-973, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31831521

ABSTRACT

Cortical circuit activity is shaped by the parvalbumin (PV) and somatostatin (SST) interneurons that inhibit principal excitatory (EXC) neurons and the vasoactive intestinal peptide (VIP) interneurons that suppress activation of other interneurons. To understand the molecular-genetic basis of functional specialization and identify potential drug targets specific to each neuron subtype, we performed a genome wide assessment of both gene expression and splicing across EXC, PV, SST and VIP neurons from male and female mouse brains. These results reveal numerous examples where neuron subtype-specific gene expression, as well as splice-isoform usage, can explain functional differences between neuron subtypes, including in presynaptic plasticity, postsynaptic receptor function, and synaptic connectivity specification. We provide a searchable web resource for exploring differential mRNA expression and splice form usage between excitatory, PV, SST, and VIP neurons (http://research-pub.gene.com/NeuronSubtypeTranscriptomes). This resource, combining a unique new dataset and novel application of analysis methods to multiple relevant datasets, identifies numerous potential drug targets for manipulating circuit function, reveals neuron subtype-specific roles for disease-linked genes, and is useful for understanding gene expression changes observed in human patient brains.SIGNIFICANCE STATEMENT Understanding the basis of functional specialization of neuron subtypes and identifying drug targets for manipulating circuit function requires comprehensive information on cell-type-specific transcriptional profiles. We sorted excitatory neurons and key inhibitory neuron subtypes from mouse brains and assessed differential mRNA expression. We used a genome-wide analysis which not only examined differential gene expression levels but could also detect differences in splice isoform usage. This analysis reveals numerous examples of neuron subtype-specific isoform usage with functional importance, identifies potential drug targets, and provides insight into the neuron subtypes involved in psychiatric disease. We also apply our analysis to two other relevant datasets for comparison, and provide a searchable website for convenient access to the resource.


Subject(s)
Cerebral Cortex/metabolism , Interneurons/metabolism , Neurons/metabolism , Transcriptome , Animals , Cells, Cultured , Female , Hippocampus/metabolism , Male , Mice, Transgenic , Parvalbumins/metabolism , RNA, Messenger/metabolism , Somatostatin/metabolism , Vasoactive Intestinal Peptide/metabolism
4.
Cell Rep ; 28(8): 2111-2123.e6, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433986

ABSTRACT

Complement pathway overactivation can lead to neuronal damage in various neurological diseases. Although Alzheimer's disease (AD) is characterized by ß-amyloid plaques and tau tangles, previous work examining complement has largely focused on amyloidosis models. We find that glial cells show increased expression of classical complement components and the central component C3 in mouse models of amyloidosis (PS2APP) and more extensively tauopathy (TauP301S). Blocking complement function by deleting C3 rescues plaque-associated synapse loss in PS2APP mice and ameliorates neuron loss and brain atrophy in TauP301S mice, improving neurophysiological and behavioral measurements. In addition, C3 protein is elevated in AD patient brains, including at synapses, and levels and processing of C3 are increased in AD patient CSF and correlate with tau. These results demonstrate that complement activation contributes to neurodegeneration caused by tau pathology and suggest that blocking C3 function might be protective in AD and other tauopathies.


Subject(s)
Alzheimer Disease/immunology , Amyloidosis/immunology , Complement C3/metabolism , Nerve Degeneration/immunology , Tauopathies/immunology , Alzheimer Disease/genetics , Animals , Atrophy , Behavior, Animal , Biomarkers/metabolism , Brain/pathology , Complement C1q/metabolism , Complement C3/cerebrospinal fluid , Complement C3/genetics , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation , Humans , Male , Mice, Transgenic , Nerve Degeneration/genetics , Neurons/metabolism , Neurons/pathology , Plaque, Amyloid/metabolism , Synapses/metabolism
5.
Neuron ; 100(6): 1322-1336.e7, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30392797

ABSTRACT

Synapse loss and Tau pathology are hallmarks of Alzheimer's disease (AD) and other tauopathies, but how Tau pathology causes synapse loss is unclear. We used unbiased proteomic analysis of postsynaptic densities (PSDs) in Tau-P301S transgenic mice to identify Tau-dependent alterations in synapses prior to overt neurodegeneration. Multiple proteins and pathways were altered in Tau-P301S PSDs, including depletion of a set of GTPase-regulatory proteins that leads to actin cytoskeletal defects and loss of dendritic spines. Furthermore, we found striking accumulation of complement C1q in the PSDs of Tau-P301S mice and AD patients. At synapses, C1q decorated perisynaptic membranes, accumulated in correlation with phospho-Tau, and was associated with augmented microglial engulfment of synapses and decline of synapse density. A C1q-blocking antibody inhibited microglial synapse removal in cultured neurons and in Tau-P301S mice, rescuing synapse density. Thus, inhibiting complement-mediated synapse removal by microglia could be a potential therapeutic target for Tau-associated neurodegeneration.


Subject(s)
Antibodies/therapeutic use , Complement C1q/immunology , Synapses/metabolism , Tauopathies/drug therapy , Tauopathies/pathology , tau Proteins/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Complement C1q/metabolism , Complement C1q/ultrastructure , Embryo, Mammalian , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Post-Synaptic Density/ultrastructure , Presenilin-2/genetics , Presenilin-2/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Proteome/metabolism , Rats , Synapses/drug effects , Synapses/ultrastructure , Tauopathies/diagnostic imaging , Tauopathies/genetics , tau Proteins/genetics
6.
Cell Rep ; 22(3): 832-847, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29346778

ABSTRACT

Microglia, the CNS-resident immune cells, play important roles in disease, but the spectrum of their possible activation states is not well understood. We derived co-regulated gene modules from transcriptional profiles of CNS myeloid cells of diverse mouse models, including new tauopathy model datasets. Using these modules to interpret single-cell data from an Alzheimer's disease (AD) model, we identified microglial subsets-distinct from previously reported "disease-associated microglia"-expressing interferon-related or proliferation modules. We then analyzed whole-tissue RNA profiles from human neurodegenerative diseases, including a new AD dataset. Correcting for altered cellular composition of AD tissue, we observed elevated expression of the neurodegeneration-related modules, but also modules not implicated using expression profiles from mouse models alone. We provide a searchable, interactive database for exploring gene expression in all these datasets (http://research-pub.gene.com/BrainMyeloidLandscape). Understanding the dimensions of CNS myeloid cell activation in human disease may reveal opportunities for therapeutic intervention.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , Microglia/metabolism , Alzheimer Disease/metabolism , Animals , Disease Models, Animal , Humans , Mice
7.
BMC Genomics ; 17: 61, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26768488

ABSTRACT

BACKGROUND: RNA-editing is a tightly regulated, and essential cellular process for a properly functioning brain. Dysfunction of A-to-I RNA editing can have catastrophic effects, particularly in the central nervous system. Thus, understanding how the process of RNA-editing is regulated has important implications for human health. However, at present, very little is known about the regulation of editing across tissues, and individuals. RESULTS: Here we present an analysis of RNA-editing patterns from 9 different tissues harvested from a single mouse. For comparison, we also analyzed data for 5 of these tissues harvested from 15 additional animals. We find that tissue specificity of editing largely reflects differential expression of substrate transcripts across tissues. We identified a surprising enrichment of editing in intronic regions of brain transcripts, that could account for previously reported higher levels of editing in brain. There exists a small but remarkable amount of editing which is tissue-specific, despite comparable expression levels of the edit site across multiple tissues. Expression levels of editing enzymes and their isoforms can explain some, but not all of this variation. CONCLUSIONS: Together, these data suggest a complex regulation of the RNA-editing process beyond transcript expression levels.


Subject(s)
Adenosine Deaminase/genetics , Organ Specificity/genetics , RNA Editing/genetics , RNA-Binding Proteins/genetics , Adenosine Deaminase/biosynthesis , Animals , Brain/growth & development , Brain/metabolism , Gene Expression Regulation , Humans , Introns/genetics , Mice , Protein Isoforms/genetics , RNA-Binding Proteins/biosynthesis , Transcription, Genetic
8.
Neuron ; 89(1): 70-82, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26687840

ABSTRACT

The blood-brain barrier (BBB) poses a major challenge for developing effective antibody therapies for neurological diseases. Using transcriptomic and proteomic profiling, we searched for proteins in mouse brain endothelial cells (BECs) that could potentially be exploited to transport antibodies across the BBB. Due to their limited protein abundance, neither antibodies against literature-identified targets nor BBB-enriched proteins identified by microarray facilitated significant antibody brain uptake. Using proteomic analysis of isolated mouse BECs, we identified multiple highly expressed proteins, including basigin, Glut1, and CD98hc. Antibodies to each of these targets were significantly enriched in the brain after administration in vivo. In particular, antibodies against CD98hc showed robust accumulation in brain after systemic dosing, and a significant pharmacodynamic response as measured by brain Aß reduction. The discovery of CD98hc as a robust receptor-mediated transcytosis pathway for antibody delivery to the brain expands the current approaches available for enhancing brain uptake of therapeutic antibodies.


Subject(s)
Antibodies/therapeutic use , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Brain/metabolism , Receptors, Transferrin/metabolism , Animals , Antibodies/immunology , Endothelial Cells/metabolism , Fusion Regulatory Protein 1, Heavy Chain/immunology , Mice , Proteomics/methods , Transcytosis/physiology
9.
Front Neurosci ; 8: 355, 2014.
Article in English | MEDLINE | ID: mdl-25414634

ABSTRACT

The availability of genome-wide expression data for the blood-brain barrier is an invaluable resource that has recently enabled the discovery of several genes and pathways involved in the development and maintenance of the blood-brain barrier, particularly in rodent models. The broad distribution of published data sets represents a viable starting point for the molecular dissection of the blood-brain barrier and will further direct the discovery of novel mechanisms of blood-brain barrier formation and function. Technical advances in purifying brain endothelial cells, the key cell that forms the critical barrier, have allowed for greater specificity in gene expression comparisons with other central nervous system cell types, and more systematic characterizations of the molecular composition of the blood-brain barrier. Nevertheless, our understanding of how the blood-brain barrier changes during aging and disease is underrepresented. Blood-brain barrier data sets from a wider range of experimental paradigms and species, including invertebrates and primates, would be invaluable for investigating the function and evolution of the blood-brain barrier. Newer technologies in gene expression profiling, such as RNA-sequencing, now allow for finer resolution of transcriptomic changes, including isoform specificity and RNA-editing. As our field continues to utilize more advanced expression profiling in its ongoing efforts to elucidate the blood-brain barrier, including in disease and drug delivery, we will continue to see rapid advances in our understanding of the molecular mediators of barrier biology. We predict that the recently published data sets, combined with forthcoming genomic and proteomic blood-brain barrier data sets, will continue to fuel the molecular genetic revolution of.

10.
Pac Symp Biocomput ; : 75-86, 2014.
Article in English | MEDLINE | ID: mdl-24297535

ABSTRACT

Cancer cells derived from different stages of tumor progression may exhibit distinct biological properties, as exemplified by the paired lung cancer cell lines H1993 and H2073. While H1993 was derived from chemo-naive metastasized tumor, H2073 originated from the chemo-resistant primary tumor from the same patient and exhibits strikingly different drug response profile. To understand the underlying genetic and epigenetic bases for their biological properties, we investigated these cells using a wide range of large-scale methods including whole genome sequencing, RNA sequencing, SNP array, DNA methylation array, and de novo genome assembly. We conducted an integrative analysis of both cell lines to distinguish between potential driver and passenger alterations. Although many genes are mutated in these cell lines, the combination of DNA- and RNA-based variant information strongly implicates a small number of genes including TP53 and STK11 as likely drivers. Likewise, we found a diverse set of genes differentially expressed between these cell lines, but only a fraction can be attributed to changes in DNA copy number or methylation. This set included the ABC transporter ABCC4, implicated in drug resistance, and the metastasis associated MET oncogene. While the rich data content allowed us to reduce the space of hypotheses that could explain most of the observed biological properties, we also caution there is a lack of statistical power and inherent limitations in such single patient case studies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Computational Biology , DNA Methylation , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Gene Dosage , Gene Expression Profiling/statistics & numerical data , Genomics/statistics & numerical data , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Models, Genetic , Mutation
11.
Bioinformatics ; 29(24): 3220-1, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24078713

ABSTRACT

UNLABELLED: It is common for computational analyses to generate large amounts of complex data that are difficult to process and share with collaborators. Standard methods are needed to transform such data into a more useful and intuitive format. We present ReportingTools, a Bioconductor package, that automatically recognizes and transforms the output of many common Bioconductor packages into rich, interactive, HTML-based reports. Reports are not generic, but have been individually designed to reflect content specific to the result type detected. Tabular output included in reports is sortable, filterable and searchable and contains context-relevant hyperlinks to external databases. Additionally, in-line graphics have been developed for specific analysis types and are embedded by default within table rows, providing a useful visual summary of underlying raw data. ReportingTools is highly flexible and reports can be easily customized for specific applications using the well-defined API. AVAILABILITY: The ReportingTools package is implemented in R and available from Bioconductor (version ≥ 2.11) at the URL: http://bioconductor.org/packages/release/bioc/html/ReportingTools.html. Installation instructions and usage documentation can also be found at the above URL.


Subject(s)
Computational Biology , Gene Expression Profiling/methods , Genomics/methods , High-Throughput Nucleotide Sequencing , Software , Algorithms , Databases, Factual
12.
Nature ; 488(7413): 660-4, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22895193

ABSTRACT

Identifying and understanding changes in cancer genomes is essential for the development of targeted therapeutics. Here we analyse systematically more than 70 pairs of primary human colon tumours by applying next-generation sequencing to characterize their exomes, transcriptomes and copy-number alterations. We have identified 36,303 protein-altering somatic changes that include several new recurrent mutations in the Wnt pathway gene TCF7L2, chromatin-remodelling genes such as TET2 and TET3 and receptor tyrosine kinases including ERBB3. Our analysis for significantly mutated cancer genes identified 23 candidates, including the cell cycle checkpoint kinase ATM. Copy-number and RNA-seq data analysis identified amplifications and corresponding overexpression of IGF2 in a subset of colon tumours. Furthermore, using RNA-seq data we identified multiple fusion transcripts including recurrent gene fusions involving R-spondin family members RSPO2 and RSPO3 that together occur in 10% of colon tumours. The RSPO fusions were mutually exclusive with APC mutations, indicating that they probably have a role in the activation of Wnt signalling and tumorigenesis. Consistent with this we show that the RSPO fusion proteins were capable of potentiating Wnt signalling. The R-spondin gene fusions and several other gene mutations identified in this study provide new potential opportunities for therapeutic intervention in colon cancer.


Subject(s)
Colonic Neoplasms/genetics , Gene Fusion/genetics , Genes, Neoplasm/genetics , Intercellular Signaling Peptides and Proteins/genetics , Thrombospondins/genetics , Ataxia Telangiectasia Mutated Proteins , Base Sequence , Cell Cycle Proteins/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Copy Number Variations/genetics , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Exome/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genes, APC , Humans , Insulin-Like Growth Factor II/genetics , Molecular Sequence Data , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor, ErbB-3/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Transcription Factor 7-Like 2 Protein/genetics , Tumor Suppressor Proteins/genetics , Wnt Proteins/metabolism
13.
Mol Biol Evol ; 24(12): 2598-609, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17602168

ABSTRACT

Repeated motifs of amino acids within proteins are an abundant feature of eukaryotic sequences and may catalyze the rapid production of genetic and even phenotypic variation among organisms. The completion of the genome sequencing projects of 12 distinct Drosophila species provides a unique dataset to study these intriguing sequence features on a phylogeny with a variety of timescales. We show that there is a higher percentage of proteins containing repeats within the Drosophila genus than most other eukaryotes, including non-Drosphila insects, which makes this collection of species particularly useful for the study of protein repeats. We also find that proteins containing repeats are overrepresented in functional categories involving developmental processes, signaling, and gene regulation. Using the set of 1-to-1 ortholog alignments for the 12 Drosophila species, we test the ability of repeats to act as reliable phylogenetic signals and find that they resolve the generally accepted phylogeny despite the noise caused by their accelerated rate of evolution. We also determine that in general the position of repeats within a protein sequence is non-random, with repeats more often being absent from the middle regions of sequences. Finally we find evidence to suggest that the presence of repeats is associated with an increase in evolutionary rate upon the entire sequence in which they are embedded. With additional evidence to suggest a corresponding elevation in positive selection we propose that some repeats may be inducing compensatory substitutions in their surrounding sequence.


Subject(s)
Drosophila/classification , Drosophila/genetics , Evolution, Molecular , Genome, Insect/genetics , Repetitive Sequences, Amino Acid , Amino Acid Sequence , Animals , Base Sequence , Chi-Square Distribution , Insect Proteins/chemistry , Molecular Sequence Data , Phylogeny , Selection, Genetic , Sequence Alignment , Species Specificity , Trinucleotide Repeats/genetics
14.
Mol Biol Evol ; 23(11): 2017-25, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16877497

ABSTRACT

Highly repetitive sequence within proteins is an abundant feature yet is considered by some to be the protein equivalent of "junk DNA." Homopolymer sequences, the most highly repetitive of this group, are typically encoded by trinucleotide repeats at the DNA level. It is thought that many of these sequences are produced by a replicative slippage mechanism. Recent studies suggest that these highly mutable regions within proteins may allow for rapid morphological evolution emerging from the increased variability afforded by such coding structures. However, in a homopolymer, it is difficult to determine if the repeated amino acid is due to slippage at the DNA level or due to selection at the protein level. Here we develop and test a model to detect cases for which the homopolymer tract has clearly been selected for, with no evidence of slippage at the DNA level. The polyserine tract within the phosphatidylserine receptor protein is used as an excellent example of one such case.


Subject(s)
Evolution, Molecular , Models, Genetic , Phylogeny , Repetitive Sequences, Amino Acid , Selection, Genetic , Amino Acid Sequence , Base Sequence , Conserved Sequence , Humans , Molecular Sequence Data , Polymers , Sequence Homology, Amino Acid , Serine/chemistry
15.
J Mol Evol ; 62(6): 693-700, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16612543

ABSTRACT

A genomic comparison of Drosophila melanogaster and Drosophila pseudoobscura provides a unique opportunity to investigate factors involved in sequence divergence. The chromosomal arrangements of these species include an autosomal segment in D. melanogaster which is homologous to part of the X chromosome in D. pseudoobscura. Using orthologues to calculate rates of nonsynonymous (d(N)) substitutions, we found genes on the X chromosome to be significantly more diverged than those on the autosomes, but it is not true for segment 3L-XR which is autosomal in D. melanogaster (3L) and X-linked in D. pseudoobscura (XR). We also found that the median d(N) values for genes having reproductive functions in either the male, the female, or both sexes are higher than those for sequences without reproductive function and even higher for sequences involved in male-specific function. These estimates of divergence for male sex-related sequences are most likely underestimates, as the very rapidly evolving reproductive genes would tend to lose homology sooner and thus not be included in the comparison of orthologues. We also noticed a high proportion of male reproductive genes among the othologous genes with the highest rates of d(N). Reproductive genes with and without an orthologue in D. pseudoobscura were compared among D. melanogaster, D. simulans, and D. yakuba and it was found that there were in fact higher rates of divergence in the group without a D. pseudoobscura orthologue. These results, from widely separated taxa, bolster the thesis that sexual system genes experience accelerated rates of change in comparison to nonsexual genes in evolution and speciation.


Subject(s)
Drosophila/genetics , Evolution, Molecular , Genes, X-Linked/genetics , Genome , Recombination, Genetic , Animals , Drosophila melanogaster/genetics , Female , Male , Reproduction/genetics , Sex Differentiation/genetics , Time Factors
16.
Genome ; 48(2): 291-301, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15838552

ABSTRACT

We examined sequences expressed in the brain and nervous system using EST data. A previous study including sequences thought to have neurological function found a deficiency of simple sequence within such sequences. This was despite many examples of neurodegenerative diseases, such as Huntington disease, which are thought to be caused by expansions of polyglutamine tracts within associated protein sequences. It may be that many of the sequences thought to have neurological function have other additional, non-neurological roles. For this reason, we examined sequences with specific expression in the brain and nervous system, using EST expression data to determine if they too are deficient of simple, repetitive sequences. Indeed, we find this class of sequences to be deficient. Unexpectedly, however, we find sequences expressed in the brain and nervous system to be consistently enriched for histidine-enriched simple sequence. Determining the function of these histidine-rich regions within brain-specific proteins requires more experimental data.


Subject(s)
Brain/metabolism , Expressed Sequence Tags , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nervous System/metabolism , Amino Acid Sequence , Animals , Computational Biology , Databases, Protein , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Xenopus laevis , Zebrafish
17.
Genetics ; 166(3): 1141-54, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15082536

ABSTRACT

Proteins associated with disease and development of the nervous system are thought to contain repetitive, simple sequences. However, genome-wide surveys for simple sequences within proteins have revealed that repetitive peptide sequences are the most frequent shared peptide segments among eukaryotic proteins, including those of Saccharomyces cerevisiae, which has few to no specialized developmental and neurological proteins. It is therefore of interest to determine if these specialized proteins have an excess of simple sequences when compared to other sets of compositionally similar proteins. We have determined the relative abundance of simple sequences within neurological proteins and find no excess of repetitive simple sequence within this class. In fact, polyglutamine repeats that are associated with many neurodegenerative diseases are no more abundant within neurological specialized proteins than within nonneurological collections of proteins. We also examined the codon composition of serine homopolymers to determine what forces may play a role in the evolution of extended homopolymers. Codon type homogeneity tends to be favored, suggesting replicative slippage instead of selection as the main force responsible for producing these homopolymers.


Subject(s)
Nerve Tissue/chemistry , Proteins/chemistry , Repetitive Sequences, Amino Acid/genetics , Alanine/chemistry , Algorithms , Animals , Codon , Conserved Sequence , Databases, Factual , Drosophila Proteins/chemistry , Eukaryotic Cells , Evolution, Molecular , Genome , Histidine/chemistry , Humans , Peptides/chemistry , Phosphotransferases/chemistry , Sequence Homology, Amino Acid , Serine/chemistry , Tandem Repeat Sequences
18.
Proteins ; 48(1): 134-40, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12012345

ABSTRACT

A simple sequence is abundant in the proteins that have been sequenced to date. But unusual protein features, such as a simple sequence, are not present in the same high frequency within structural databases. A subset of these simple sequences, a group with a highly repetitive nature has been shown to be abundant in eukaryotes but not in prokaryotes. In this study, an examination of the eukaryotic proteins in the Protein Data Bank (PDB) has revealed a large deficiency of low complexity, highly repetitive protein repeats. Through simulated databases of similar samples of eukaryotic proteins taken from the National Center for Biotechnology Information (NCBI) database, it is shown that the PDB contains a significantly less highly repetitive, simple sequence than artificial databases of similar composition randomly derived from NCBI. When the structural data for those few PDB sequences that did contain a highly repetitive simple sequence is examined in detail, it is found that in most cases the tertiary structure is unknown for the regions consisting of a simple sequence. This lack of a simple sequence both in the PDB database and in the structural information suggests that this type of simple sequence may produce disordered structures that make structural characterization difficult.


Subject(s)
Databases, Protein , Proteins/chemistry , Amino Acid Sequence , Animals , Models, Molecular , Molecular Sequence Data , Repetitive Sequences, Amino Acid , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...