Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Nucleic Acids Res ; 50(D1): D1010-D1015, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34718735

ABSTRACT

FlyAtlas 2 (flyatlas2.org) is a database and web application for studying the expression of the genes of Drosophila melanogaster in different tissues of adults and larvae. It is based on RNA-Seq data, and incorporates both genes encoding proteins and microRNAs. We have now completed the population of the database with 13 tissues from both male and female adults, five sex-specific tissues, and eight larval tissues. Larval garland cell nephrocytes have also been included. Major enhancements have been made to the application. First, a facility has been added for a 'Profile' search for genes with a similar pattern of tissue expression as a query gene. This may help establish the function of genes for which this is currently unknown. Second, a facility has been added dedicated to the larval midgut, where the difference in gene expression in the five regions of different pH can be explored. A variety of further improvements to the interface are described.


Subject(s)
Databases, Genetic , Drosophila melanogaster/genetics , Gene Expression Regulation/genetics , Software , Animals , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila melanogaster/classification , Female , Larva/genetics , Larva/growth & development , Male , MicroRNAs/classification , MicroRNAs/genetics
2.
Cell Mol Life Sci ; 78(9): 4335-4364, 2021 May.
Article in English | MEDLINE | ID: mdl-33619613

ABSTRACT

The Synaptotagmin (SYT) family of proteins play key roles in regulating membrane trafficking at neuronal synapses. Using both Ca2+-dependent and Ca2+-independent interactions, several SYT isoforms participate in synchronous and asynchronous fusion of synaptic vesicles (SVs) while preventing spontaneous release that occurs in the absence of stimulation. Changes in the function or abundance of the SYT1 and SYT7 isoforms alter the number and route by which SVs fuse at nerve terminals. Several SYT family members also regulate trafficking of other subcellular organelles at synapses, including dense core vesicles (DCV), exosomes, and postsynaptic vesicles. Although SYTs are linked to trafficking of multiple classes of synaptic membrane compartments, how and when they interact with lipids, the SNARE machinery and other release effectors are still being elucidated. Given mutations in the SYT family cause disorders in both the central and peripheral nervous system in humans, ongoing efforts are defining how these proteins regulate vesicle trafficking within distinct neuronal compartments. Here, we review the Drosophila SYT family and examine their role in synaptic communication. Studies in this invertebrate model have revealed key similarities and several differences with the predicted activity of their mammalian counterparts. In addition, we highlight the remaining areas of uncertainty in the field and describe outstanding questions on how the SYT family regulates membrane trafficking at nerve terminals.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Synaptotagmins/metabolism , Animals , Calcium/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Exocytosis , Humans , Neurotransmitter Agents/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Synaptic Vesicles/metabolism , Synaptotagmins/chemistry , Synaptotagmins/classification
3.
J Biol Chem ; 295(12): 3794-3807, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32024695

ABSTRACT

The troponin complex regulates the Ca2+ activation of myofilaments during striated muscle contraction and relaxation. Troponin genes emerged 500-700 million years ago during early animal evolution. Troponin T (TnT) is the thin-filament-anchoring subunit of troponin. Vertebrate and invertebrate TnTs have conserved core structures, reflecting conserved functions in regulating muscle contraction, and they also contain significantly diverged structures, reflecting muscle type- and species-specific adaptations. TnT in insects contains a highly-diverged structure consisting of a long glutamic acid-rich C-terminal extension of ∼70 residues with unknown function. We found here that C-terminally truncated Drosophila TnT (TpnT-CD70) retains binding of tropomyosin, troponin I, and troponin C, indicating a preserved core structure of TnT. However, the mutant TpnTCD70 gene residing on the X chromosome resulted in lethality in male flies. We demonstrate that this X-linked mutation produces dominant-negative phenotypes, including decreased flying and climbing abilities, in heterozygous female flies. Immunoblot quantification with a TpnT-specific mAb indicated expression of TpnT-CD70 in vivo and normal stoichiometry of total TnT in myofilaments of heterozygous female flies. Light and EM examinations revealed primarily normal sarcomere structures in female heterozygous animals, whereas Z-band streaming could be observed in the jump muscle of these flies. Although TpnT-CD70-expressing flies exhibited lower resistance to cardiac stress, their hearts were significantly more tolerant to Ca2+ overloading induced by high-frequency electrical pacing. Our findings suggest that the Glu-rich long C-terminal extension of insect TnT functions as a myofilament Ca2+ buffer/reservoir and is potentially critical to the high-frequency asynchronous contraction of flight muscles.


Subject(s)
Drosophila Proteins/metabolism , Glutamic Acid/metabolism , Muscle, Skeletal/metabolism , Troponin T/metabolism , Alternative Splicing , Animals , CD27 Ligand/chemistry , CD27 Ligand/metabolism , Calcium/metabolism , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Drosophila Proteins/genetics , Female , Flight, Animal , Male , Muscle Contraction , Mutagenesis , Myofibrils/metabolism , Phylogeny , Protein Domains , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Tropomyosin/chemistry , Tropomyosin/metabolism , Troponin T/chemistry , Troponin T/classification , Troponin T/genetics , X Chromosome
4.
Mol Phylogenet Evol ; 158: 107061, 2021 05.
Article in English | MEDLINE | ID: mdl-33387647

ABSTRACT

The Drosophila montium species group is a clade of 94 named species, closely related to the model species D. melanogaster. The montium species group is distributed over a broad geographic range throughout Asia, Africa, and Australasia. Species of this group possess a wide range of morphologies, mating behaviors, and endosymbiont associations, making this clade useful for comparative analyses. We use genomic data from 42 available species to estimate the phylogeny and relative divergence times within the montium species group, and its relative divergence time from D. melanogaster. To assess the robustness of our phylogenetic inferences, we use 3 non-overlapping sets of 20 single-copy coding sequences and analyze all 60 genes with both Bayesian and maximum likelihood methods. Our analyses support monophyly of the group. Apart from the uncertain placement of a single species, D. baimaii, our analyses also support the monophyly of all seven subgroups proposed within the montium group. Our phylograms and relative chronograms provide a highly resolved species tree, with discordance restricted to estimates of relatively short branches deep in the tree. In contrast, age estimates for the montium crown group, relative to its divergence from D. melanogaster, depend critically on prior assumptions concerning variation in rates of molecular evolution across branches, and hence have not been reliably determined. We discuss methodological issues that limit phylogenetic resolution - even when complete genome sequences are available - as well as the utility of the current phylogeny for understanding the evolutionary and biogeographic history of this clade.


Subject(s)
Drosophila/classification , Animals , Bayes Theorem , DNA/chemistry , DNA/isolation & purification , DNA/metabolism , Drosophila/genetics , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila melanogaster/classification , Drosophila melanogaster/genetics , Evolution, Molecular , Phylogeny , Sequence Analysis, DNA
5.
Biochem Biophys Res Commun ; 523(2): 322-327, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31864711

ABSTRACT

While many instances of GPCR dimerization have been reported for vertebrate receptors, invertebrate GPCR dimerization remains poorly investigated, with few invertebrate GPCRs having been shown to assemble as dimers. To date, no Drosophila GPCRs have been shown to assemble as dimers. To explore the evolutionary conservation of GPCR dimerization, we employed an acceptor-photobleaching FRET methodology to evaluate whether multiple subclasses of Drosophila GPCRs assembled as homodimers when heterologously expressed in HEK-293 T cells. We C-terminally tagged multiple Drosophila neuropeptide GPCRs that exhibited structural homology with a vertebrate GPCR family member previously shown to assemble as a dimer with CFP and YFP fluorophores and visualized these receptors through confocal microscopy. FRET responses were determined based on the increase in CFP emission intensity following YFP photobleaching for each receptor pair tested. A significant FRET response was observed for each receptor expressed as a homodimer pair, while non-significant FRET responses were displayed by both cytosolic CFP and YFP expressed alone, and a heterodimeric pair of receptors from unrelated families. These findings suggest that receptors exhibiting positive FRET responses assemble as homodimers at the plasma membrane and are the first to suggest that Drosophila GPCRs assemble as homodimeric complexes. We propose that GPCR dimerization arose early in metazoan evolution and likely plays an important and underappreciated role in the cellular signaling of all animals.


Subject(s)
Drosophila Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, Neuropeptide/chemistry , Animals , Cell Membrane/metabolism , Dimerization , Drosophila Proteins/classification , Drosophila Proteins/genetics , Evolution, Molecular , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Neuropeptides/metabolism , Photobleaching , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/classification , Receptors, Neuropeptide/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
Cell Mol Life Sci ; 76(4): 667-680, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30343321

ABSTRACT

The complexity of neuronal wiring relies on the extraordinary recognition diversity of cell surface molecules. Drosophila Dscam1 and vertebrate clustered protocadherins (Pcdhs) are two classic examples of the striking diversity from a complex genomic locus, wherein the former encodes more than 10,000 distinct isoforms via alternative splicing, while the latter employs alternative promoters to attain isoform diversity. These structurally unrelated families show remarkably striking molecular parallels and even similar functions. Recent studies revealed a novel Dscam gene family with tandemly arrayed 5' cassettes in Chelicerata (e.g., the scorpion Mesobuthus martensii and the tick Ixodes scapularis), similar to vertebrate clustered Pcdhs. Likewise, octopus shows a more remarkable expansion of the Pcdh isoform repertoire than human. These discoveries of Dscam and Pcdh diversification reshape the evolutionary landscape of recognition molecule diversity and provide a greater understanding of convergent molecular strategies for isoform diversity. This article reviews new insights into the evolution, regulatory mechanisms, and functions of Dscam and Pcdh isoform diversity. In particular, the convergence of clustered Dscams and Pcdhs is highlighted.


Subject(s)
Alternative Splicing , Cadherins/genetics , Cell Adhesion Molecules/genetics , Drosophila Proteins/genetics , Drosophila/genetics , Animals , Cadherins/classification , Cadherins/metabolism , Cell Adhesion Molecules/classification , Cell Adhesion Molecules/metabolism , Drosophila/metabolism , Drosophila Proteins/classification , Drosophila Proteins/metabolism , Evolution, Molecular , Humans , Neurons/metabolism , Phylogeny , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism
7.
Genomics ; 111(4): 549-559, 2019 07.
Article in English | MEDLINE | ID: mdl-29545002

ABSTRACT

This article introduces an alignment-free clustering method in order to cluster all the 66 DORs sequentially diverse protein sequences. Two different methods are discussed: one is utilizing twenty standard amino acids (without grouping) and another one is using chemical grouping of amino acids (with grouping). Two grayscale images (representing two protein sequences by order pair frequency matrices) are compared to find the similarity index using morphology technique. We could achieve the correlation coefficients of 0.9734 and 0.9403 for without and with grouping methods respectively with the ClustalW result in the ND5 dataset, which are much better than some of the existing alignment-free methods. Based on the similarity index, the 66 DORs are clustered into three classes - Highest, Moderate and Lowest - which are seen to be best fitted for 66 DORs protein sequences. OR83b is the distinguished olfactory receptor expressed in divergent insect population which is substantiated through our investigation.


Subject(s)
Drosophila Proteins/chemistry , Receptors, Odorant/chemistry , Sequence Alignment/methods , Animals , Cluster Analysis , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila melanogaster , Phylogeny , Receptors, Odorant/classification , Receptors, Odorant/genetics
8.
Proc Natl Acad Sci U S A ; 112(40): 12450-5, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26385968

ABSTRACT

Contrary to the pattern seen in mammalian sex chromosomes, where most Y-linked genes have X-linked homologs, the Drosophila X and Y chromosomes appear to be unrelated. Most of the Y-linked genes have autosomal paralogs, so autosome-to-Y transposition must be the main source of Drosophila Y-linked genes. Here we show how these genes were acquired. We found a previously unidentified gene (flagrante delicto Y, FDY) that originated from a recent duplication of the autosomal gene vig2 to the Y chromosome of Drosophila melanogaster. Four contiguous genes were duplicated along with vig2, but they became pseudogenes through the accumulation of deletions and transposable element insertions, whereas FDY remained functional, acquired testis-specific expression, and now accounts for ∼20% of the vig2-like mRNA in testis. FDY is absent in the closest relatives of D. melanogaster, and DNA sequence divergence indicates that the duplication to the Y chromosome occurred ∼2 million years ago. Thus, FDY provides a snapshot of the early stages of the establishment of a Y-linked gene and demonstrates how the Drosophila Y has been accumulating autosomal genes.


Subject(s)
Drosophila melanogaster/genetics , Genes, Insect/genetics , Genes, Y-Linked/genetics , Y Chromosome/genetics , Animals , Chromosome Mapping , Chromosomes, Insect/genetics , Drosophila Proteins/classification , Drosophila Proteins/genetics , Female , Gene Duplication , Gene Expression , INDEL Mutation , Male , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , X Chromosome/genetics
9.
Cell Mol Life Sci ; 73(9): 1939-54, 2016 May.
Article in English | MEDLINE | ID: mdl-26715182

ABSTRACT

Ubiquitin-related modifier 1 (Urm1) is a ubiquitin-like molecule (UBL) with the dual capacity to act both as a sulphur carrier and posttranslational protein modifier. Here we characterize the Drosophila melanogaster homologues of Urm1 (CG33276) and its E1 activating enzyme Uba4 (CG13090), and show that they function together to induce protein urmylation in vivo. Urm1 conjugation to target proteins in general, and to the evolutionary conserved substrate Peroxiredoxin 5 (Prx5) specifically, is dependent on Uba4. A complete loss of Urm1 is lethal in flies, although a small number of adult zygotic Urm1 (n123) mutant escapers can be recovered. These escapers display a decreased general fitness and shortened lifespan, but in contrast to their S. cerevisiae counterparts, they are resistant to oxidative stress. Providing a molecular explanation, we demonstrate that cytoprotective JNK signaling is increased in Urm1 deficient animals. In agreement, molecular and genetic evidence suggest that elevated activity of the JNK downstream target genes Jafrac1 and gstD1 strongly contributes to the tolerance against oxidative stress displayed by Urm1 (n123) null mutants. In conclusion, Urm1 is a UBL that is involved in the regulation of JNK signaling and the response against oxidative stress in the fruit fly.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified/metabolism , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Drosophila melanogaster/growth & development , Larva/metabolism , Longevity , MAP Kinase Signaling System , Molecular Sequence Data , Mutagenesis , Nucleotidyltransferases/classification , Nucleotidyltransferases/metabolism , Oxidative Stress , Paraquat/toxicity , Peroxidases/genetics , Peroxidases/metabolism , Phylogeny , Sequence Alignment , Ubiquitin/classification , Ubiquitin/genetics
10.
Biochem J ; 473(13): 1905-16, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27114558

ABSTRACT

CMP-sialic acid synthetase (CSAS) is a key enzyme of the sialylation pathway. CSAS produces the activated sugar donor, CMP-sialic acid, which serves as a substrate for sialyltransferases to modify glycan termini with sialic acid. Unlike other animal CSASs that normally localize in the nucleus, Drosophila melanogaster CSAS (DmCSAS) localizes in the cell secretory compartment, predominantly in the Golgi, which suggests that this enzyme has properties distinct from those of its vertebrate counterparts. To test this hypothesis, we purified recombinant DmCSAS and characterized its activity in vitro Our experiments revealed several unique features of this enzyme. DmCSAS displays specificity for N-acetylneuraminic acid as a substrate, shows preference for lower pH and can function with a broad range of metal cofactors. When tested at a pH corresponding to the Golgi compartment, the enzyme showed significant activity with several metal cations, including Zn(2+), Fe(2+), Co(2+) and Mn(2+), whereas the activity with Mg(2+) was found to be low. Protein sequence analysis and site-specific mutagenesis identified an aspartic acid residue that is necessary for enzymatic activity and predicted to be involved in co-ordinating a metal cofactor. DmCSAS enzymatic activity was found to be essential in vivo for rescuing the phenotype of DmCSAS mutants. Finally, our experiments revealed a steep dependence of the enzymatic activity on temperature. Taken together, our results indicate that DmCSAS underwent evolutionary adaptation to pH and ionic environment different from that of counterpart synthetases in vertebrates. Our data also suggest that environmental temperatures can regulate Drosophila sialylation, thus modulating neural transmission.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Amino Acid Sequence , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Cells, Cultured , Drosophila , Drosophila Proteins/classification , Drosophila Proteins/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Magnesium/metabolism , Manganese/metabolism , Mutation , N-Acylneuraminate Cytidylyltransferase/classification , N-Acylneuraminate Cytidylyltransferase/genetics , Phylogeny , Structure-Activity Relationship , Substrate Specificity , Temperature
11.
Genome Res ; 23(6): 928-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23471540

ABSTRACT

Cys2-His2 zinc finger proteins (ZFPs) are the largest group of transcription factors in higher metazoans. A complete characterization of these ZFPs and their associated target sequences is pivotal to fully annotate transcriptional regulatory networks in metazoan genomes. As a first step in this process, we have characterized the DNA-binding specificities of 129 zinc finger sets from Drosophila using a bacterial one-hybrid system. This data set contains the DNA-binding specificities for at least one encoded ZFP from 70 unique genes and 23 alternate splice isoforms representing the largest set of characterized ZFPs from any organism described to date. These recognition motifs can be used to predict genomic binding sites for these factors within the fruit fly genome. Subsets of fingers from these ZFPs were characterized to define their orientation and register on their recognition sequences, thereby allowing us to define the recognition diversity within this finger set. We find that the characterized fingers can specify 47 of the 64 possible DNA triplets. To confirm the utility of our finger recognition models, we employed subsets of Drosophila fingers in combination with an existing archive of artificial zinc finger modules to create ZFPs with novel DNA-binding specificity. These hybrids of natural and artificial fingers can be used to create functional zinc finger nucleases for editing vertebrate genomes.


Subject(s)
Binding Sites , Drosophila Proteins/genetics , Drosophila/genetics , Nucleotide Motifs , Zinc Fingers/genetics , Alternative Splicing , Animals , Base Sequence , Cluster Analysis , Computational Biology/methods , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Models, Molecular , Phylogeny , Position-Specific Scoring Matrices , Protein Binding , Protein Conformation
12.
Bioinformatics ; 31(23): 3835-7, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26243019

ABSTRACT

UNLABELLED: The recent explosion of comparative genomics data presents an unprecedented opportunity to construct gene networks via the evolutionary rate covariation (ERC) signature. ERC is used to identify genes that experienced similar evolutionary histories, and thereby draws functional associations between them. The ERC Analysis website allows researchers to exploit genome-wide datasets to infer novel genes in any biological function and to explore deep evolutionary connections between distinct pathways and complexes. The website provides five analytical methods, graphical output, statistical support and access to an increasing number of taxonomic groups. AVAILABILITY AND IMPLEMENTATION: Analyses and data at http://csb.pitt.edu/erc_analysis/ CONTACT: nclark@pitt.edu.


Subject(s)
Drosophila Proteins/genetics , Evolution, Molecular , Fungal Proteins/genetics , Gene Regulatory Networks , Genomics/methods , Internet , Metabolic Networks and Pathways , Animals , Drosophila Proteins/classification , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Fungal Proteins/classification , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Genome , Phylogeny , Species Specificity , Yeasts/genetics
13.
PLoS Biol ; 10(11): e1001420, 2012.
Article in English | MEDLINE | ID: mdl-23139640

ABSTRACT

Changes in the physical interaction between cis-regulatory DNA sequences and proteins drive the evolution of gene expression. However, it has proven difficult to accurately quantify evolutionary rates of such binding change or to estimate the relative effects of selection and drift in shaping the binding evolution. Here we examine the genome-wide binding of CTCF in four species of Drosophila separated by between ∼2.5 and 25 million years. CTCF is a highly conserved protein known to be associated with insulator sequences in the genomes of human and Drosophila. Although the binding preference for CTCF is highly conserved, we find that CTCF binding itself is highly evolutionarily dynamic and has adaptively evolved. Between species, binding divergence increased linearly with evolutionary distance, and CTCF binding profiles are diverging rapidly at the rate of 2.22% per million years (Myr). At least 89 new CTCF binding sites have originated in the Drosophila melanogaster genome since the most recent common ancestor with Drosophila simulans. Comparing these data to genome sequence data from 37 different strains of Drosophila melanogaster, we detected signatures of selection in both newly gained and evolutionarily conserved binding sites. Newly evolved CTCF binding sites show a significantly stronger signature for positive selection than older sites. Comparative gene expression profiling revealed that expression divergence of genes adjacent to CTCF binding site is significantly associated with the gain and loss of CTCF binding. Further, the birth of new genes is associated with the birth of new CTCF binding sites. Our data indicate that binding of Drosophila CTCF protein has evolved under natural selection, and CTCF binding evolution has shaped both the evolution of gene expression and genome evolution during the birth of new genes.


Subject(s)
Adaptation, Biological , Drosophila Proteins/genetics , Drosophila/genetics , Evolution, Molecular , Genome, Insect , Repressor Proteins/genetics , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Conserved Sequence , Drosophila/chemistry , Drosophila/classification , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Genetic Drift , Genetics, Population/methods , Oligonucleotide Array Sequence Analysis , Phylogeny , Protein Binding , Repressor Proteins/chemistry , Repressor Proteins/classification , Selection, Genetic , Species Specificity , Time Factors
14.
PLoS Genet ; 8(5): e1002667, 2012.
Article in English | MEDLINE | ID: mdl-22589731

ABSTRACT

The pairing of homologous chromosomes is a fundamental feature of the meiotic cell. In addition, a number of species exhibit homolog pairing in nonmeiotic, somatic cells as well, with evidence for its impact on both gene regulation and double-strand break (DSB) repair. An extreme example of somatic pairing can be observed in Drosophila melanogaster, where homologous chromosomes remain aligned throughout most of development. However, our understanding of the mechanism of somatic homolog pairing remains unclear, as only a few genes have been implicated in this process. In this study, we introduce a novel high-throughput fluorescent in situ hybridization (FISH) technology that enabled us to conduct a genome-wide RNAi screen for factors involved in the robust somatic pairing observed in Drosophila. We identified both candidate "pairing promoting genes" and candidate "anti-pairing genes," providing evidence that pairing is a dynamic process that can be both enhanced and antagonized. Many of the genes found to be important for promoting pairing are highly enriched for functions associated with mitotic cell division, suggesting a genetic framework for a long-standing link between chromosome dynamics during mitosis and nuclear organization during interphase. In contrast, several of the candidate anti-pairing genes have known interphase functions associated with S-phase progression, DNA replication, and chromatin compaction, including several components of the condensin II complex. In combination with a variety of secondary assays, these results provide insights into the mechanism and dynamics of somatic pairing.


Subject(s)
Chromosome Pairing/genetics , Drosophila Proteins , Drosophila melanogaster , Heterochromatin/genetics , Meiosis , RNA Interference , Anaphase-Promoting Complex-Cyclosome , Aneuploidy , Animals , Cell Culture Techniques , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , In Situ Hybridization, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitosis , Recombination, Genetic , Ubiquitin-Protein Ligase Complexes/genetics , Cohesins
15.
Gen Comp Endocrinol ; 209: 162-70, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25230393

ABSTRACT

The corticotropin releasing hormone receptors (CRHR) and the arthropod diuretic hormone 44 receptors (DH44R) are structurally and functionally related members of the G protein-coupled receptors (GPCR) of the secretin-like receptor superfamily. We show here that they derive from a bilaterian predecessor. In protostomes, the receptor became DH44R that has been identified and functionally characterised in several arthropods but the gene seems to be absent from nematode genomes. Duplicate DH44R genes (DH44 R1 and DH44R2) have been described in some arthropods resulting from lineage-specific duplications. Recently, CRHR-DH44R-like receptors have been identified in the genomes of some lophotrochozoans (molluscs, which have a lineage-specific gene duplication, and annelids) as well as representatives of early diverging deuterostomes. Vertebrates have previously been reported to have two CRHR receptors that were named CRHR1 and CRHR2. To resolve their origin we have analysed recently assembled genomes from representatives of early vertebrate divergencies including elephant shark, spotted gar and coelacanth. We show here by analysis of synteny conservation that the two CRHR genes arose from a common ancestral gene in the early vertebrate tetraploidizations (2R) approximately 500 million years ago. Subsequently, the teleost-specific tetraploidization (3R) resulted in a duplicate of CRHR1 that has been lost in some teleost lineages. These results help distinguish orthology and paralogy relationships and will allow studies of functional conservation and changes during evolution of the individual members of the receptor family and their multiple native peptide agonists.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Drosophila Proteins/genetics , Evolution, Molecular , Invertebrates/genetics , Receptors, Cell Surface/genetics , Vertebrates/genetics , Animals , Conserved Sequence , Corticotropin-Releasing Hormone/classification , Corticotropin-Releasing Hormone/metabolism , Drosophila Proteins/classification , Drosophila Proteins/metabolism , Humans , Insect Hormones/genetics , Insect Hormones/metabolism , Invertebrates/metabolism , Phylogeny , Receptors, Cell Surface/classification , Receptors, Cell Surface/metabolism , Vertebrates/metabolism
16.
Dev Biol ; 363(1): 266-78, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22245052

ABSTRACT

As the complexity of animal nervous systems has increased during evolution, developmental control of neuronal connectivity has become increasingly refined. How has functional diversification within related axon guidance molecules contributed to the evolution of nervous systems? To address this question, we explore the evolution of functional diversity within the Roundabout (Robo) family of axon guidance receptors. In Drosophila, Robo and Robo2 promote midline repulsion, while Robo2 and Robo3 specify the position of longitudinal axon pathways. The Robo family has expanded by gene duplication in insects; robo2 and robo3 exist as distinct genes only within dipterans, while other insects, like the flour beetle Tribolium castaneum, retain an ancestral robo2/3 gene. Both Robos from Tribolium can mediate midline repulsion in Drosophila, but unlike the fly Robos cannot be down-regulated by Commissureless. The overall architecture and arrangement of longitudinal pathways are remarkably conserved in Tribolium, despite it having only two Robos. Loss of TcSlit causes midline collapse of axons in the beetle, a phenotype recapitulated by simultaneous knockdown of both Robos. Single gene knockdowns reveal that beetle Robos have specialized axon guidance functions: TcRobo is dedicated to midline repulsion, while TcRobo2/3 also regulates longitudinal pathway formation. TcRobo2/3 knockdown reproduces aspects of both Drosophila robo2 and robo3 mutants, suggesting that TcRobo2/3 has two functions that in Drosophila are divided between Robo2 and Robo3. The ability of Tribolium to organize longitudinal axons into three discrete medial-lateral zones with only two Robo receptors demonstrates that beetle and fly achieve equivalent developmental outcomes using divergent genetic programs.


Subject(s)
Axons/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Insect Proteins/genetics , Tribolium/genetics , Amino Acid Sequence , Animals , Axons/physiology , Drosophila Proteins/classification , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Evolution, Molecular , Female , Gene Expression Regulation, Developmental , Genetic Variation , Immunohistochemistry , Insect Proteins/classification , Insect Proteins/metabolism , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Nervous System/metabolism , Phylogeny , RNA Interference , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction/genetics , Tribolium/embryology , Tribolium/metabolism , Roundabout Proteins
17.
J Mol Evol ; 76(5): 311-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23619741

ABSTRACT

Drosophila nasuta nasuta (2n = 8) and D. n. albomicans (2n = 6) are morphologically identical, cross fertile and karyotypically dissimilar pair of chromosomal races belonging to nasuta subgroup of immigrans group of Drosophila. Interracial hybridization between these two races yielded karyotypically stabilized newly evolved Cytoraces with new combinations of chromosomes and DNA content, and are called nasuta-albomicans complex of Drosophila. Along with many other features, striking plasticity in the lifespan has been observed in the karyotypically stabilized members of nasuta-albomicans complex of Drosophila. These findings provide a strong background to understand any changes at the molecular levels. In view of this, we cloned and characterized Sod1 and Rpd3 in the members of nasuta-albomicans complex of Drosophila. The evolution of Sod1 and Rpd3 in D. n. nasuta and D. n. albomicans is contrasting with the other species of Drosophila, at the level of synonymous mutations, intron variation, InDels and secondary structure changes in protein. In the members of NAC of Drosophila there were synonymous changes, variations in intron sequences of Sod1, whereas, in Rpd3, synonymous, nonsynonymous, intron variation, and secondary structure changes in protein were observed. The contrasting differences in the levels of Rpd3 (and Sir2) proteins were also noticed among short-lived and long-lived Cytoraces. The Cytoraces have exhibited not only specific changes in Sod1 and Rpd3, but also show pronounced changes in the levels of synthesis of these proteins, which indicates rapid evolution of these Cytoraces in laboratory. Further these Cytoraces have become a model system to understand the process of anagenesis.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Evolution, Molecular , Histone Deacetylase 1/genetics , Mutation , Superoxide Dismutase/genetics , Amino Acid Sequence , Animals , Chromosomes, Insect , Drosophila/classification , Drosophila Proteins/classification , Histone Deacetylase 1/classification , Humans , Hybridization, Genetic , Introns , Karyotyping , Longevity , Male , Molecular Sequence Data , Protein Structure, Secondary , Sequence Alignment , Superoxide Dismutase/classification , Superoxide Dismutase-1
18.
Parasitol Res ; 112(10): 3527-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23873618

ABSTRACT

The Eurasian red squirrel (Sciurus vulgaris) is the only naturally occurring tree squirrel throughout its range. We aim at improving current knowledge on its macroparasite fauna, expecting that it will have a poor parasite diversity because in species that have no sympatric congeners parasite richness should be lower than in hosts sharing their range with several closely related species, where host-switching events and lateral transmission are promoted. We examined gastro-intestinal helminth and ectoparasite communities (excluding mites) of, respectively, 147 and 311 red squirrel roadkills collected in four biogeographic regions in Italy and France. As expected, the macroparasite fauna was poor: we found five species of nematodes and some unidentified cestodes, three fleas, two sucking lice and two hard ticks. The helminth community was dominated by a single species, the oxyurid Trypanoxyuris (Rodentoxyuris) sciuri (prevalence, 87%; mean abundance, 373 ± 65 worms/host). Its abundance varied among seasons and biogeographic regions and increased with body mass in male hosts while decreased in females. The most prevalent ectoparasites were the flea Ceratophyllus (Monopsyllus) sciurorum (28%), whose presence was affected by season, and the generalist tick Ixodes (Ixodes) ricinus that was found only in France (34%). All the other helminths and arthropod species were rare, with prevalence below 10%. However, the first record of Strongyloides robustus, a common nematode of North American Eastern grey squirrels (S. carolinensis), in two red squirrels living in areas where this alien species co-inhabits, deserves further attention, since low parasite richness could result in native red squirrels being particularly vulnerable to parasite spillover.


Subject(s)
Ectoparasitic Infestations/veterinary , Helminthiasis, Animal/parasitology , Helminths/classification , Sciuridae/parasitology , Animals , Drosophila Proteins/classification , Ectoparasitic Infestations/parasitology , Female , Male , Protein Kinases/classification , Siphonaptera/classification , Ticks/classification
19.
Mol Biol Evol ; 28(1): 237-47, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20651048

ABSTRACT

The FoxP gene subfamily of transcription factors is defined by its characteristic 110 amino acid long DNA-binding forkhead domain and plays essential roles in vertebrate biology. Its four members, FoxP1-P4, have been extensively characterized functionally. FoxP1, FoxP2, and FoxP4 are involved in lung, heart, gut, and central nervous system (CNS) development. FoxP3 is necessary and sufficient for the specification of regulatory T cells (Tregs) of the adaptive immune system. In Drosophila melanogaster, in silico predictions identify one unique FoxP subfamily gene member (CG16899) with no described function. We characterized this gene and established that it generates by alternative splicing two isoforms that differ in the forkhead DNA-binding domain. In D. melanogaster, both isoforms are expressed in the embryonic CNS, but in hemocytes, only isoform A is expressed, hinting to a putative modulation through alternative splicing of FoxP1 function in immunity and/or other hemocyte-dependent processes. Furthermore, we show that in vertebrates, this novel alternative splicing pattern is conserved for FoxP1. In mice, this new FoxP1 isoform is expressed in brain, liver, heart, testes, thymus, and macrophages (equivalent in function to hemocytes). This alternative splicing pattern has arisen at the base of the Bilateria, probably through exon tandem duplication. Moreover, our phylogenetic analysis suggests that in vertebrates, FoxP1 is more related to the FoxP gene ancestral form and the other three paralogues, originated through serial duplications, which only retained one of the alternative exons. Also, the newly described isoform differs from the other in amino acids critical for DNA-binding specificity. The integrity of its fold is maintained, but the molecule has lost the direct hydrogen bonding to DNA bases leading to a putatively lower specificity and possibly affinity toward DNA. With the present comparative study, through the integration of experimental and in silico studies of the FoxP gene subfamily across the animal kingdom, we establish a new model for the FoxP gene in invertebrates and for the vertebrate FoxP1 paralogue. Furthermore, we present a scenario for the structural evolution of this gene class and reveal new previously unsuspected levels of regulation for FoxP1 in the vertebrate system.


Subject(s)
Alternative Splicing , Drosophila Proteins/genetics , Evolution, Molecular , Forkhead Transcription Factors/genetics , Gene Duplication , Protein Isoforms/genetics , Repressor Proteins/genetics , Amino Acid Sequence , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/classification , Drosophila melanogaster/genetics , Exons , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/classification , Hemocytes/physiology , Humans , Mice , Models, Molecular , Molecular Sequence Data , Multigene Family , Phylogeny , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/classification , Repressor Proteins/chemistry , Repressor Proteins/classification , Sequence Alignment
20.
Trends Genet ; 25(9): 414-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19716619

ABSTRACT

Polycomb Group (PcG) proteins form an epigenetic memory system that is conserved in plants and animals and controls gene expression during development. Loss of plant PcG proteins leads to loss of organ identity and to cell overproliferation. Our understanding of plant PcG protein function has recently been advanced by the identification of additional proteins required for transcriptional repression by PcG and by the purification of an Arabidopsis PcG protein complex. These data indicate that Polycomb Repressive Complex 2 (PRC2)-like complexes in animals and plants have to associate with Plant Homeo Domain (PHD)-finger proteins for efficient deposition of histone H3 trimethylated at lysine 27 (H3K27me3) and transcriptional repression. Subsequently, H3K27me3 at target genes assist to recruit additional PcG protein complexes - PRC1 in animals and potentially LIKE HETEROCHROMATIN PROTEIN-1 (LHP1) and the RING finger gene product AtRING1 in plants. A picture is emerging in which the general mechanisms of PcG protein function are well conserved between animals and plants, but in which individual players have been exchanged during evolution.


Subject(s)
Evolution, Molecular , Genetic Variation , Phylogeny , Plant Proteins/genetics , Repressor Proteins/genetics , Animals , Arabidopsis/genetics , Arabidopsis Proteins/classification , Arabidopsis Proteins/genetics , Arabidopsis Proteins/physiology , Chromosomal Proteins, Non-Histone/genetics , Drosophila/genetics , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Plant Proteins/classification , Plant Proteins/physiology , Polycomb-Group Proteins , Repressor Proteins/classification , Repressor Proteins/physiology , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL