Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873137

ABSTRACT

Long-read sequencing is driving rapid progress in genome assembly across all major groups of life, including species of the family Drosophilidae, a longtime model system for genetics, genomics, and evolution. We previously developed a cost-effective hybrid Oxford Nanopore (ONT) long-read and Illumina short-read sequencing approach and used it to assemble 101 drosophilid genomes from laboratory cultures, greatly increasing the number of genome assemblies for this taxonomic group. The next major challenge is to address the laboratory culture bias in taxon sampling by sequencing genomes of species that cannot easily be reared in the lab. Here, we build upon our previous methods to perform amplification-free ONT sequencing of single wild flies obtained either directly from the field or from ethanol-preserved specimens in museum collections, greatly improving the representation of lesser studied drosophilid taxa in whole-genome data. Using Illumina Novaseq X Plus and ONT P2 sequencers with R10.4.1 chemistry, we set a new benchmark for inexpensive hybrid genome assembly at US $150 per genome while assembling genomes from as little as 35 ng of genomic DNA from a single fly. We present 183 new genome assemblies for 179 species as a resource for drosophilid systematics, phylogenetics, and comparative genomics. Of these genomes, 62 are from pooled lab strains and 121 from single adult flies. Despite the sample limitations of working with small insects, most single-fly diploid assemblies are comparable in contiguity (>1Mb contig N50), completeness (>98% complete dipteran BUSCOs), and accuracy (>QV40 genome-wide with ONT R10.4.1) to assemblies from inbred lines. We present a well-resolved multi-locus phylogeny for 360 drosophilid and 4 outgroup species encompassing all publicly available (as of August 2023) genomes for this group. Finally, we present a Progressive Cactus whole-genome, reference-free alignment built from a subset of 298 suitably high-quality drosophilid genomes. The new assemblies and alignment, along with updated laboratory protocols and computational pipelines, are released as an open resource and as a tool for studying evolution at the scale of an entire insect family.

2.
PLoS Biol ; 21(6): e3002148, 2023 06.
Article in English | MEDLINE | ID: mdl-37379332

ABSTRACT

To respond to the world around them, animals rely on the input of a network of sensory organs distributed throughout the body. Distinct classes of sensory organs are specialized for the detection of specific stimuli such as strain, pressure, or taste. The features that underlie this specialization relate both to the neurons that innervate sensory organs and the accessory cells they comprise. To understand the genetic basis of this diversity of cell types, both within and between sensory organs, we performed single-cell RNA sequencing on the first tarsal segment of the male Drosophila melanogaster foreleg during pupal development. This tissue displays a wide variety of functionally and structurally distinct sensory organs, including campaniform sensilla, mechanosensory bristles, and chemosensory taste bristles, as well as the sex comb, a recently evolved male-specific structure. In this study, we characterize the cellular landscape in which the sensory organs reside, identify a novel cell type that contributes to the construction of the neural lamella, and resolve the transcriptomic differences among support cells within and between sensory organs. We identify the genes that distinguish between mechanosensory and chemosensory neurons, resolve a combinatorial transcription factor code that defines 4 distinct classes of gustatory neurons and several types of mechanosensory neurons, and match the expression of sensory receptor genes to specific neuron classes. Collectively, our work identifies core genetic features of a variety of sensory organs and provides a rich, annotated resource for studying their development and function.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Male , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Sensory Receptor Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Dev Genes Evol ; 232(5-6): 89-102, 2022 12.
Article in English | MEDLINE | ID: mdl-35939093

ABSTRACT

The origin, diversification, and secondary loss of sexually dimorphic characters are common in animal evolution. In some cases, structurally and functionally similar traits have evolved independently in multiple lineages. Prominent examples of such traits include the male-specific grasping structures that develop on the front legs of many dipteran insects. In this report, we describe the evolution and development of one of these structures, the male-specific "sex brush." The sex brush is composed of densely packed, irregularly arranged modified bristles and is found in several distantly related lineages in the family Drosophilidae. Phylogenetic analysis using 250 genes from over 200 species provides modest support for a single origin of the sex brush followed by many secondary losses; however, independent origins of the sex brush cannot be ruled out completely. We show that sex brushes develop in very similar ways in all brush-bearing lineages. The dense packing of brush hairs is explained by the specification of bristle precursor cells at a near-maximum density permitted by the lateral inhibition mechanism, as well as by the reduced size of the surrounding epithelial cells. In contrast to the female and the ancestral male condition, where bristles are arranged in stereotypical, precisely spaced rows, cell migration does not contribute appreciably to the formation of the sex brush. The complex phylogenetic history of the sex brush can make it a valuable model for investigating coevolution of sex-specific morphology and mating behavior.


Subject(s)
Biological Evolution , Drosophilidae , Animals , Male , Female , Phylogeny , Drosophilidae/genetics , Drosophila melanogaster/genetics , Phenotype , Sex Characteristics
4.
Evolution ; 76(9): 2089-2104, 2022 09.
Article in English | MEDLINE | ID: mdl-35841603

ABSTRACT

Animal evolution is characterized by frequent turnover of sexually dimorphic traits-new sex-specific characters are gained, and some ancestral sex-specific characters are lost, in many lineages. In insects, sexual differentiation is predominantly cell autonomous and depends on the expression of the doublesex (dsx) transcription factor. In most cases, cells that transcribe dsx have the potential to undergo sex-specific differentiation, while those that lack dsx expression do not. Consistent with this mode of development, comparative research has shown that the origin of new sex-specific traits can be associated with the origin of new spatial domains of dsx expression. In this report, we examine the opposite situation-a secondary loss of the sex comb, a male-specific grasping structure that develops on the front legs of some drosophilid species. We show that while the origin of the sex comb is linked to an evolutionary gain of dsx expression in the leg, sex comb loss in a newly identified species of Lordiphosa (Drosophilidae) is associated with a secondary loss of dsx expression. We discuss how the developmental control of sexual dimorphism affects the mechanisms by which sex-specific traits can evolve.


Subject(s)
Drosophila Proteins , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Male , Sex Characteristics , Sex Differentiation
5.
J Comp Neurol ; 530(14): 2439-2450, 2022 10.
Article in English | MEDLINE | ID: mdl-35603778

ABSTRACT

Drosophila males use leg gustatory bristles to discriminate between male and female cuticular pheromones as an important part of courtship behavior. In Drosophila melanogaster, several male-specific gustatory bristles are present on the anterior surface of the first tarsal segment of the prothoracic leg, in addition to a larger set of gustatory bristles found in both sexes. These bristles are thought to be specialized for pheromone detection. Here, we report the number and location of sex-specific gustatory bristles in 27 other Drosophila species. Although some species have a pattern similar to D. melanogaster, others lack anterior male-specific bristles but have many dorsal male-specific gustatory bristles instead. Some species have both anterior and dorsal male-specific bristles, while others lack sexual dimorphism entirely. In several distantly related species, the number of gustatory bristles is much greater in males than in females due to a male-specific transformation of ancestrally mechanosensory bristles to a chemosensory identity. This variation in the extent and pattern of sexual dimorphism may affect the formation and function of neuronal circuits that control Drosophila courtship and contribute to the evolution of mating behavior.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Courtship , Drosophila , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Male , Pheromones , Sexual Behavior, Animal/physiology
7.
Elife ; 102021 07 19.
Article in English | MEDLINE | ID: mdl-34279216

ABSTRACT

Over 100 years of studies in Drosophila melanogaster and related species in the genus Drosophila have facilitated key discoveries in genetics, genomics, and evolution. While high-quality genome assemblies exist for several species in this group, they only encompass a small fraction of the genus. Recent advances in long-read sequencing allow high-quality genome assemblies for tens or even hundreds of species to be efficiently generated. Here, we utilize Oxford Nanopore sequencing to build an open community resource of genome assemblies for 101 lines of 93 drosophilid species encompassing 14 species groups and 35 sub-groups. The genomes are highly contiguous and complete, with an average contig N50 of 10.5 Mb and greater than 97% BUSCO completeness in 97/101 assemblies. We show that Nanopore-based assemblies are highly accurate in coding regions, particularly with respect to coding insertions and deletions. These assemblies, along with a detailed laboratory protocol and assembly pipelines, are released as a public resource and will serve as a starting point for addressing broad questions of genetics, ecology, and evolution at the scale of hundreds of species.


Subject(s)
Drosophila melanogaster/genetics , Genome Size , Genomics/methods , Animals , Cell Line , Chromosomes , Computational Biology/methods , Female , Genome , High-Throughput Nucleotide Sequencing/methods , Nanopores
8.
Development ; 146(14)2019 07 25.
Article in English | MEDLINE | ID: mdl-31285355

ABSTRACT

The ability of a single genome to produce distinct and often dramatically different male and female forms is one of the wonders of animal development. In Drosophila melanogaster, most sexually dimorphic traits are controlled by sex-specific isoforms of the doublesex (dsx) transcription factor, and dsx expression is mostly limited to cells that give rise to sexually dimorphic traits. However, it is unknown how this mosaic of sexually dimorphic and monomorphic organs arises. Here, we characterize the cis-regulatory sequences that control dsx expression in the foreleg, which contains multiple types of sex-specific sensory organs. We find that separate modular enhancers are responsible for dsx expression in each sexually dimorphic organ. Expression of dsx in the sex comb is co-regulated by two enhancers with distinct spatial and temporal specificities that are separated by a genitalia-specific enhancer. The sex comb-specific enhancer from D. willistoni, a species that primitively lacks sex combs, is not active in the foreleg. Thus, the mosaic of sexually dimorphic and monomorphic organs depends on modular regulation of dsx transcription by dedicated cell type-specific enhancers.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Enhancer Elements, Genetic/physiology , Genitalia/embryology , Genitalia/metabolism , Sex Differentiation/genetics , Animals , Animals, Genetically Modified , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Male , Organ Specificity/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Sex Characteristics
9.
PLoS Genet ; 14(4): e1007320, 2018 04.
Article in English | MEDLINE | ID: mdl-29634724

ABSTRACT

Hox genes are involved in the patterning of animal body parts at multiple levels of regulatory hierarchies. Early expression of Hox genes in different domains along the embryonic anterior-posterior (A/P) axis in insects, vertebrates, and other animals establishes segmental or regional identity. However, Hox gene function is also required later in development for the patterning and morphogenesis of limbs and other organs. In Drosophila, spatiotemporal modulation of Sex combs reduced (Scr) expression within the first thoracic (T1) leg underlies the generation of segment- and sex-specific sense organ patterns. High Scr expression in defined domains of the T1 leg is required for the development of T1-specific transverse bristle rows in both sexes and sex combs in males, implying that the patterning of segment-specific sense organs involves incorporation of Scr into the leg development and sex determination gene networks. We sought to gain insight into this process by identifying the cis-and trans-regulatory factors that direct Scr expression during leg development. We have identified two cis-regulatory elements that control spatially modulated Scr expression within T1 legs. One of these enhancers directs sexually dimorphic expression and is required for the formation of T1-specific bristle patterns. We show that the Distalless and Engrailed homeodomain transcription factors act through sequences in this enhancer to establish elevated Scr expression in spatially defined domains. This enhancer functions to integrate Scr into the intrasegmental gene regulatory network, such that Scr serves as a link between leg patterning, sex determination, and sensory organ development.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Enhancer Elements, Genetic/genetics , Homeodomain Proteins/genetics , Sense Organs/metabolism , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Extremities/growth & development , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Male , Mutation , Sense Organs/growth & development , Sex Factors , Transcription Factors/metabolism
10.
Evol Dev ; 20(2): 78-88, 2018 03.
Article in English | MEDLINE | ID: mdl-29372584

ABSTRACT

Male ornaments and other sex-specific traits present some of the most dramatic examples of evolutionary innovations. Comparative studies of similar but independently evolved traits are particularly important for identifying repeated patterns in the evolution of these traits. Male-specific modifications of the front legs have evolved repeatedly in Drosophilidae and other Diptera. The best understood of these novel structures is the sex comb of Drosophila melanogaster and its close relatives. Here, we examine the evolution of another male foreleg modification, the sex brush, found in the distantly related Drosophila immigrans species group. Similar to the sex comb, we find that the origin of the sex brush correlates with novel, spatially restricted expression of the doublesex (dsx) transcription factor, the primary effector of the Drosophila sex determination pathway. The diversity of Dsx expression patterns in the immigrans species group closely reflects the differences in the presence, position, and size of the sex brush. Together with previous work on sex comb evolution, these observations suggest that tissue-specific activation of dsx expression may be a common mechanism responsible for the evolution of sexual dimorphism and particularly for the origin of novel male-specific ornaments.


Subject(s)
Biological Evolution , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/classification , Drosophila/physiology , Gene Expression Regulation , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Extremities/physiology , Female , Male , Phylogeny , Sexual Behavior, Animal
11.
Genome Res ; 24(7): 1209-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24985915

ABSTRACT

Accurate gene model annotation of reference genomes is critical for making them useful. The modENCODE project has improved the D. melanogaster genome annotation by using deep and diverse high-throughput data. Since transcriptional activity that has been evolutionarily conserved is likely to have an advantageous function, we have performed large-scale interspecific comparisons to increase confidence in predicted annotations. To support comparative genomics, we filled in divergence gaps in the Drosophila phylogeny by generating draft genomes for eight new species. For comparative transcriptome analysis, we generated mRNA expression profiles on 81 samples from multiple tissues and developmental stages of 15 Drosophila species, and we performed cap analysis of gene expression in D. melanogaster and D. pseudoobscura. We also describe conservation of four distinct core promoter structures composed of combinations of elements at three positions. Overall, each type of genomic feature shows a characteristic divergence rate relative to neutral models, highlighting the value of multispecies alignment in annotating a target genome that should prove useful in the annotation of other high priority genomes, especially human and other mammalian genomes that are rich in noncoding sequences. We report that the vast majority of elements in the annotation are evolutionarily conserved, indicating that the annotation will be an important springboard for functional genetic testing by the Drosophila community.


Subject(s)
Computational Biology/methods , Drosophila melanogaster/genetics , Gene Expression Profiling , Molecular Sequence Annotation , Transcriptome , Animals , Cluster Analysis , Drosophila melanogaster/classification , Evolution, Molecular , Exons , Female , Genome, Insect , Humans , Male , Nucleotide Motifs , Phylogeny , Position-Specific Scoring Matrices , Promoter Regions, Genetic , RNA Editing , RNA Splice Sites , RNA Splicing , Reproducibility of Results , Transcription Initiation Site
12.
Genetics ; 192(4): 1465-75, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23086218

ABSTRACT

Phylogenetic analyses suggest that violations of "Dollo's law"--that is, re-evolution of lost complex structures--do occur, albeit infrequently. However, the genetic basis of such reversals has not been examined. Here, we address this question using the Drosophila sex comb, a recently evolved, male-specific morphological structure composed of modified bristles. In some species, sex comb development involves only the modification of individual bristles, while other species have more complex "rotated" sex combs that are shaped by coordinated migration of epithelial tissues. Rotated sex combs were lost in the ananassae species subgroup and subsequently re-evolved, ∼12 million years later, in Drosophila bipectinata and its sibling species. We examine the genetic basis of the differences in sex comb morphology between D. bipectinata and D. malerkotliana, a closely related species with a much simpler sex comb representing the ancestral condition. QTL mapping reveals that >50% of this difference is controlled by one chromosomal inversion that covers ∼5% of the genome. Several other, larger inversions do not contribute appreciably to the phenotype. This genetic architecture suggests that rotating sex combs may have re-evolved through changes in relatively few genes. We discuss potential developmental mechanisms that may allow lost complex structures to be regained.


Subject(s)
Biological Evolution , Drosophila/anatomy & histology , Drosophila/genetics , Animals , Chimera , Chromosome Inversion , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Female , Genome, Insect , Male , Phenotype , Quantitative Trait Loci , Species Specificity , Transcription Factors/genetics
13.
J Neuroimmunol ; 240-241: 129-36, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079192

ABSTRACT

Intrathecal antibody production manifest as oligoclonal bands (OCBs) is a hallmark of multiple sclerosis (MS). Once present, OCBs can be detected in CSF throughout the lifetime of MS patients. To determine the specificity of the OCBs, we applied CSF IgG obtained from 2 consecutive lumbar punctures of 5 MS patients to screen phage-displayed random peptide libraries, and selected identical and related peptides that reacted with the paired CSF IgGs from each patient. Highly sensitive phage-mediated immuno-PCR revealed that the phage peptides bound specifically to IgG in MS CSF collected over time. IEF immunoblots also showed that these peptides were recognized by OCBs in MS CSF. We further demonstrated that the peptides represented linear epitopes, indicating that they represent natural epitopes of corresponding protein antigens. A database search combined with alanine scan mutagenesis of peptides that bound to CSF IgG from 3 MS patients revealed that they are derived from proteins including serine/threonine-protein kinase, protein ZIP2 and MHC class II. Identification of epitopes that are recognized by IgG in MS CSF over time provides a critical tool to investigate the specificity of OCBs, which may determine the cause of disease, leading to strategies for diagnostic and therapeutic intervention.


Subject(s)
Cerebrospinal Fluid Proteins/biosynthesis , Epitopes/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Adult , Cerebrospinal Fluid Proteins/metabolism , Epitopes/metabolism , Female , Humans , Immunoglobulin G/metabolism , Male , Middle Aged , Peptides/metabolism , Protein Binding/immunology , Time Factors
14.
PLoS Biol ; 9(8): e1001131, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21886483

ABSTRACT

Almost every animal lineage is characterized by unique sex-specific traits, implying that such traits are gained and lost frequently in evolution. However, the genetic mechanisms responsible for these changes are not understood. In Drosophila, the activity of the sex determination pathway is restricted to sexually dimorphic tissues, suggesting that spatial regulation of this pathway may contribute to the evolution of sex-specific traits. We examine the regulation and function of doublesex (dsx), the main transcriptional effector of the sex determination pathway, in the development and evolution of Drosophila sex combs. Sex combs are a recent evolutionary innovation and show dramatic diversity in the relatively few Drosophila species that have them. We show that dsx expression in the presumptive sex comb region is activated by the HOX gene Sex combs reduced (Scr), and that the male isoform of dsx up-regulates Scr so that both genes become expressed at high levels in this region in males but not in females. Precise spatial regulation of dsx is essential for defining sex comb position and morphology. Comparative analysis of Scr and dsx expression reveals a tight correlation between sex comb morphology and the expression patterns of both genes. In species that primitively lack sex combs, no dsx expression is observed in the homologous region, suggesting that the origin and diversification of this structure were linked to the gain of a new dsx expression domain. Two other, distantly related fly lineages that independently evolved novel male-specific structures show evolutionary gains of dsx expression in the corresponding tissues, where dsx may also be controlled by Scr. These findings suggest that changes in the spatial regulation of sex-determining genes are a key mechanism that enables the evolution of new sex-specific traits, contributing to some of the most dramatic examples of phenotypic diversification in nature.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Evolution, Molecular , Sex Differentiation/genetics , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epistasis, Genetic , Female , Gene Expression , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Larva/metabolism , Male , Morphogenesis , Organ Specificity , Phylogeny , RNA Interference , Sex Characteristics , Transcription Factors/genetics
15.
J Neuroimmunol ; 233(1-2): 192-203, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21176973

ABSTRACT

We employed 19 recombinant antibodies (rAbs) generated from clonally expanded plasma cells, and native IgG from cerebrospinal fluid (CSF) of three multiple sclerosis (MS) patients for panning with phage displayed random peptide libraries. Specific peptide epitopes/mimotopes were identified and characterized. Importantly, peptide-antibody interactions were shared by rAbs and native IgG from the same patient. Three peptides strongly interacted with at least one other MS CSF, but not to inflammatory CNS controls. Database searches revealed several protein candidates including stress proteins, cell surface proteins, and neuronal proteins. Peptides derived from the candidate proteins were recognized by rAbs. Identification of peptide epitopes/mimotopes in MS may provide clues regarding disease-relevant antigens.


Subject(s)
Autoantibodies/biosynthesis , Immunoglobulin G/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Peptide Fragments/metabolism , Plasma Cells/immunology , Recombinant Proteins/metabolism , Clone Cells , Humans , Peptide Fragments/immunology , Peptide Library , Plasma Cells/pathology , Recombinant Proteins/immunology
16.
Proc Natl Acad Sci U S A ; 106(12): 4764-9, 2009 Mar 24.
Article in English | MEDLINE | ID: mdl-19255422

ABSTRACT

Similar selective pressures can lead to independent origin of similar morphological structures in multiple evolutionary lineages. Developmental mechanisms underlying convergent evolution remain poorly understood. In this report, we show that similar sex comb morphology in closely related Drosophila species is produced by different cellular mechanisms. The sex comb is a recently evolved, male-specific array of modified bristles derived from transverse bristle rows found on the first thoracic legs in both sexes. "Longitudinal" sex combs oriented along the proximo-distal leg axis evolved independently in several Drosophila lineages. We show that in some of these lineages, sex combs originate as one or several transverse bristle rows that subsequently rotate 90 degrees and align to form a single longitudinal row. In other species, bristle cells that make up the sex combs arise in their final longitudinal orientation. Thus, sex combs can develop through either sex-specific patterning of bristle precursor cells or male-specific morphogenesis of sexually monomorphic precursors. Surprisingly, the two mechanisms produce nearly identical morphology in some species. Phylogenetic analysis shows that each of these mechanisms has probably evolved repeatedly in different Drosophila lineages, suggesting that selection can recruit different cellular processes to produce similar functional solutions.


Subject(s)
Animal Structures/embryology , Biological Evolution , Drosophila/anatomy & histology , Drosophila/embryology , Animals , Morphogenesis , Phylogeny , Rotation
17.
J Virol Methods ; 156(1-2): 169-73, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19095007

ABSTRACT

Phage-displayed random peptide libraries, in which high affinity phage peptides are enriched by repetitive selection (panning) on target antibody, provide a unique tool for identifying antigen specificity. This paper describes a new panning method that enables selection of peptides in 1 day as compared to about 6 days required in traditional panning to identify virus-specific epitopes. The method, termed ultra-fast selection of peptide (UFSP), utilizes phage produced by bacterial infection (phage amplification) directly for subsequent panning. Phage amplified in less than 1h of infection in Escherichia coli are used for binding to target antibody pre-coated in the same wells of an ELISA plate, obviating the need for traditional large-scale amplification and purification. Importantly, phage elution at 37 degrees C was superior to that at room temperature, and phage amplification in a 150-microl volume of E. coli cells was superior to that in 250-microl volume. Application of UFSP to two monoclonal antibodies generated from clonally expanded plasma cells in subacute sclerosing panencephalitis (SSPE) brain identified high-affinity measles virus-specific-peptide epitopes. The UFSP panning methodology will expedite identification of peptides reacting with antibodies generated in other diseases of unknown antigenic specificity such as multiple sclerosis (MS), sarcoidosis and Behcet's disease.


Subject(s)
Antigens, Viral/isolation & purification , Epitopes/isolation & purification , Measles virus/chemistry , Peptide Library , Antibodies, Viral , Antibody Specificity , Enzyme-Linked Immunosorbent Assay , Escherichia coli/metabolism , Escherichia coli/virology
18.
Genetics ; 180(1): 421-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18723880

ABSTRACT

Pigmentation is a rapidly evolving trait that can play important roles in mimicry, sexual selection, thermoregulation, and other adaptive processes in many groups of animals. In Drosophila, pigmentation can differ dramatically among closely related taxa, presenting a good opportunity to dissect the genetic changes underlying species divergence. In this report, we investigate the genetic basis of color pattern variation between two allopatric subspecies of Drosophila malerkotliana, a widespread member of the ananassae species subgroup. In D. malerkotliana malerkotliana, the last three abdominal segments are darkly pigmented in males but not in females, while in D. malerkotliana pallens both sexes lack dark pigmentation. Composite interval mapping in F2 hybrid progeny shows that this difference is largely controlled by three quantitative trait loci (QTL) located on the 2L chromosome arm, which is homologous to the 3R of D. melanogaster (Muller element E). Using highly recombinant introgression strains produced by repeated backcrossing and phenotypic selection, we show that these QTL do not correspond to any of the candidate genes known to be involved in pigment patterning and synthesis in Drosophila. These results, in combination with similar analyses in other Drosophila species, indicate that different genetic and molecular changes are responsible for the evolution of similar phenotypic traits in different lineages. This feature makes Drosophila color patterns a powerful model for investigating how the genetic basis of trait evolution is influenced by the intrinsic organization of regulatory pathways controlling the development of these traits.


Subject(s)
Drosophila/genetics , Drosophila/physiology , Gene Expression Regulation , Genes, Insect , Pigmentation/genetics , Animals , Behavior, Animal , Crosses, Genetic , Epistasis, Genetic , Female , Genotype , Male , Models, Genetic , Quantitative Trait Loci , Sex Factors , Sexual Behavior, Animal/physiology
19.
Mol Biol Evol ; 25(6): 1081-92, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18296696

ABSTRACT

Host plant shifts by phytophagous insects play a key role in insect evolution and plant ecology. Such shifts often involve major behavioral changes as the insects must acquire an attraction and/or lose the repulsion to the new host plant's odor and taste. The evolution of chemotactic behavior may be due, in part, to gene expression changes in the peripheral sensory system. To test this hypothesis, we compared gene expression in the olfactory organs of Drosophila sechellia, a narrow ecological specialist that feeds on the fruit of Morinda citrifolia, with its close relatives Drosophila simulans and Drosophila melanogaster, which feed on a wide variety of decaying plant matter. Using whole-genome microarrays and quantitative polymerase chain reaction, we surveyed the entire repertoire of Drosophila odorant receptors (ORs) and odorant-binding proteins (OBPs) expressed in the antennae. We found that the evolution of OR and OBP expression was accelerated in D. sechellia compared both with the genome average in that species and with the rate of OR and OBP evolution in the other species. However, some of the gene expression changes that correlate with D. sechellia's increased sensitivity to Morinda odorants may predate its divergence from D. simulans. Interspecific divergence of olfactory gene expression cannot be fully explained by changes in the relative abundance of different sensilla as some ORs and OBPs have evolved independently of other genes expressed in the same sensilla. A number of OR and OBP genes are upregulated in D. sechellia compared with its generalist relatives. These genes include Or22a, which likely responds to a key odorant of M. citrifolia, and several genes that are yet to be characterized in detail. Increased expression of these genes in D. sechellia may have contributed to the evolution of its unique chemotactic behavior.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Evolution, Molecular , Gene Expression , Olfactory Pathways/metabolism , Smell/genetics , Animals , Drosophila/metabolism , Genes, Insect , Genetic Variation , Multigene Family , Oligonucleotide Array Sequence Analysis
20.
Dev Biol ; 311(2): 277-86, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17868668

ABSTRACT

Animal diversity is shaped by the origin and diversification of new morphological structures. Many examples of evolutionary innovations are provided by male-specific traits involved in mating and sexual selection. The origin of new sex-specific characters requires the evolution of new regulatory interactions between sex-determining genes and genes that control spatial patterning and cell differentiation. Here, we show that sex-specific regulation of the HOX gene Sex combs reduced (Scr) is associated with the origin and evolution of the Drosophila sex comb - a novel and rapidly diversifying male-specific organ. In species that primitively lack sex combs, Scr expression shows little spatial modulation, whereas in species that have sex combs, Scr is upregulated in the presumptive sex comb region and is frequently sexually dimorphic. Phylogenetic analysis shows that sex-specific regulation of Scr has been gained and lost multiple times in Drosophila evolution and correlates with convergent origin of similar sex comb morphologies in several independent lineages. Some of these transitions occurred on microevolutionary timescales, indicating that HOX gene expression can evolve with surprising ease. This is the first example of a sex-specific regulation of a HOX gene contributing to the development and evolution of a secondary sexual trait.


Subject(s)
Biological Evolution , Drosophila Proteins , Drosophila melanogaster , Gene Expression Regulation, Developmental , Genes, Homeobox , Morphogenesis , Sex Differentiation/genetics , Transcription Factors , Animals , Drosophila Proteins/classification , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Extremities/anatomy & histology , Extremities/embryology , Extremities/growth & development , Female , Homeodomain Proteins/classification , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Phylogeny , Sex Characteristics , Transcription Factors/classification , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...