fbpx
Wikipedia

Drosophila melanogaster

Drosophila melanogaster is a species of fly (an insect of the order Diptera) in the family Drosophilidae. The species is often referred to as the fruit fly or lesser fruit fly, or less commonly the "vinegar fly", "pomace fly",[a][4] or "banana fly".[5] In the wild, D. melanogaster are attracted to rotting fruit and fermenting beverages, and are often found in orchards, kitchens and pubs.

Drosophila melanogaster
Fruit fly feeding off a banana
Scientific classification
Domain: Eukaryota
Kingdom: Animalia
Phylum: Arthropoda
Class: Insecta
Order: Diptera
Family: Drosophilidae
Genus: Drosophila
Subgenus: Sophophora
Species group: melanogaster
Species subgroup: melanogaster
Species complex: melanogaster
Species:
D. melanogaster
Binomial name
Drosophila melanogaster
Meigen, 1830[1]

Starting with Charles W. Woodworth's 1901 proposal of the use of this species as a model organism,[6][7] D. melanogaster continues to be widely used for biological research in genetics, physiology, microbial pathogenesis, and life history evolution. As of 2017, six Nobel Prizes have been awarded to drosophilists for their work using the insect.[8][9]

D. melanogaster is typically used in research owing to its rapid life cycle, relatively simple genetics with only four pairs of chromosomes, and large number of offspring per generation.[10] It was originally an African species, with all non-African lineages having a common origin.[11] Its geographic range includes all continents, including islands.[12] D. melanogaster is a common pest in homes, restaurants, and other places where food is served.[13]

Flies belonging to the family Tephritidae are also called "fruit flies". This can cause confusion, especially in the Mediterranean, Australia, and South Africa, where the Mediterranean fruit fly Ceratitis capitata is an economic pest.

Etymology edit

The term "Drosophila", meaning "dew-loving", is a modern scientific Latin adaptation from Greek words δρόσος, drósos, "dew", and φιλία, philía, "lover". The term "melanogaster" meaning "black-belly", comes from Ancient Greek μέλας, mélas, “black”, and γᾰστήρ, gastḗr, "belly".

Physical appearance edit

 
Female (left) and male (right) D. melanogaster
 
View from above
 
Front view

Wild type fruit flies are yellow-brown, with brick-red eyes and transverse black rings across the abdomen. The black portions of the abdomen are the inspiration for the species name (melanogaster = "black-bellied"). The brick-red color of the eyes of the wild type fly are due to two pigments:[14] xanthommatin, which is brown and is derived from tryptophan, and drosopterins, which are red and are derived from guanosine triphosphate.[14] They exhibit sexual dimorphism; females are about 2.5 mm (0.10 in) long; males are slightly smaller with darker backs. Males are easily distinguished from females based on colour differences, with a distinct black patch at the abdomen, less noticeable in recently emerged flies, and the sex combs (a row of dark bristles on the tarsus of the first leg). Furthermore, males have a cluster of spiky hairs (claspers) surrounding the reproducing parts used to attach to the female during mating. Extensive images are found at FlyBase.[15]

Drosophila melanogaster can be distinguished from related species by the following combination of features: gena ~1/10 diameter of eye at greatest vertical height; wing hyaline and with costal index 2.4; male protarsus with a single row of ~12 setae forming a sex comb; male epandrial posterior lobe small and nearly triangular; female abdominal tergite 6 with dark band running to its ventral margin; female oviscapt small, pale, without dorsodistal depression and with 12-13 peg-like outer ovisensilla.[16][17]

Drosophila melanogaster flies can sense air currents with the hairs on their backs. Their eyes are sensitive to slight differences in light intensity and will instinctively fly away when a shadow or other movement is detected.[18]

Lifecycle and reproduction edit

 
Egg of D. melanogaster

Under optimal growth conditions at 25 °C (77 °F), the D. melanogaster lifespan is about 50 days from egg to death.[19] The developmental period for D. melanogaster varies with temperature, as with many ectothermic species. The shortest development time (egg to adult), 7 days, is achieved at 28 °C (82 °F).[20][21] Development times increase at higher temperatures (11 days at 30 °C or 86 °F) due to heat stress. Under ideal conditions, the development time at 25 °C (77 °F) is 8.5 days,[20][21][22] at 18 °C (64 °F) it takes 19 days[20][21] and at 12 °C (54 °F) it takes over 50 days.[20][21] Under crowded conditions, development time increases,[23] while the emerging flies are smaller.[23][24] Females lay some 400 eggs (embryos), about five at a time, into rotting fruit or other suitable material such as decaying mushrooms and sap fluxes. Drosophila melanogaster is a holometabolous insect, so it undergoes a full metamorphosis. Their life cycle is broken down into 4 stages: embryo, larva, pupa, adult.[25] The eggs, which are about 0.5 mm long, hatch after 12–15 hours (at 25 °C or 77 °F).[20][21] The resulting larvae grow for about 4 days (at 25 °C) while molting twice (into second- and third-instar larvae), at about 24 and 48 h after hatching.[20][21] During this time, they feed on the microorganisms that decompose the fruit, as well as on the sugar of the fruit itself. The mother puts feces on the egg sacs to establish the same microbial composition in the larvae's guts that has worked positively for herself.[26] Then the larvae encapsulate in the puparium and undergo a 4-day-long metamorphosis (at 25 °C), after which the adults eclose (emerge).[20][21]

Sound of Drosophila heartbeat

Males perform a sequence of five behavioral patterns to court females. First, males orient themselves while playing a courtship song by horizontally extending and vibrating their wings. Soon after, the male positions himself at the rear of the female's abdomen in a low posture to tap and lick the female genitalia. Finally, the male curls his abdomen and attempts copulation. Females can reject males by moving away, kicking, and extruding their ovipositor.[27] Copulation lasts around 15–20 minutes,[28] during which males transfer a few hundred, very long (1.76 mm) sperm cells in seminal fluid to the female.[29] Females store the sperm in a tubular receptacle and in two mushroom-shaped spermathecae; sperm from multiple matings compete for fertilization. A last male precedence is believed to exist; the last male to mate with a female sires about 80% of her offspring. This precedence was found to occur through both displacement and incapacitation.[30] The displacement is attributed to sperm handling by the female fly as multiple matings are conducted and is most significant during the first 1–2 days after copulation. Displacement from the seminal receptacle is more significant than displacement from the spermathecae.[30] Incapacitation of first male sperm by second male sperm becomes significant 2–7 days after copulation. The seminal fluid of the second male is believed to be responsible for this incapacitation mechanism (without removal of first male sperm) which takes effect before fertilization occurs.[30] The delay in effectiveness of the incapacitation mechanism is believed to be a protective mechanism that prevents a male fly from incapacitating his own sperm should he mate with the same female fly repetitively. Sensory neurons in the uterus of female D. melanogaster respond to a male protein, sex peptide, which is found in semen.[31] This protein makes the female reluctant to copulate for about 10 days after insemination. The signal pathway leading to this change in behavior has been determined. The signal is sent to a brain region that is a homolog of the hypothalamus and the hypothalamus then controls sexual behavior and desire.[31] Gonadotropic hormones in Drosophila maintain homeostasis and govern reproductive output via a cyclic interrelationship, not unlike the mammalian estrous cycle.[32] Sex peptide perturbs this homeostasis and dramatically shifts the endocrine state of the female by inciting juvenile hormone synthesis in the corpus allatum.[33]

D. melanogaster is often used for life extension studies, such as to identify genes purported to increase lifespan when mutated.[34] D. melanogaster is also used in studies of aging. Werner syndrome is a condition in humans characterized by accelerated aging. It is caused by mutations in the gene WRN that encodes a protein with essential roles in repair of DNA damage. Mutations in the D. melanogaster homolog of WRN also cause increased physiologic signs of aging, such as shorter lifespan, higher tumor incidence, muscle degeneration, reduced climbing ability, altered behavior and reduced locomotor activity.[35]

Females edit

 
Mating in captivity

Females become receptive to courting males about 8–12 hours after emergence.[36] Specific neuron groups in females have been found to affect copulation behavior and mate choice. One such group in the abdominal nerve cord allows the female fly to pause her body movements to copulate.[31] Activation of these neurons induces the female to cease movement and orient herself towards the male to allow for mounting. If the group is inactivated, the female remains in motion and does not copulate. Various chemical signals such as male pheromones often are able to activate the group.[31]

Also, females exhibit mate choice copying. When virgin females are shown other females copulating with a certain type of male, they tend to copulate more with this type of male afterwards than naïve females (which have not observed the copulation of others). This behavior is sensitive to environmental conditions, and females copulate less in bad weather conditions.[37]

Males edit

Courtship behavior in male. The male first showed wing extension (stage 1), and later other steps such as abdomen bending (stage 2), then frequent attempted copulation, licking and even ejaculation (stage 3), finally the male fell over and was on its back (stage 4).

D. melanogaster males exhibit a strong reproductive learning curve. That is, with sexual experience, these flies tend to modify their future mating behavior in multiple ways. These changes include increased selectivity for courting only intraspecifically, as well as decreased courtship times.

Sexually naïve D. melanogaster males are known to spend significant time courting interspecifically, such as with D. simulans flies. Naïve D. melanogaster will also attempt to court females that are not yet sexually mature, and other males. D. melanogaster males show little to no preference for D. melanogaster females over females of other species or even other male flies. However, after D. simulans or other flies incapable of copulation have rejected the males' advances, D. melanogaster males are much less likely to spend time courting nonspecifically in the future. This apparent learned behavior modification seems to be evolutionarily significant, as it allows the males to avoid investing energy into futile sexual encounters.[38]

In addition, males with previous sexual experience modify their courtship dance when attempting to mate with new females—the experienced males spend less time courting, so have lower mating latencies, meaning that they are able to reproduce more quickly. This decreased mating latency leads to a greater mating efficiency for experienced males over naïve males.[39] This modification also appears to have obvious evolutionary advantages, as increased mating efficiency is extremely important in the eyes of natural selection.

Polygamy edit

Both male and female D. melanogaster flies act polygamously (having multiple sexual partners at the same time).[40] In both males and females, polygamy results in a decrease in evening activity compared to virgin flies, more so in males than females.[40] Evening activity consists of those in which the flies participate other than mating and finding partners, such as finding food.[41] The reproductive success of males and females varies, because a female only needs to mate once to reach maximum fertility.[41] Mating with multiple partners provides no advantage over mating with one partner, so females exhibit no difference in evening activity between polygamous and monogamous individuals.[41] For males, however, mating with multiple partners increases their reproductive success by increasing the genetic diversity of their offspring.[41] This benefit of genetic diversity is an evolutionary advantage because it increases the chance that some of the offspring will have traits that increase their fitness in their environment.

The difference in evening activity between polygamous and monogamous male flies can be explained with courtship. For polygamous flies, their reproductive success increases by having offspring with multiple partners, and therefore they spend more time and energy on courting multiple females.[41] On the other hand, monogamous flies only court one female, and expend less energy doing so.[41] While it requires more energy for male flies to court multiple females, the overall reproductive benefits it produces has kept polygamy as the preferred sexual choice.[41]

The mechanism that affects courtship behavior in Drosophila is controlled by the oscillator neurons DN1s and LNDs.[42] Oscillation of the DN1 neurons was found to be effected by sociosexual interactions, and is connected to mating-related decrease of evening activity.[42]

Model organism in genetics edit

D. melanogaster remains one of the most studied organisms in biological research, particularly in genetics and developmental biology. It is also employed in studies of environmental mutagenesis.

History of use in genetic analysis edit

 
Alfred Sturtevant's Drosophila melanogaster genetic linkage map: This was the first successful gene mapping work and provides important evidence for the chromosome theory of inheritance. The map shows the relative positions of allelic characteristics on the second Drosophila chromosome. The distance between the genes (map units) are equal to the percentage of crossing-over events that occurs between different alleles.

D. melanogaster was among the first organisms used for genetic analysis, and today it is one of the most widely used and genetically best-known of all eukaryotic organisms. All organisms use common genetic systems; therefore, comprehending processes such as transcription and replication in fruit flies helps in understanding these processes in other eukaryotes, including humans.[43]

Thomas Hunt Morgan began using fruit flies in experimental studies of heredity at Columbia University in 1910 in a laboratory known as the Fly Room. The Fly Room was cramped with eight desks, each occupied by students and their experiments. They started off experiments using milk bottles to rear the fruit flies and handheld lenses for observing their traits. The lenses were later replaced by microscopes, which enhanced their observations. Morgan and his students eventually elucidated many basic principles of heredity, including sex-linked inheritance, epistasis, multiple alleles, and gene mapping.[43]

D. melanogaster had historically been used in laboratories to study genetics and patterns of inheritance. However, D. melanogaster also has importance in environmental mutagenesis research, allowing researchers to study the effects of specific environmental mutagens.[44]

Reasons for use in laboratories edit

 
D. melanogaster multiple mutants (clockwise from top): brown eyes and black cuticle (2 mutations), cinnabar eyes and wildtype cuticle (1 mutation), sepia eyes and ebony cuticle, vermilion eyes and yellow cuticle, white eyes and yellow cuticle, wildtype eyes and yellow cuticle.

There are many reasons the fruit fly is a popular choice as a model organism:

  • Its care and culture require little equipment, space, and expense even when using large cultures.
  • It can be safely and readily anesthetized (usually with ether, carbon dioxide gas, by cooling, or with products such as FlyNap).
  • Its morphology is easy to identify once anesthetized.
  • It has a short generation time (about 10 days at room temperature), so several generations can be studied within a few weeks.
  • It has a high fecundity (females lay up to 100 eggs per day, and perhaps 2000 in a lifetime).[10]
  • Males and females are readily distinguished, and virgin females can be easily identified by their light-colored, translucent abdomen, facilitating genetic crossing.
  • The mature larva has giant chromosomes in the salivary glands called polytene chromosomes, "puffs", which indicate regions of transcription, hence gene activity. The under-replication of rDNA occurs resulting in only 20% of DNA compared to the brain. Compare to the 47%, less rDNA in Sarcophaga barbata ovaries.
  • It has only four pairs of chromosomes – three autosomes, and one pair of sex chromosomes.
  • Males do not show meiotic recombination, facilitating genetic studies.
  • Recessive lethal "balancer chromosomes" carrying visible genetic markers can be used to keep stocks of lethal alleles in a heterozygous state without recombination due to multiple inversions in the balancer.
  • The development of this organism—from fertilized egg to mature adult—is well understood.
  • Genetic transformation techniques have been available since 1987. One approach of inserting foreign genes into the Drosophila genome involves P elements. The transposable P elements, also known as transposons, are segments of bacterial DNA that are transferred into the fly genome. Transgenic flies have already contributed to many scientific advances, e.g., modeling such human diseases as Parkinson's, neoplasia, obesity, and diabetes.
  • Its complete genome was sequenced and first published in 2000.[45]
  • Sexual mosaics can be readily produced, providing an additional tool for studying the development and behavior of these flies.[46]

Genetic markers edit

 
D. melanogaster which carries the Cy allele (right), hence showing a characteristic phenotype of curly wings in adult flies[47]

Genetic markers are commonly used in Drosophila research, for example within balancer chromosomes or P-element inserts, and most phenotypes are easily identifiable either with the naked eye or under a microscope. In the list of a few common markers below, the allele symbol is followed by the name of the gene affected and a description of its phenotype. (Note: Recessive alleles are in lower case, while dominant alleles are capitalised.)

  • Cy1: Curly; the wings curve away from the body, flight may be somewhat impaired
  • e1: Ebony; black body and wings (heterozygotes are also visibly darker than wild type)
  • Sb1: Stubble; bristles are shorter and thicker than wild type
  • w1: White; eyes lack pigmentation and appear white
  • bw: Brown; eye color determined by various pigments combined.
  • y1: Yellow; body pigmentation and wings appear yellow, the fly analog of albinism

Classic genetic mutations edit

Drosophila genes are traditionally named after the phenotype they cause when mutated. For example, the absence of a particular gene in Drosophila will result in a mutant embryo that does not develop a heart. Scientists have thus called this gene tinman, named after the Oz character of the same name.[48] Likewise changes in the Shavenbaby gene cause the loss of dorsal cuticular hairs in Drosophila sechellia larvae.[49] This system of nomenclature results in a wider range of gene names than in other organisms.

  • b: black- The black mutation was discovered in 1910 by Thomas Hunt Morgan.[50] The black mutation results in a darker colored body, wings, veins, and segments of the fruit fly's leg.[51] This occurs due to the fly's inability to create beta-alanine, a beta amino acid.[50] The phenotypic expression of this mutation varies based on the genotype of the individual; for example, whether the specimen is homozygotic or heterozygotic results in a darker or less dark appearance.[51] This genetic mutation is x-linked recessive.[52]
  • bw: brown- The brown eye mutation results from inability to produce or synthesize pteridine (red) pigments, due to a point mutation on chromosome II.[53] When the mutation is homozygous, the pteridine pigments are unable to be synthesized because in the beginning of the pteridine pathway, a defective enzyme is being coded by homozygous recessive genes.[54][unreliable source?] In all, mutations in the pteridine pathway produces a darker eye color, hence the resulting color of the biochemical defect in the pteridine pathway being brown.
  • m: miniature- One of the first records of the miniature mutation of wings was also made by Thomas Hunt Morgan in 1911. He described the wings as having a similar shape as the wild-type phenotype. However, their miniature designation refers to the lengths of their wings, which do not stretch beyond their body and, thus, are notably shorter than the wild-type length. He also noted its inheritance is connected to the sex of the fly and could be paired with the inheritance of other sex-determined traits such as white eyes.[55] The wings may also demonstrate other characteristics deviant from the wild-type wing, such as a duller and cloudier color.[56] Miniature wings are 1.5x shorter than wild-type but are believed to have the same number of cells. This is due to the lack of complete flattening by these cells, making the overall structure of the wing seem shorter in comparison. The pathway of wing expansion is regulated by a signal-receptor pathway, where the neurohormone bursicon interacts with its complementary G protein-coupled receptor; this receptor drives one of the G-protein subunits to signal further enzyme activity and results in development in the wing, such as apoptosis and growth.[57]
  • se: sepia- The eye color of the sepia mutant is sepia, a reddish-brown color. In wild-type flies, ommochromes (brown) and drosopterins (red) give the eyes the typical red color.[58][59] The drosopterins are made via a pathway that involves a pyrimidodiazepine synthase,[60] which is encoded on chromosome 3L. The gene has a premature stop codon in sepia flies, so that the flies cannot produce the pyrimidodiazepine synthase and thus no red pigment, so that the eyes stay sepia.[58] The sepia allele is recessive and thus offspring from sepia flies and homozygous wild type flies, has red eyes. The sepia phenotype does not depend on the sex of the fly.[61]
  • v: vermilion- The vermilion mutants cannot produce the brown ommochromes leaving the red drosopterins so that the eyes are vermilion colored (a radiant red) compared to a wild-type D. melanogaster. The vermilion mutation is sex-linked and recessive. The gene that is defect lies on the X chromosome.[62] The brown ommochromes are synthesised from kynurenine, which is made from tryptophane. Vermilion flies cannot convert tryptophane into kynurenine and thus cannot make ommochromes, either.[62] Vermilion mutants live longer than wild-type flies. This longer life span may be associated with the reduced amount of tryptophan converted to kynurenine in vermilion flies.[63]
 
Triple mutant male fruit fly (Drosophila melanogaster) exhibiting black body, vestigial wings, and brown eyes mutations
  • vg: vestigial- A spontaneous mutation, discovered in 1919 by Thomas Morgan and Calvin Bridges. Vestigial wings are those not fully developed and that have lost function. Since the discovery of the vestigial gene in Drosophila melanogaster, there have been many discoveries of the vestigial gene in other vertebrates and their functions within the vertebrates.[64] The vestigial gene is considered to be one of the most important genes for wing formation, but when it becomes over expressed the issue of ectopic wings begin to form.[65] The vestigial gene acts to regulate the expression of the wing imaginal discs in the embryo and acts with other genes to regulate the development of the wings. A mutated vestigial allele removes an essential sequence of the DNA required for correct development of the wings.[66]
  • w: white- Drosophila melanogaster wild type typically expresses a brick red eye color. The white eye mutation in fruit flies is caused due to the absence of two pigments associated with red and brown eye colors; peridines (red) and ommochromes (brown).[59] In January 1910, Thomas Hunt Morgan first discovered the white gene and denoted it as w. The discovery of the white-eye mutation by Morgan brought about the beginnings of genetic experimentation and analysis of Drosophila melanogaster. Hunt eventually discovered that the gene followed a similar pattern of inheritance related to the meiotic segregation of the X chromosome. He discovered that the gene was located on the X chromosome with this information. This led to the discovery of sex-linked genes and also to the discovery of other mutations in Drosophila melanogaster.[67] The white-eye mutation leads to several disadvantages in flies, such as a reduced climbing ability, shortened life span, and lowered resistance to stress when compared to wild type flies.[68] Drosophila melanogaster has a series of mating behaviors that enable them to copulate within a given environment and therefore contribute to their fitness. After Morgan's discovery of the white-eye mutation being sex-linked, a study led by Sturtevant (1915) concluded that white-eyed males were less successful than wild-type males in terms of mating with females.[69] It was found that the greater the density in eye pigmentation, the greater the success in mating for the males of Drosophila melanogaster.[69]
  • y: yellow- The yellow gene is a genetic mutation known as Dmel\y within the widely used data base called FlyBase. This mutation can be easily identified by the atypical yellow pigment observed in the cuticle of the adult flies and the mouth pieces of the larva.[70] The y mutation comprises the following phenotypic classes: the mutants that show a complete loss of pigmentation from the cuticle (y-type) and other mutants that show a mosaic pigment pattern with some regions of the cuticle (wild type, y2-type).[71] The role of the yellow gene is diverse and is responsible for changes in behaviour, sex-specific reproductive maturation and, epigenetic reprogramming.[72] The y gene is an ideal gene to study as it is visibly clear when an organism has this gene, making it easier to understand the passage of DNA to offspring.[72]
 
Wild-type wing (left) vs. miniature wing (right)

Genome edit

Genomic information
 
D. melanogaster chromosomes to scale with megabase-pair references oriented as in the National Center for Biotechnology Information database, centimorgan distances are approximate and estimated from the locations of selected mapped loci.
NCBI genome ID47
Ploidydiploid
Number of chromosomes8
Year of completion2015

The genome of D. melanogaster (sequenced in 2000, and curated at the FlyBase database[45]) contains four pairs of chromosomes – an X/Y pair, and three autosomes labeled 2, 3, and 4. The fourth chromosome is relatively very small and therefore often ignored, aside from its important eyeless gene. The D. melanogaster sequenced genome of 139.5 million base pairs has been annotated[73] and contains around 15,682 genes according to Ensemble release 73. More than 60% of the genome appears to be functional non-protein-coding DNA[74] involved in gene expression control. Determination of sex in Drosophila occurs by the X:A ratio of X chromosomes to autosomes, not because of the presence of a Y chromosome as in human sex determination. Although the Y chromosome is entirely heterochromatic, it contains at least 16 genes, many of which are thought to have male-related functions.[75]

There are three transferrin orthologs, all of which are dramatically divergent from those known in chordate models.[76]

Similarity to humans edit

A March 2000 study by National Human Genome Research Institute comparing the fruit fly and human genome estimated that about 60% of genes are conserved between the two species.[77] About 75% of known human disease genes have a recognizable match in the genome of fruit flies,[78] and 50% of fly protein sequences have mammalian homologs[citation needed]. An online database called Homophila is available to search for human disease gene homologues in flies and vice versa.[79]

Drosophila is being used as a genetic model for several human diseases including the neurodegenerative disorders Parkinson's, Huntington's, spinocerebellar ataxia and Alzheimer's disease.[80] The fly is also being used to study mechanisms underlying aging and oxidative stress, immunity, diabetes, and cancer, as well as drug abuse.[81][82][83]

Development edit

The life cycle of this insect has four stages: fertilized egg, larva, pupa, and adult.[12]

Embryogenesis in Drosophila has been extensively studied, as its small size, short generation time, and large brood size makes it ideal for genetic studies. It is also unique among model organisms in that cleavage occurs in a syncytium.

 
D. melanogaster oogenesis

During oogenesis, cytoplasmic bridges called "ring canals" connect the forming oocyte to nurse cells. Nutrients and developmental control molecules move from the nurse cells into the oocyte. In the figure to the left, the forming oocyte can be seen to be covered by follicular support cells.

After fertilization of the oocyte, the early embryo (or syncytial embryo) undergoes rapid DNA replication and 13 nuclear divisions until about 5000 to 6000 nuclei accumulate in the unseparated cytoplasm of the embryo. By the end of the eighth division, most nuclei have migrated to the surface, surrounding the yolk sac (leaving behind only a few nuclei, which will become the yolk nuclei). After the 10th division, the pole cells form at the posterior end of the embryo, segregating the germ line from the syncytium. Finally, after the 13th division, cell membranes slowly invaginate, dividing the syncytium into individual somatic cells. Once this process is completed, gastrulation starts.[84]

Nuclear division in the early Drosophila embryo happens so quickly, no proper checkpoints exist, so mistakes may be made in division of the DNA. To get around this problem, the nuclei that have made a mistake detach from their centrosomes and fall into the centre of the embryo (yolk sac), which will not form part of the fly.

The gene network (transcriptional and protein interactions) governing the early development of the fruit fly embryo is one of the best understood gene networks to date, especially the patterning along the anteroposterior (AP) and dorsoventral (DV) axes (See under morphogenesis).[84]

The embryo undergoes well-characterized morphogenetic movements during gastrulation and early development, including germ-band extension, formation of several furrows, ventral invagination of the mesoderm, and posterior and anterior invagination of endoderm (gut), as well as extensive body segmentation until finally hatching from the surrounding cuticle into a first-instar larva.

During larval development, tissues known as imaginal discs grow inside the larva. Imaginal discs develop to form most structures of the adult body, such as the head, legs, wings, thorax, and genitalia. Cells of the imaginal disks are set aside during embryogenesis and continue to grow and divide during the larval stages—unlike most other cells of the larva, which have differentiated to perform specialized functions and grow without further cell division. At metamorphosis, the larva forms a pupa, inside which the larval tissues are reabsorbed and the imaginal tissues undergo extensive morphogenetic movements to form adult structures.

Developmental plasticity edit

Biotic and abiotic factors experienced during development will affect developmental resource allocation leading to phenotypic variation, also referred to as developmental plasticity.[85][86] As in all insects,[86] environmental factors can influence several aspects of development in Drosophila melanogaster.[87][88] Fruit flies reared under a hypoxia treatment experience decreased thorax length, while hyperoxia produces smaller flight muscles, suggesting negative developmental effects of extreme oxygen levels.[89] Circadian rhythms are also subject to developmental plasticity. Light conditions during development affect daily activity patterns in Drosophila melanogaster, where flies raised under constant dark or light are less active as adults than those raised under a 12-hour light/dark cycle.[90]

Temperature is one of the most pervasive factors influencing arthropod development. In Drosophila melanogaster temperature-induced developmental plasticity can be beneficial and/or detrimental.[91][92] Most often lower developmental temperatures reduce growth rates which influence many other physiological factors.[93] For example, development at 25 °C increases walking speed, thermal performance breadth, and territorial success, while development at 18 °C increases body mass, wing size, all of which are tied to fitness.[88][91] Moreover, developing at certain low temperatures produces proportionally large wings which improve flight and reproductive performance at similarly low temperatures (See acclimation).[94]

While certain effects of developmental temperature, like body size, are irreversible in ectotherms, others can be reversible.[86][95] When Drosophila melanogaster develop at cold temperatures they will have greater cold tolerance, but if cold-reared flies are maintained at warmer temperatures their cold tolerance decreases and heat tolerance increases over time.[95][96] Because insects typically only mate in a specific range of temperatures, their cold/heat tolerance is an important trait in maximizing reproductive output.[97]

While the traits described above are expected to manifest similarly across sexes, developmental temperature can also produce sex-specific effects in D. melanogaster adults.

  • Females- Ovariole number is significantly affected by developmental temperature in D. melanogaster.[98] Egg size is also affected by developmental temperature, and exacerbated when both parents develop at warm temperatures (See Maternal effect).[91] Under stressful temperatures, these structures will develop to smaller ultimate sizes and decrease a female's reproductive output.[98][91] Early fecundity (total eggs laid in first 10 days post-eclosion) is maximized when reared at 25 °C (versus 17 °C and 29 °C) regardless of adult temperature.[99] Across a wide range of developmental temperatures, females tend to have greater heat tolerance than males.[100]
  • Males- Stressful developmental temperatures will cause sterility in D. melanogaster males; although the upper temperature limit can be increased by maintaining strains at high temperatures (See acclimation).[92] Male sterility can be reversible if adults are returned to an optimal temperature after developing at stressful temperatures.[101] Male flies are smaller and more successful at defending food/oviposition sites when reared at 25 °C versus 18 °C; thus smaller males will have increased mating success and reproductive output.[88]

Sex determination edit

Drosophila flies have both X and Y chromosomes, as well as autosomes. Unlike humans, the Y chromosome does not confer maleness; rather, it encodes genes necessary for making sperm. Sex is instead determined by the ratio of X chromosomes to autosomes.[102] Furthermore, each cell "decides" whether to be male or female independently of the rest of the organism, resulting in the occasional occurrence of gynandromorphs.

X Chromosomes Autosomes Ratio of X:A Sex
XXXX AAAA 1 Normal Female
XXX AAA 1 Normal Female
XXY AA 1 Normal Female
XXYY AA 1 Normal Female
XX AA 1 Normal Female
XY AA 0.50 Normal Male
X AA 0.50 Normal Male (sterile)
XXX AA 1.50 Metafemale
XXXX AAA 1.33 Metafemale
XX AAA 0.66 Intersex
X AAA 0.33 Metamale

Three major genes are involved in determination of Drosophila sex. These are sex-lethal, sisterless, and deadpan. Deadpan is an autosomal gene which inhibits sex-lethal, while sisterless is carried on the X chromosome and inhibits the action of deadpan. An AAX cell has twice as much deadpan as sisterless, so sex-lethal will be inhibited, creating a male. However, an AAXX cell will produce enough sisterless to inhibit the action of deadpan, allowing the sex-lethal gene to be transcribed to create a female.

Later, control by deadpan and sisterless disappears and what becomes important is the form of the sex-lethal gene. A secondary promoter causes transcription in both males and females. Analysis of the cDNA has shown that different forms are expressed in males and females. Sex-lethal has been shown to affect the splicing of its own mRNA. In males, the third exon is included which encodes a stop codon, causing a truncated form to be produced. In the female version, the presence of sex-lethal causes this exon to be missed out; the other seven amino acids are produced as a full peptide chain, again giving a difference between males and females.[103]

Presence or absence of functional sex-lethal proteins now go on to affect the transcription of another protein known as doublesex. In the absence of sex-lethal, doublesex will have the fourth exon removed and be translated up to and including exon 6 (DSX-M[ale]), while in its presence the fourth exon which encodes a stop codon will produce a truncated version of the protein (DSX-F[emale]). DSX-F causes transcription of Yolk proteins 1 and 2 in somatic cells, which will be pumped into the oocyte on its production.

Immunity edit

The D. melanogaster immune system can be divided into two responses: humoral and cell-mediated. The former is a systemic response mediated in large part through the toll and Imd pathways, which are parallel systems for detecting microbes. Other pathways including the stress response pathways JAK-STAT and P38, nutritional signalling via FOXO, and JNK cell death signalling are all involved in key physiological responses to infection. D. melanogaster has an organ called the "fat body", which is analogous to the human liver. The fat body is the primary secretory organ and produces key immune molecules upon infection, such as serine proteases and antimicrobial peptides (AMPs). AMPs are secreted into the hemolymph and bind infectious bacteria and fungi, killing them by forming pores in their cell walls or inhibiting intracellular processes. The cellular immune response instead refers to the direct activity of blood cells (hemocytes) in Drosophila, which are analogous to mammalian monocytes/macrophages. Hemocytes also possess a significant role in mediating humoral immune responses such as the melanization reaction.[104]

The immune response to infection can involve up to 2,423 genes, or 13.7% of the genome. Although the fly's transcriptional response to microbial challenge is highly specific to individual pathogens, Drosophila differentially expresses a core group of 252 genes upon infection with most bacteria. This core group of genes is associated with gene ontology categories such as antimicrobial response, stress response, secretion, neuron-like, reproduction, and metabolism among others.[105][106] Drosophila also possesses several immune mechanisms to both shape the microbiota and prevent excessive immune responses upon detection of microbial stimuli. For instance, secreted PGRPs with amidase activity scavenge and degrade immunostimulatory DAP-type PGN in order to block Imd activation.[107]

Unlike mammals, Drosophila have innate immunity but lack an adaptive immune response. However, the core elements of this innate immune response are conserved between humans and fruit flies. As a result, the fruit fly offers a useful model of innate immunity for disentangling genetic interactions of signalling and effector function, as flies do not have to contend with interference of adaptive immune mechanisms that could confuse results. Various genetic tools, protocols, and assays make Drosophila a classical model for studying the innate immune system,[108] which has even included immune research on the international space station.[109]

JAK-STAT signalling edit

Multiple elements of the Drosophila JAK-STAT signalling pathway bear direct homology to human JAK-STAT pathway genes. JAK-STAT signalling is induced upon various organismal stresses such as heat stress, dehydration, or infection. JAK-STAT induction leads to the production of a number of stress response proteins including Thioester-containing proteins (TEPs),[110] Turandots,[111] and the putative antimicrobial peptide Listericin.[112] The mechanisms through which many of these proteins act is still under investigation. For instance, the TEPs appear to promote phagocytosis of Gram-positive bacteria and the induction of the toll pathway. As a consequence, flies lacking TEPs are susceptible to infection by toll pathway challenges.[110]

Drosophila hemocytes (green) engulfing Escherichia coli bacteria (red)

The cellular response to infection edit

Circulating hemocytes are key regulators of infection. This has been demonstrated both through genetic tools to generate flies lacking hemocytes, or through injecting microglass beads or lipid droplets that saturate hemocyte ability to phagocytose a secondary infection.[113][114] Flies treated like this fail to phagocytose bacteria upon infection, and are correspondingly susceptible to infection.[115] These hemocytes derive from two waves of hematopoiesis, one occurring in the early embryo and one occurring during development from larva to adult.[116] However Drosophila hemocytes do not renew over the adult lifespan, and so the fly has a finite number of hemocytes that decrease over the course of its lifespan.[117] Hemocytes are also involved in regulating cell-cycle events and apoptosis of aberrant tissue (e.g. cancerous cells) by producing Eiger, a tumor necrosis factor signalling molecule that promotes JNK signalling and ultimately cell death and apoptosis.[118]

Behavioral genetics and neuroscience edit

In 1971, Ron Konopka and Seymour Benzer published "Clock mutants of Drosophila melanogaster", a paper describing the first mutations that affected an animal's behavior. Wild-type flies show an activity rhythm with a frequency of about a day (24 hours). They found mutants with faster and slower rhythms, as well as broken rhythms—flies that move and rest in random spurts. Work over the following 30 years has shown that these mutations (and others like them) affect a group of genes and their products that form a biochemical or biological clock. This clock is found in a wide range of fly cells, but the clock-bearing cells that control activity are several dozen neurons in the fly's central brain.

Since then, Benzer and others have used behavioral screens to isolate genes involved in vision, olfaction, audition, learning/memory, courtship, pain, and other processes, such as longevity.

Following the pioneering work of Alfred Henry Sturtevant[119] and others, Benzer and colleagues[46] used sexual mosaics to develop a novel fate mapping technique. This technique made it possible to assign a particular characteristic to a specific anatomical location. For example, this technique showed that male courtship behavior is controlled by the brain.[46] Mosaic fate mapping also provided the first indication of the existence of pheromones in this species.[120] Males distinguish between conspecific males and females and direct persistent courtship preferentially toward females thanks to a female-specific sex pheromone which is mostly produced by the female's tergites.

The first learning and memory mutants (dunce, rutabaga, etc.) were isolated by William "Chip" Quinn while in Benzer's lab, and were eventually shown to encode components of an intracellular signaling pathway involving cyclic AMP, protein kinase A, and a transcription factor known as CREB. These molecules were shown to be also involved in synaptic plasticity in Aplysia and mammals.[121]

The Nobel Prize in Physiology or Medicine for 2017 was awarded to Jeffrey C. Hall, Michael Rosbash, Michael W. Young for their works using fruit flies in understanding the "molecular mechanisms controlling the circadian rhythm".[122]

Male flies sing to the females during courtship using their wings to generate sound, and some of the genetics of sexual behavior have been characterized. In particular, the fruitless gene has several different splice forms, and male flies expressing female splice forms have female-like behavior and vice versa. The TRP channels nompC, nanchung, and inactive are expressed in sound-sensitive Johnston's organ neurons and participate in the transduction of sound.[123][124] Mutating the Genderblind gene, also known as CG6070, alters the sexual behavior of Drosophila, turning the flies bisexual.[125]

Flies use a modified version of Bloom filters to detect novelty of odors, with additional features including similarity of novel odor to that of previously experienced examples, and time elapsed since previous experience of the same odor.[126]

Aggression edit

As with most insects, aggressive behaviors between male flies commonly occur in the presence of courting a female and when competing for resources. Such behaviors often involve raising wings and legs towards the opponent and attacking with the whole body.[127] Thus, it often causes wing damage, which reduces their fitness by removing their ability to fly and mate.[128]

Acoustic communication edit

In order for aggression to occur, male flies produce sounds to communicate their intent. A 2017 study found that songs promoting aggression contain pulses occurring at longer intervals.[129] RNA sequencing from fly mutants displaying over-aggressive behaviors found more than 50 auditory-related genes (important for transient receptor potentials, Ca2+ signaling, and mechanoreceptor potentials) to be upregulated in the AB neurons located in Johnston's organ.[129] In addition, aggression levels were reduced when these genes were knocked out via RNA interference.[129] This signifies the major role of hearing as a sensory modality in communicating aggression.

Pheromone signaling edit

Other than hearing, another sensory modality that regulates aggression is pheromone signaling, which operates through either the olfactory system or the gustatory system depending on the pheromone.[130] An example is cVA, an anti-aphrodisiac pheromone used by males to mark females after copulation and to deter other males from mating.[131] This male-specific pheromone causes an increase in male-male aggression when detected by another male's gustatory system.[130] However, upon inserting a mutation that makes the flies irresponsive to cVA, no aggressive behaviors were seen.[132] This shows how there are multiple modalities for promoting aggression in flies.

Competition for food edit

Specifically, when competing for food, aggression occurs based on amount of food available and is independent of any social interactions between males.[133] Specifically, sucrose was found to stimulate gustatory receptor neurons, which was necessary to stimulate aggression.[133] However, once the amount of food becomes greater than a certain amount, the competition between males lowers.[133] This is possibly due to an over-abundance of food resources. On a larger scale, food was found to determine the boundaries of a territory since flies were observed to be more aggressive at the food's physical perimeter.

Effect of sleep deprivation edit

However, like most behaviors requiring arousal and wakefulness, aggression was found to be impaired via sleep deprivation. Specifically, this occurs through the impairment of Octopamine and dopamine signaling, which are important pathways for regulating arousal in insects.[134][135] Due to reduced aggression, sleep-deprived male flies were found to be disadvantaged at mating compared to normal flies.[135] However, when octopamine agonists were administered upon these sleep-deprived flies, aggression levels were seen to be increased and sexual fitness was subsequently restored.[135] Therefore, this finding implicates the importance of sleep in aggression between male flies.

Vision edit

 
Stereo images of the eye

The compound eye of the fruit fly contains 760 unit eyes or ommatidia, and are one of the most advanced among insects. Each ommatidium contains eight photoreceptor cells (R1-8), support cells, pigment cells, and a cornea. Wild-type flies have reddish pigment cells, which serve to absorb excess blue light so the fly is not blinded by ambient light. Eye color genes regulate cellular vesicular transport. The enzymes needed for pigment synthesis are then transported to the cell's pigment granule, which holds pigment precursor molecules.[59]

Each photoreceptor cell consists of two main sections, the cell body and the rhabdomere. The cell body contains the nucleus, while the 100-μm-long rhabdomere is made up of toothbrush-like stacks of membrane called microvilli. Each microvillus is 1–2 μm in length and about 60 nm in diameter.[136] The membrane of the rhabdomere is packed with about 100 million opsin molecules, the visual protein that absorbs light. The other visual proteins are also tightly packed into the microvilli, leaving little room for cytoplasm.

Opsins and spectral sensitivity edit

 
The arrangement of the photoreceptor cells in an pale and yellow ommatidia of Drosophila melanogaster: The top row shows two of the six outer photoreceptor cells (R1-R6) and the inner R7 and R8 cells. The bottom row shows the different opsins (Rh1, Rh3, Rh4, Rh5, and Rh6) the cells express. Figure from Sharkey et al. (2020).[137]
 
Expression of the opsin Rh1 in the photoreceptor cells R1-R6

The genome of Drosophila encodes seven opsins,[138] five of those are expressed in the omatidia of the eye. The photoreceptor cells R1-R6 express the opsin Rh1,[139] which absorbs maximally blue light (around 480 nm),[140][141][142] however the R1-R6 cells cover a broader range of the spectrum than an opsin would allow due to a sensitising pigment[143][144] that adds two sensitivity maxima in the UV-range (355 and 370 nm).[142] The R7 cells come in two types with yellow and pale rhabdomeres (R7y and R7p).[145][146] The pale R7p cells express the opsin Rh3,[147][148] which maximally absorbs UV-light (345 nm).[149] The R7p cells are strictly paired with the R8p cells that express Rh5,[148] which maximally absorbs violet light (437 nm).[142] The other, the yellow R7y cells express a blue-absorbing screening pigment[145] and the opsin Rh4,[150] which maximally absorbs UV-light (375 nm).[149] The R7y cells are strictly paired with R8y cells that express Rh6,[151] which maximally absorbs UV-light (508 nm).[142] In a subset of omatidia both R7 and R8 cells express the opsin Rh3.[148]

However, these absorption maxima of the opsins where measured in white eyed flies without screening pigments (Rh3-Rh6),[149][142] or from the isolated opsin directly (Rh1).[140] Those pigments reduce the light that reaches the opsins depending on the wavelength. Thus in fully pigmented flies, the effective absorption maxima of opsins differs and thus also the sensitivity of their photoreceptor cells. With screening pigment, the opsin Rh3 is short wave shifted from 345 nm[b] to 330 nm and Rh4 from 375 nm to 355 nm. Whether screening pigment is present does not make a practical difference for the opsin Rh5 (435 nm and 437 nm), while the opsin R6 is long wave shifted by 92 nm from 508 nm to 600 nm.[137]

Additionally of the opsins of the eye, Drosophila has two more opsins: The ocelli express the opsin Rh2,[152][153] which maximally absorbs violet light (~420 nm).[153] And the opsin Rh7, which maximally absorbs UV-light (350 nm) with an unusually long wavelength tail up to 500 nm. The long tail disappears if a lysine at position 90 is replaced by glutamic acid. This mutant then absorbs maximally violet light (450 nm).[154] The opsin Rh7 entrains with cryptochrome the circadian rhythm of Drosophila to the day-night-cycle in the central pacemaker neurons.[155]

Each Drosophila opsin binds the carotenoid chromophore 11-cis-3-hydroxyretinal via a lysine.[156][157] This lysine is conserved in almost all opsins, only a few opsins have lost it during evolution.[158] Opsins without it are not light sensitive.[159][160][161] In particular, the Drosophila opsins Rh1, Rh4, and Rh7 function not only as photoreceptors, but also as chemoreceptors for aristolochic acid. These opsins still have the lysine like other opsins. However, if it is replaced by an arginine in Rh1, then Rh1 loses light sensitivity but still responds to aristolochic acid. Thus, the lysine is not needed for Rh1 to function as chemoreceptor.[160]

Phototransduction edit

As in vertebrate vision, visual transduction in invertebrates occurs via a G protein-coupled pathway. However, in vertebrates, the G protein is transducin, while the G protein in invertebrates is Gq (dgq in Drosophila). When rhodopsin (Rh) absorbs a photon of light its chromophore, 11-cis-3-hydroxyretinal, is isomerized to all-trans-3-hydroxyretinal. Rh undergoes a conformational change into its active form, metarhodopsin. Metarhodopsin activates Gq, which in turn activates a phospholipase Cβ (PLCβ) known as NorpA.[162]

PLCβ hydrolyzes phosphatidylinositol (4,5)-bisphosphate (PIP2), a phospholipid found in the cell membrane, into soluble inositol triphosphate (IP3) and diacylglycerol (DAG), which stays in the cell membrane. DAG, a derivative of DAG, or PIP2 depletion cause a calcium-selective ion channel known as transient receptor potential (TRP) to open and calcium and sodium flows into the cell.[163] IP3 is thought to bind to IP3 receptors in the subrhabdomeric cisternae, an extension of the endoplasmic reticulum, and cause release of calcium, but this process does not seem to be essential for normal vision.[162]

Calcium binds to proteins such as calmodulin (CaM) and an eye-specific protein kinase C (PKC) known as InaC. These proteins interact with other proteins and have been shown to be necessary for shut off of the light response. In addition, proteins called arrestins bind metarhodopsin and prevent it from activating more Gq. A sodium-calcium exchanger known as CalX pumps the calcium out of the cell. It uses the inward sodium gradient to export calcium at a stoichiometry of 3 Na+/ 1 Ca++.[164]

TRP, InaC, and PLC form a signaling complex by binding a scaffolding protein called InaD. InaD contains five binding domains called PDZ domain proteins, which specifically bind the C termini of target proteins. Disruption of the complex by mutations in either the PDZ domains or the target proteins reduces the efficiency of signaling. For example, disruption of the interaction between InaC, the protein kinase C, and InaD results in a delay in inactivation of the light response.

Unlike vertebrate metarhodopsin, invertebrate metarhodopsin can be converted back into rhodopsin by absorbing a photon of orange light (580 nm).

About two-thirds of the Drosophila brain is dedicated to visual processing.[165] Although the spatial resolution of their vision is significantly worse than that of humans, their temporal resolution is around 10 times better.

Grooming edit

Drosophila are known to exhibit grooming behaviors that are executed in a predictable manner. Drosophila consistently begin a grooming sequence by using their front legs to clean the eyes, then the head and antennae. Using their hind legs, Drosophila proceed to groom their abdomen, and finally the wings and thorax. Throughout this sequence, Drosophila periodically rub their legs together to get rid of excess dust and debris that accumulates during the grooming process.[166]

Grooming behaviors have been shown to be executed in a suppression hierarchy. This means that grooming behaviors that occur at the beginning of the sequence prevent those that come later in the sequence from occurring simultaneously, as the grooming sequence consists of mutually exclusive behaviors.[167][168] This hierarchy does not prevent Drosophila from returning to grooming behaviors that have already been accessed in the grooming sequence.[167] The order of grooming behaviors in the suppression hierarchy is thought to be related to the priority of cleaning a specific body part. For example, the eyes and antennae are likely executed early on in the grooming sequence to prevent debris from interfering with the function of D. melanogaster's sensory organs.[167][168]

Walking edit

 
Top view of a walking Drosophila (left) with legs tracked with DeepLabCut[169] (right)

Like many other hexapod insects, Drosophila typically walk using a tripod gait.[170] This means that three of the legs swing together while the other three remain stationary, or in stance. Specifically, the middle leg moves in phase with the contralateral front and hind legs. However, variability around the tripod configuration exists along a continuum, meaning that flies do not exhibit distinct transitions between different gaits.[171] At fast walking speeds, the walking configuration is mostly tripod (3 legs in stance), but at slower walking speeds, flies are more likely to have four (tetrapod) or five legs in stance (wave).[172][173] These transitions may help to optimize static stability.[174] Because flies are so small, inertial forces are negligible compared with the elastic forces of their muscles and joints or the viscous forces of the surrounding air.[175]

Flight edit

Flies fly via straight sequences of movement interspersed by rapid turns called saccades.[176] During these turns, a fly is able to rotate 90° in less than 50 milliseconds.[176]

Characteristics of Drosophila flight may be dominated by the viscosity of the air, rather than the inertia of the fly body, but the opposite case with inertia as the dominant force may occur.[176] However, subsequent work showed that while the viscous effects on the insect body during flight may be negligible, the aerodynamic forces on the wings themselves actually cause fruit flies' turns to be damped viscously.[177]

Connectome edit

Drosophila is one of the few animals (C. elegans being another) where detailed neural circuits (a connectome) are available.

A high-level connectome, at the level of brain compartments and interconnecting tracts of neurons, exists for the full fly brain.[178] A version of this is available online.[179]

Detailed circuit-level connectomes exist for the lamina[180][181] and a medulla[182] column, both in the visual system of the fruit fly, and the alpha lobe of the mushroom body.[183]

In May 2017 a paper published in bioRxiv presented an electron microscopy image stack of the whole adult female brain at synaptic resolution. The volume is available for sparse tracing of selected circuits.[184][185] Since then, multiple datasets have been collected including a dense connectome of half the central brain of Drosophila in 2020,[186][187] and a dense connectome of the entire female adult nerve cord in 2021.[188] Generally, these datasets are acquired by sectioning the tissue (e.g. the brain) into thin sections (on order of ten or hundreds of nanometers). Each section is then imaged using an electron microscope and these images are stitched and aligned together to create a 3D image volume. The methods used in reconstruction and initial analysis of the such datasets followed.[189] Due to advancements in deep learning, automated methods for image segmentation have made large scale reconstruction providing dense reconstructions of all the neurites within the volume.[190] Furthermore, the resolution of electron microscopy illuminates ultrastructural variations between neurons as well as the location of individual synapses, thereby providing a wiring diagram of synaptic connectivity between all neurites within the given dataset.

In 2023, the complete map of a Drosophila larval brain at the synapse level, and an analysis of its architecture was published. The larval brain consists of 3016 neurons and 548,000 synaptic sites,[191] whereas the adult brain has about 150,000 neurons and 150 million synapses.

Misconceptions edit

Drosophila is sometimes referred to as a pest due to its tendency to live in human settlements where fermenting fruit is found. Flies may collect in homes, restaurants, stores, and other locations.[13] The name and behavior of this species of fly have led to the misconception that it is a biological security risk in Australia and elsewhere. While other "fruit fly" species do pose a risk, D. melanogaster is attracted to fruit that is already rotting, rather than causing fruit to rot.[192][193]

See also edit

Notes edit

  1. ^ "Vinegar fly" is preferred by a handful of recent publications as being a more accurate description than "fruit fly".[2][3][4]
  2. ^ Sharkey et al.[137] give the absorption maximum of Rh3 as 334 nm in their result section. However, in the introduction and the material and methods section they give it as 345 nm. For both values, they cite Feiler et al., who reported 345 nm only.[149] Therefore, this seems to be a mistake and they probably meant there 345 nm, too.

References edit

  1. ^ Meigen JW (1830). (PDF) (in German). Schulz-Wundermann. Archived from the original (PDF) on February 9, 2012.
  2. ^ "Drosophila | insect genus". Encyclopedia Britannica. Retrieved October 30, 2021.
  3. ^ "Vinegar Flies". Penn State Extension. Retrieved October 30, 2021.
  4. ^ a b Green MM (September 2002). "It really is not a fruit fly". Genetics. 162 (1): 1–3. doi:10.1093/genetics/162.1.1. PMC 1462251. PMID 12242218.
  5. ^ Schilthuizen, Menno (April 28, 2015). "Semen's Chemical Cocktail Can Hijack a Mate's Brain". Discover. Retrieved September 11, 2023. Even in the ejaculate of the lowly banana fly Drosophila melanogaster, researchers have identified no fewer than 133 different kinds of proteins.
  6. ^
  7. ^ Holden B (January 1, 2015). Charles W. Woodworth: The Remarkable Life of U.C.'s First Entomologist (1st ed.). Brian Holden Publishing. pp. 135–137. ISBN 978-0-9864105-3-6.
  8. ^ "Nobel Prizes". The Guardian. October 7, 2017.
  9. ^ "FruitFly-ResearchGate".
  10. ^ a b Sang JH, Reeve EC (June 23, 2001). "Drosophila melanogaster: The Fruit Fly". Encyclopedia of genetics. USA: Fitzroy Dearborn Publishers, I. p. 157. ISBN 978-1-884964-34-3. Retrieved July 1, 2009.
  11. ^ Baudry E, Viginier B, Veuille M (August 2004). "Non-African populations of Drosophila melanogaster have a unique origin". Molecular Biology and Evolution. 21 (8): 1482–91. doi:10.1093/molbev/msh089. PMID 15014160.
  12. ^ a b Markow TA (June 2015). "The secret lives of Drosophila flies". eLife. 4. doi:10.7554/eLife.06793. PMC 4454838. PMID 26041333.
  13. ^ a b "Vinegar Flies, Drosophila species, Family: Drosophilidae". Department of Entomology, College of Agricultural Sciences, Pennsylvania State University. 2017. Retrieved July 20, 2017.
  14. ^ a b Ewart GD, Howells AJ (January 1, 1998). "ABC transporters involved in transport of eye pigment precursors in Drosophila melanogaster". ABC Transporters: Biochemical, Cellular, and Molecular Aspects. Methods in Enzymology. Vol. 292. Academic Press. pp. 213–24. doi:10.1016/S0076-6879(98)92017-1. ISBN 978-0-12-182193-7. PMID 9711556.
  15. ^ . Genetics Society of America. 2009. Archived from the original on August 15, 2009. Retrieved August 11, 2009.
  16. ^ Yuzuki, Keven; Tidon, Rosana (2020). "Identification key for drosophilid species (Diptera, Drosophilidae) exotic to the Neotropical Region and occurring in Brazil". Revista Brasileira de Entomologia. 64 (1). doi:10.1590/1806-9665-rbent-2019-100. ISSN 1806-9665. S2CID 211570766.
  17. ^ Miller, M. E.; Marshall, S. A.; Grimaldi, D. A. (2017). "A Review of the Species of Drosophila (Diptera: Drosophilidae) and Genera of Drosophilidae of Northeastern North America". Canadian Journal of Arthropod Identification. 31. doi:10.3752/cjai.2017.31.
  18. ^ . Animal Diversity Web. 2000. Archived from the original on November 30, 2014. Retrieved August 11, 2009.
  19. ^ Linford NJ, Bilgir C, Ro J, Pletcher SD (January 2013). "Measurement of lifespan in Drosophila melanogaster". Journal of Visualized Experiments (71). doi:10.3791/50068. PMC 3582515. PMID 23328955.
  20. ^ a b c d e f g Ashburner M, Thompson JN (1978). "The laboratory culture of Drosophila". In Ashburner M, Wright TRF (ed.). The genetics and biology of Drosophila. Vol. 2A. Academic Press. 1–81.
  21. ^ a b c d e f g Ashburner M, Golic KG, Hawley RS (2005). Drosophila: A Laboratory Handbook (2nd ed.). Cold Spring Harbor Laboratory Press. pp. 162–4. ISBN 978-0-87969-706-8.
  22. ^ Bloomington Drosophila Stock Center at Indiana University: Basic Methods of Culturing Drosophila 2006-09-01 at the Wayback Machine
  23. ^ a b Chiang HC, Hodson AC (1950). "An analytical study of population growth in Drosophila melanogaster". Ecological Monographs. 20 (3): 173–206. Bibcode:1950EcoM...20..173C. doi:10.2307/1948580. JSTOR 1948580.
  24. ^ Bakker K (1961). "An analysis of factors which determine success in competition for food among larvae of Drosophila melanogaster". Archives Néerlandaises de Zoologie. 14 (2): 200–281. doi:10.1163/036551661X00061. S2CID 85129022.
  25. ^ Fernández-Moreno MA, Farr CL, Kaguni LS, Garesse R (2007). "Drosophila melanogaster as a Model System to Study Mitochondrial Biology". Mitochondria. Methods in Molecular Biology. Vol. 372. pp. 33–49. doi:10.1007/978-1-59745-365-3_3. ISBN 978-1-58829-667-2. PMC 4876951. PMID 18314716.
  26. ^ Blum JE, Fischer CN, Miles J, Handelsman J (November 2013). "Frequent replenishment sustains the beneficial microbiome of Drosophila melanogaster". mBio. 4 (6): e00860-13. doi:10.1128/mBio.00860-13. PMC 3892787. PMID 24194543.
  27. ^ Cook R, Connolly K (1973). "Rejection Responses by Female Drosophila melanogaster: Their Ontogeny, Causality and Effects upon the Behaviour of the Courting Male". Behaviour. 44 (1/2): 142–166. doi:10.1163/156853973x00364. JSTOR 4533484. S2CID 85393769.
  28. ^ Houot B, Svetec N, Godoy-Herrera R, Ferveur JF (July 2010). "Effect of laboratory acclimation on the variation of reproduction-related characters in Drosophila melanogaster". The Journal of Experimental Biology. 213 (Pt 13): 2322–31. doi:10.1242/jeb.041566. PMID 20543131.
  29. ^ Gilbert SF (2006). . Developmental Biology (8th ed.). Sinauer Associates. ISBN 978-0-87893-250-4. Archived from the original on February 7, 2007.
  30. ^ a b c Price CS, Dyer KA, Coyne JA (July 1999). "Sperm competition between Drosophila males involves both displacement and incapacitation". Nature. 400 (6743): 449–52. Bibcode:1999Natur.400..449P. doi:10.1038/22755. PMID 10440373. S2CID 4393369.
  31. ^ a b c d "Fruit fly research may reveal what happens in female brains during courtship, mating". Retrieved October 5, 2014.
  32. ^ Meiselman M, Lee SS, Tran RT, Dai H, Ding Y, Rivera-Perez C, et al. (May 2017). "Drosophila melanogaster". Proceedings of the National Academy of Sciences of the United States of America. 114 (19): E3849–E3858. doi:10.1073/pnas.1620760114. PMC 5441734. PMID 28439025.
  33. ^ Moshitzky P, Fleischmann I, Chaimov N, Saudan P, Klauser S, Kubli E, Applebaum SW (1996). "Sex-peptide activates juvenile hormone biosynthesis in the Drosophila melanogaster corpus allatum". Archives of Insect Biochemistry and Physiology. 32 (3–4): 363–74. doi:10.1002/(SICI)1520-6327(1996)32:3/4<363::AID-ARCH9>3.0.CO;2-T. PMID 8756302.
  34. ^ Carnes MU, Campbell T, Huang W, Butler DG, Carbone MA, Duncan LH, et al. (2015). "The Genomic Basis of Postponed Senescence in Drosophila melanogaster". PLOS ONE. 10 (9): e0138569. Bibcode:2015PLoSO..1038569C. doi:10.1371/journal.pone.0138569. PMC 4574564. PMID 26378456.
  35. ^ Cassidy D, Epiney DG, Salameh C, Zhou LT, Salomon RN, Schirmer AE, et al. (November 2019). "Evidence for premature aging in a Drosophila model of Werner syndrome". Experimental Gerontology. 127: 110733. doi:10.1016/j.exger.2019.110733. PMC 6935377. PMID 31518666.
  36. ^ Pitnick S (1996). "Investment in testes and the cost of making long sperm in Drosophila". American Naturalist. 148: 57–80. doi:10.1086/285911. S2CID 83654824.
  37. ^ Dagaeff AC, Pocheville A, Nöbel S, Loyau A, Isabel G, Danchin E (2016). "Drosophila mate copying correlates with atmospheric pressure in a speed learning situation". Animal Behaviour. 121: 163–174. doi:10.1016/j.anbehav.2016.08.022.
  38. ^ Dukas R (2004). "Male fruit flies learn to avoid interspecific courtship". Behavioral Ecology. 15 (4): 695–698. doi:10.1093/beheco/arh068.
  39. ^ Saleem S, Ruggles PH, Abbott WK, Carney GE (2014). "Sexual experience enhances Drosophila melanogaster male mating behavior and success". PLOS ONE. 9 (5): e96639. Bibcode:2014PLoSO...996639S. doi:10.1371/journal.pone.0096639. PMC 4013029. PMID 24805129.
  40. ^ a b von Haartman L (1951). "Successive Polygamy". Behaviour. 3 (1): 256–273. doi:10.1163/156853951x00296.
  41. ^ a b c d e f g Vartak VR, Varma V, Sharma VK (February 2015). "Effects of polygamy on the activity/rest rhythm of male fruit flies Drosophila melanogaster". Die Naturwissenschaften. 102 (1–2): 1252. Bibcode:2015SciNa.102....3V. doi:10.1007/s00114-014-1252-5. PMID 25604736. S2CID 7529509.
  42. ^ a b Bateman AJ (December 1948). "Intra-sexual selection in Drosophila". Heredity. 2 (Pt. 3): 349–68. doi:10.1038/hdy.1948.21. PMID 18103134.
  43. ^ a b Pierce BA (2004). Genetics: A Conceptual Approach (2nd ed.). W. H. Freeman. ISBN 978-0-7167-8881-2.
  44. ^ Kilbey BJ, MacDonald DJ, Auerbach C, Sobels FH, Vogel EW (June 1981). "The use of Drosophila melanogaster in tests for environmental mutagens". Mutation Research. 85 (3): 141–6. doi:10.1016/0165-1161(81)90029-7. PMID 6790982.
  45. ^ a b Adams MD; Celniker SE; Holt RA; Evans CA; Gocayne JD; Amanatides PG; et al. (March 2000). "The genome sequence of Drosophila melanogaster". Science. 287 (5461): 2185–95. Bibcode:2000Sci...287.2185.. CiteSeerX 10.1.1.549.8639. doi:10.1126/science.287.5461.2185. PMID 10731132.
  46. ^ a b c Hotta Y, Benzer S (December 1972). "Mapping of behaviour in Drosophila mosaics". Nature. 240 (5383): 527–35. Bibcode:1972Natur.240..527H. doi:10.1038/240527a0. PMID 4568399. S2CID 4181921.
  47. ^ Meiers S (2018). "Exploiting emerging DNA sequencing technologies to study genomic rearrangements". archiv.ub.uni-heidelberg.de. doi:10.11588/heidok.00024506. Retrieved June 28, 2021.
  48. ^ Azpiazu N, Frasch M (July 1993). "tinman and bagpipe: two homeo box genes that determine cell fates in the dorsal mesoderm of Drosophila". Genes & Development. 7 (7B): 1325–40. doi:10.1101/gad.7.7b.1325. PMID 8101173.
  49. ^ Stern DL, Frankel N (December 2013). "The structure and evolution of cis-regulatory regions: the shavenbaby story". Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences. 368 (1632): 20130028. doi:10.1098/rstb.2013.0028. PMC 3826501. PMID 24218640.
  50. ^ a b Phillips AM, Smart R, Strauss R, Brembs B, Kelly LE (May 2005). "The Drosophila black enigma: the molecular and behavioural characterization of the black1 mutant allele" (PDF). Gene. 351: 131–42. doi:10.1016/j.gene.2005.03.013. PMID 15878647.
  51. ^ a b "FlyBase Gene Report: Dmel\b". flybase.org. Retrieved March 26, 2019.
  52. ^ Sherald AF (September 1981). "Intergenic suppression of the black mutation of Drosophila melanogaster". Molecular & General Genetics. 183 (1): 102–6. doi:10.1007/bf00270146. PMID 6799739. S2CID 1210971.
  53. ^ Shoup JR (May 1966). "The development of pigment granules in the eyes of wild type and mutant Drosophila melanogaster". The Journal of Cell Biology. 29 (2): 223–49. doi:10.1083/jcb.29.2.223. PMC 2106902. PMID 5961338.
  54. ^ "TEACHER REFERENCE PAGES-FLY EYE PIGMENTS LAB" (PDF).
  55. ^ Morgan TH (March 1911). "The Origin of Nine Wing Mutations in Drosophila". Science. 33 (848): 496–9. Bibcode:1911Sci....33..496M. doi:10.1126/science.33.848.496. JSTOR 1638587. PMID 17774436.
  56. ^ "FlyBase Gene Report: Dmel\m". flybase.org. Retrieved March 26, 2019.
  57. ^ Bilousov OO, Katanaev VL, Demydov SV, Kozeretska IA (March–April 2013). "The downregulation of the miniature gene does not replicate miniature loss-of-function phenotypes in Drosophila melanogaster wing to the full extent". TSitologiia I Genetika. 47 (2): 77–81. PMID 23745366.
  58. ^ a b Kim J, Suh H, Kim S, Kim K, Ahn C, Yim J (September 2006). "Identification and characteristics of the structural gene for the Drosophila eye colour mutant sepia, encoding PDA synthase, a member of the omega class glutathione S-transferases". The Biochemical Journal. 398 (3): 451–60. doi:10.1042/BJ20060424. PMC 1559464. PMID 16712527.
  59. ^ a b c Grant P, Maga T, Loshakov A, Singhal R, Wali A, Nwankwo J, et al. (October 2016). "An Eye on Trafficking Genes: Identification of Four Eye Color Mutations in Drosophila". G3. 6 (10): 3185–3196. doi:10.1534/g3.116.032508. PMC 5068940. PMID 27558665.
  60. ^ Wiederrecht GJ, Brown GM (1984). "Purification and properties of the enzymes from Drosophila melanogaster that catalyze the conversion of dihydroneopterin triphosphate to the pyrimidodiazepine precursor of the drosopterins". J. Biol. Chem. 259 (22): 14121–7. doi:10.1016/S0021-9258(18)89865-9. PMID 6438092.
  61. ^ "Inheritance Patterns in Drosophila Melanogaster". Retrieved March 26, 2019.
  62. ^ a b Green MM (April 1952). "Mutant Isoalleles at the Vermilion Locus in Drosophila Melanogaster". Proceedings of the National Academy of Sciences of the United States of America. 38 (4): 300–5. Bibcode:1952PNAS...38..300G. doi:10.1073/pnas.38.4.300. PMC 1063551. PMID 16589094.
  63. ^ Oxenkrug GF (January 2010). "The extended life span of Drosophila melanogaster eye-color (white and vermilion) mutants with impaired formation of kynurenine". Journal of Neural Transmission. 117 (1): 23–26. doi:10.1007/s00702-009-0341-7. PMC 3013506. PMID 19941150.
  64. ^ Simon E, Faucheux C, Zider A, Thézé N, Thiébaud P (July 2016). "From vestigial to vestigial-like: the Drosophila gene that has taken wing". Development Genes and Evolution. 226 (4): 297–315. doi:10.1007/s00427-016-0546-3. PMID 27116603. S2CID 16651247.
  65. ^ Tomoyasu Y, Ohde T, Clark-Hachtel C (March 14, 2017). "What serial homologs can tell us about the origin of insect wings". F1000Research. 6: 268. doi:10.12688/f1000research.10285.1. PMC 5357031. PMID 28357056.
  66. ^ Williams JA, Bell JB, Carroll SB (December 1991). "Control of Drosophila wing and haltere development by the nuclear vestigial gene product". Genes & Development. 5 (12B): 2481–95. doi:10.1101/gad.5.12b.2481. PMID 1752439.
  67. ^ Green MM (January 2010). "2010: A century of Drosophila genetics through the prism of the white gene". Genetics. 184 (1): 3–7. doi:10.1534/genetics.109.110015. PMC 2815926. PMID 20061564.
  68. ^ Ferreiro MJ, Pérez C, Marchesano M, Ruiz S, Caputi A, Aguilera P, et al. (2018). "rosophila melanogaster White Mutant w1118 Undergo Retinal Degeneration". Frontiers in Neuroscience. 11: 732. doi:10.3389/fnins.2017.00732. PMC 5758589. PMID 29354028.
  69. ^ a b Xiao C, Qiu S, Robertson RM (August 2017). "The white gene controls copulation success in Drosophila melanogaster". Scientific Reports. 7 (1): 7712. Bibcode:2017NatSR...7.7712X. doi:10.1038/s41598-017-08155-y. PMC 5550479. PMID 28794482.
  70. ^ "Gene:Dmel\y". Flybase.org. The FlyBase Consortium. Retrieved March 26, 2019.
  71. ^ Wittkopp PJ, True JR, Carroll SB (April 2002). "Reciprocal functions of the Drosophila yellow and ebony proteins in the development and evolution of pigment patterns". Development. 129 (8): 1849–58. doi:10.1242/dev.129.8.1849. PMID 11934851.
  72. ^ a b Biessmann H (November 1985). "Molecular analysis of the yellow gene (y) region of Drosophila melanogaster". Proceedings of the National Academy of Sciences of the United States of America. 82 (21): 7369–73. Bibcode:1985PNAS...82.7369B. doi:10.1073/pnas.82.21.7369. PMC 391346. PMID 3933004.
  73. ^ "NCBI (National Center for Biotechnology Information) Genome Database". Retrieved November 30, 2011.
  74. ^ Halligan DL, Keightley PD (July 2006). "Ubiquitous selective constraints in the Drosophila genome revealed by a genome-wide interspecies comparison". Genome Research. 16 (7): 875–84. doi:10.1101/gr.5022906. PMC 1484454. PMID 16751341.
  75. ^ Carvalho AB (December 2002). "Origin and evolution of the Drosophila Y chromosome". Current Opinion in Genetics & Development. 12 (6): 664–8. doi:10.1016/S0959-437X(02)00356-8. PMID 12433579.
  76. ^ Gabaldón T, Koonin EV (May 2013). "Functional and evolutionary implications of gene orthology". Nature Reviews. Genetics. Nature Portfolio. 14 (5): 360–366. doi:10.1038/nrg3456. PMC 5877793. PMID 23552219.
  77. ^ "Background on Comparative Genomic Analysis". US National Human Genome Research Institute. December 2002.
  78. ^ Reiter LT, Potocki L, Chien S, Gribskov M, Bier E (June 2001). "A systematic analysis of human disease-associated gene sequences in Drosophila melanogaster". Genome Research. 11 (6): 1114–25. doi:10.1101/gr.169101. PMC 311089. PMID 11381037.
  79. ^ Chien S, Reiter LT, Bier E, Gribskov M (January 2002). "Homophila: human disease gene cognates in Drosophila". Nucleic Acids Research. 30 (1): 149–51. doi:10.1093/nar/30.1.149. PMC 99119. PMID 11752278.
  80. ^ Jaiswal M, Sandoval H, Zhang K, Bayat V, Bellen HJ (2012). "Probing mechanisms that underlie human neurodegenerative diseases in Drosophila". Annual Review of Genetics. 46: 371–96. doi:10.1146/annurev-genet-110711-155456. PMC 3663445. PMID 22974305.
  81. ^ Pick L (2017). Fly Models of Human Diseases. Volume 121 of Current Topics in Developmental Biology. Academic Press. ISBN 978-0-12-802905-3.
  82. ^ Buchon N, Silverman N, Cherry S (December 2014). "Immunity in Drosophila melanogaster--from microbial recognition to whole-organism physiology". Nature Reviews. Immunology. 14 (12): 796–810. doi:10.1038/nri3763. PMC 6190593. PMID 25421701.
  83. ^ Kaun KR, Devineni AV, Heberlein U (June 2012). "Drosophila melanogaster as a model to study drug addiction". Human Genetics. 131 (6): 959–75. doi:10.1007/s00439-012-1146-6. PMC 3351628. PMID 22350798.
  84. ^ a b Weigmann K, Klapper R, Strasser T, Rickert C, Technau G, Jäckle H, et al. (June 2003). "FlyMove--a new way to look at development of Drosophila". Trends in Genetics. 19 (6): 310–1. doi:10.1016/S0168-9525(03)00050-7. PMID 12801722.
  85. ^ West-Eberhard MJ (May 2005). "Developmental plasticity and the origin of species differences". Proceedings of the National Academy of Sciences of the United States of America. 102 (suppl 1): 6543–9. Bibcode:2005PNAS..102.6543W. doi:10.1073/pnas.0501844102. PMC 1131862. PMID 15851679.
  86. ^ a b c Abram PK, Boivin G, Moiroux J, Brodeur J (November 2017). "Behavioural effects of temperature on ectothermic animals: unifying thermal physiology and behavioural plasticity". Biological Reviews of the Cambridge Philosophical Society. 92 (4): 1859–1876. doi:10.1111/brv.12312. PMID 28980433. S2CID 9099834.
  87. ^ Gibert P, Huey RB, Gilchrist GW (January 2001). "Locomotor performance of Drosophila melanogaster: interactions among developmental and adult temperatures, age, and geography". Evolution; International Journal of Organic Evolution. 55 (1): 205–9. doi:10.1111/j.0014-3820.2001.tb01286.x. PMID 11263741. S2CID 2991855.
  88. ^ a b c Zamudio KR, Huey RB, Crill WD (1995). "Bigger isn't always better: body size, developmental and parental temperature and male territorial success in Drosophila melanogaster". Animal Behaviour. 49 (3): 671–677. doi:10.1016/0003-3472(95)80200-2. ISSN 0003-3472. S2CID 9124942.
  89. ^ Harrison JF, Waters JS, Biddulph TA, Kovacevic A, Klok CJ, Socha JJ (April 2018). "Developmental plasticity and stability in the tracheal networks supplying Drosophila flight muscle in response to rearing oxygen level". Journal of Insect Physiology. The limits of respiratory function: external and internal constraints on insect gas exchange. 106 (Pt 3): 189–198. doi:10.1016/j.jinsphys.2017.09.006. PMID 28927826.
  90. ^ Sheeba V, Chandrashekaran MK, Joshi A, Sharma VK (January 2002). "Developmental plasticity of the locomotor activity rhythm of Drosophila melanogaster". Journal of Insect Physiology. 48 (1): 25–32. doi:10.1016/S0022-1910(01)00139-1. PMID 12770129.
  91. ^ a b c d Crill WD, Huey RB, Gilchrist GW (June 1996). "Within- and Between-Generation Effects of Temperature on the Morphology and Physiology of Drosophila Melanogaster". Evolution; International Journal of Organic Evolution. 50 (3): 1205–1218. doi:10.2307/2410661. JSTOR 2410661. PMID 28565273.
  92. ^ a b David JR, Araripe LO, Chakir M, Legout H, Lemos B, Pétavy G, et al. (July 2005). "Male sterility at extreme temperatures: a significant but neglected phenomenon for understanding Drosophila climatic adaptations". Journal of Evolutionary Biology. 18 (4): 838–46. doi:10.1111/j.1420-9101.2005.00914.x. PMID 16033555. S2CID 23847613.
  93. ^ French V, Feast M, Partridge L (November 1998). "Body size and cell size in Drosophila: the developmental response to temperature". Journal of Insect Physiology. 44 (11): 1081–1089. doi:10.1016/S0022-1910(98)00061-4. PMID 12770407.
  94. ^ Frazier MR, Harrison JF, Kirkton SD, Roberts SP (July 2008). "Cold rearing improves cold-flight performance in Drosophila via changes in wing morphology". The Journal of Experimental Biology. 211 (Pt 13): 2116–22. doi:10.1242/jeb.019422. PMID 18552301.
  95. ^ a b Slotsbo S, Schou MF, Kristensen TN, Loeschcke V, Sørensen JG (September 2016). "Reversibility of developmental heat and cold plasticity is asymmetric and has long-lasting consequences for adult thermal tolerance". The Journal of Experimental Biology. 219 (Pt 17): 2726–32. doi:10.1242/jeb.143750. PMID 27353229.
  96. ^ Gilchrist GW, Huey RB (January 2001). "Parental and developmental temperature effects on the thermal dependence of fitness in Drosophila melanogaster". Evolution; International Journal of Organic Evolution. 55 (1): 209–14. doi:10.1111/j.0014-3820.2001.tb01287.x. PMID 11263742. S2CID 1329035.
  97. ^ Austin CJ, Moehring AJ (May 2013). "Optimal temperature range of a plastic species, Drosophila simulans". The Journal of Animal Ecology. 82 (3): 663–72. Bibcode:2013JAnEc..82..663A. doi:10.1111/1365-2656.12041. PMID 23360477.
  98. ^ a b Hodin J, Riddiford LM (October 2000). "Different mechanisms underlie phenotypic plasticity and interspecific variation for a reproductive character in drosophilids (Insecta: Diptera)". Evolution; International Journal of Organic Evolution. 54 (5): 1638–53. doi:10.1111/j.0014-3820.2000.tb00708.x. PMID 11108591. S2CID 6875815.
  99. ^ Klepsatel P, Girish TN, Dircksen H, Gáliková M (May 2019). "Drosophila is maximised by optimal developmental temperature". The Journal of Experimental Biology. 222 (Pt 10): jeb202184. doi:10.1242/jeb.202184. PMID 31064855.
  100. ^ Schou MF, Kristensen TN, Pedersen A, Karlsson BG, Loeschcke V, Malmendal A (February 2017). "Metabolic and functional characterization of effects of developmental temperature in Drosophila melanogaster". American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 312 (2): R211–R222. doi:10.1152/ajpregu.00268.2016. PMC 5336569. PMID 27927623.
  101. ^ Cohet Y, David J (January 1978). "Control of the adult reproductive potential by preimaginal thermal conditions: A study in Drosophila melanogaster". Oecologia. 36 (3): 295–306. Bibcode:1978Oecol..36..295C. doi:10.1007/BF00348055. PMID 28309916. S2CID 12465060.
  102. ^ Rideout EJ, Narsaiya MS, Grewal SS (December 2015). "The Sex Determination Gene transformer Regulates Male-Female Differences in Drosophila Body Size". PLOS Genetics. 11 (12): e1005683. doi:10.1371/journal.pgen.1005683. PMC 4692505. PMID 26710087.
  103. ^ Gilbert SF (2000). Developmental Biology (6th ed.). Sunderland (MA): Sinauer Associates; 2000. ISBN 978-0-87893-243-6.
  104. ^ Lemaitre B, Hoffmann J (2007). "The host defense of Drosophila melanogaster" (PDF). Annual Review of Immunology. 25: 697–743. doi:10.1146/annurev.immunol.25.022106.141615. PMID 17201680.
  105. ^ Troha K, Im JH, Revah J, Lazzaro BP, Buchon N (February 2018). "Comparative transcriptomics reveals CrebA as a novel regulator of infection tolerance in D. melanogaster". PLOS Pathogens. 14 (2): e1006847. doi:10.1371/journal.ppat.1006847. PMC 5812652. PMID 29394281.
  106. ^ De Gregorio E, Spellman PT, Tzou P, Rubin GM, Lemaitre B (June 2002). "The Toll and Imd pathways are the major regulators of the immune response in Drosophila". The EMBO Journal. 21 (11): 2568–79. doi:10.1093/emboj/21.11.2568. PMC 126042. PMID 12032070.
  107. ^ Paredes JC, Welchman DP, Poidevin M, Lemaitre B (November 2011). "Negative regulation by amidase PGRPs shapes the Drosophila antibacterial response and protects the fly from innocuous infection" (PDF). Immunity. 35 (5): 770–9. doi:10.1016/j.immuni.2011.09.018. PMID 22118526.
  108. ^ Troha K, Buchon N (September 2019). "Methods for the study of innate immunity in Drosophila melanogaster". Wiley Interdisciplinary Reviews. Developmental Biology. 8 (5): e344. doi:10.1002/wdev.344. PMID 30993906. S2CID 119527642.
  109. ^ Gilbert R, Torres M, Clemens R, Hateley S, Hosamani R, Wade W, Bhattacharya S (February 2020). "Drosophila melanogaster infection model". npj Microgravity. 6 (1): 4. doi:10.1038/s41526-019-0091-2. PMC 7000411. PMID 32047838.
  110. ^ a b Dostálová A, Rommelaere S, Poidevin M, Lemaitre B (September 2017). "Thioester-containing proteins regulate the Toll pathway and play a role in Drosophila defence against microbial pathogens and parasitoid wasps". BMC Biology. 15 (1): 79. doi:10.1186/s12915-017-0408-0. PMC 5584532. PMID 28874153.
  111. ^ Srinivasan N, Gordon O, Ahrens S, Franz A, Deddouche S, Chakravarty P, et al. (November 2016). "Drosophila melanogaster". eLife. 5. doi:10.7554/eLife.19662. PMC 5138034. PMID 27871362.
  112. ^ Goto A, Yano T, Terashima J, Iwashita S, Oshima Y, Kurata S (May 2010). "Cooperative regulation of the induction of the novel antibacterial Listericin by peptidoglycan recognition protein LE and the JAK-STAT pathway". The Journal of Biological Chemistry. 285 (21): 15731–8. doi:10.1074/jbc.M109.082115. PMC 2871439. PMID 20348097.
  113. ^ Wang L, Kounatidis I, Ligoxygakis P (January 2014). "Drosophila as a model to study the role of blood cells in inflammation, innate immunity and cancer". Frontiers in Cellular and Infection Microbiology. 3: 113. doi:10.3389/fcimb.2013.00113. PMC 3885817. PMID 24409421.
  114. ^ Neyen C, Bretscher AJ, Binggeli O, Lemaitre B (June 2014). "Methods to study Drosophila immunity" (PDF). Methods. 68 (1): 116–28. doi:10.1016/j.ymeth.2014.02.023. PMID 24631888.
  115. ^ Hashimoto Y, Tabuchi Y, Sakurai K, Kutsuna M, Kurokawa K, Awasaki T, et al. (December 2009). "Identification of lipoteichoic acid as a ligand for draper in the phagocytosis of Staphylococcus aureus by Drosophila hemocytes". Journal of Immunology. 183 (11): 7451–60. doi:10.4049/jimmunol.0901032. PMID 19890048.
  116. ^ Holz A, Bossinger B, Strasser T, Janning W, Klapper R (October 2003). "The two origins of hemocytes in Drosophila". Development. 130 (20): 4955–62. doi:10.1242/dev.00702. PMID 12930778.
  117. ^ Sanchez Bosch P, Makhijani K, Herboso L, Gold KS, Baginsky R, Woodcock KJ, et al. (December 2019). "Adult Drosophila Lack Hematopoiesis but Rely on a Blood Cell Reservoir at the Respiratory Epithelia to Relay Infection Signals to Surrounding Tissues". Developmental Cell. 51 (6): 787–803.e5. doi:10.1016/j.devcel.2019.10.017. PMC 7263735. PMID 31735669.
  118. ^ Parvy JP, Yu Y, Dostalova A, Kondo S, Kurjan A, Bulet P, et al. (July 2019). "Drosophila". eLife. 8: e45061. doi:10.7554/eLife.45061. PMC 6667213. PMID 31358113.
  119. ^ Sturtevant AH (1929). "The claret mutant type of Drosophila simulans: a study of chromosome elimination and cell-lineage". Zeitschrift für Wissenschaftliche Zoologie. 135: 323–356.
  120. ^ Nissani M (May 1975). "A new behavioral bioassay for an analysis of sexual attraction and pheromones in insects". The Journal of Experimental Zoology. 192 (2): 271–5. Bibcode:1975JEZ...192..271N. doi:10.1002/jez.1401920217. PMID 805823.
  121. ^ Khan FA (2011). Biotechnology Fundamentals. CRC Press. p. 213. ISBN 978-1-4398-2009-4.
  122. ^ "The 2017 Nobel Prize in Physiology or Medicine jointly to Jeffrey C. Hall, Michael Rosbash and Michael W. Young for their discoveries of molecular mechanisms controlling the circadian rhythm". Nobelprize.org. October 2, 2017. Retrieved October 5, 2017.
  123. ^ Lehnert BP, Baker AE, Gaudry Q, Chiang AS, Wilson RI (January 2013). "Distinct roles of TRP channels in auditory transduction and amplification in Drosophila". Neuron. 77 (1): 115–28. doi:10.1016/j.neuron.2012.11.030. PMC 3811118. PMID 23312520.
  124. ^ Zhang W, Yan Z, Jan LY, Jan YN (August 2013). "Sound response mediated by the TRP channels NOMPC, NANCHUNG, and INACTIVE in chordotonal organs of Drosophila larvae". Proceedings of the National Academy of Sciences of the United States of America. 110 (33): 13612–7. Bibcode:2013PNAS..11013612Z. doi:10.1073/pnas.1312477110. PMC 3746866. PMID 23898199.
  125. ^ "Homosexuality Turned On and Off in Fruit Flies"
  126. ^ Dasgupta S, Sheehan TC, Stevens CF, Navlakha S (December 2018). "A neural data structure for novelty detection". Proceedings of the National Academy of Sciences of the United States of America. 115 (51): 13093–13098. Bibcode:2018PNAS..11513093D. doi:10.1073/pnas.1814448115. PMC 6304992. PMID 30509984.
  127. ^ Zwarts L, Versteven M, Callaerts P (January 1, 2012). "Genetics and neurobiology of aggression in Drosophila". Fly. 6 (1): 35–48. doi:10.4161/fly.19249. PMC 3365836. PMID 22513455.
  128. ^ Davis SM, Thomas AL, Liu L, Campbell IM, Dierick HA (January 2018). "Drosophila Using a Screen for Wing Damage". Genetics. 208 (1): 273–282. doi:10.1534/genetics.117.300292. PMC 5753862. PMID 29109180.
  129. ^ a b c Versteven M, Vanden Broeck L, Geurten B, Zwarts L, Decraecker L, Beelen M, et al. (February 2017). "Drosophila aggression". Proceedings of the National Academy of Sciences of the United States of America. 114 (8): 1958–1963. doi:10.1073/pnas.1605946114. PMC 5338383. PMID 28115690.
  130. ^ a b Sengupta S, Smith DP (2014). "How Drosophila Detect Volatile Pheromones: Signaling, Circuits, and Behavior". In Mucignat-Caretta C (ed.). Neurobiology of Chemical Communication. Frontiers in Neuroscience. CRC Press/Taylor & Francis. ISBN 978-1-4665-5341-5. PMID 24830032. Retrieved May 30, 2019.
  131. ^ Laturney M, Billeter JC (August 2016). "Drosophila melanogaster females restore their attractiveness after mating by removing male anti-aphrodisiac pheromones". Nature Communications. 7 (1): 12322. Bibcode:2016NatCo...712322L. doi:10.1038/ncomms12322. PMC 4976142. PMID 27484362.
  132. ^ Wang L, Han X, Mehren J, Hiroi M, Billeter JC, Miyamoto T, et al. (June 2011). "Hierarchical chemosensory regulation of male-male social interactions in Drosophila". Nature Neuroscience. 14 (6): 757–62. doi:10.1038/nn.2800. PMC 3102769. PMID 21516101.
  133. ^ a b c Lim RS, Eyjólfsdóttir E, Shin E, Perona P, Anderson DJ (August 27, 2014). "How food controls aggression in Drosophila". PLOS ONE. 9 (8): e105626. Bibcode:2014PLoSO...9j5626L. doi:10.1371/journal.pone.0105626. PMC 4146546. PMID 25162609.
  134. ^ Erion R, DiAngelo JR, Crocker A, Sehgal A (September 2012). "Interaction between sleep and metabolism in Drosophila with altered octopamine signaling". The Journal of Biological Chemistry. 287 (39): 32406–14. doi:10.1074/jbc.M112.360875. PMC 3463357. PMID 22829591.
  135. ^ a b c Kayser MS, Mainwaring B, Yue Z, Sehgal A (July 2015). Griffith LC (ed.). "Sleep deprivation suppresses aggression in Drosophila". eLife. 4: e07643. doi:10.7554/eLife.07643. PMC 4515473. PMID 26216041.
  136. ^ Hardie RC, Raghu P (September 2001). "Visual transduction in Drosophila". Nature. 413 (6852): 186–93. Bibcode:2001Natur.413..186H. doi:10.1038/35093002. PMID 11557987. S2CID 4415605.
  137. ^ a b c d Sharkey CR, Blanco J, Leibowitz MM, Pinto-Benito D, Wardill TJ (October 2020). "The spectral sensitivity of Drosophila photoreceptors". Scientific Reports. 10 (1): 18242. Bibcode:2020NatSR..1018242S. doi:10.1038/s41598-020-74742-1. PMC 7588446. PMID 33106518. S2CID 215551298.   Material was copied and adapted from this source, which is available under a Creative Commons Attribution 4.0 International License.
  138. ^ Feuda R, Goulty M, Zadra N, Gasparetti T, Rosato E, Pisani D, et al. (August 2021). "Phylogenomics of Opsin Genes in Diptera Reveals Lineage-Specific Events and Contrasting Evolutionary Dynamics in Anopheles and Drosophila". Genome Biology and Evolution. 13 (8): evab170. doi:10.1093/gbe/evab170. PMC 8369074. PMID 34270718.
  139. ^ Harris WA, Stark WS, Walker JA (April 1976). "Genetic dissection of the photoreceptor system in the compound eye of Drosophila melanogaster". The Journal of Physiology. 256 (2): 415–439. doi:10.1113/jphysiol.1976.sp011331. PMC 1309314. PMID 16992509. S2CID 45888026.
  140. ^ a b Ostroy SE, Wilson M, Pak WL (August 1974). "Drosophila rhodopsin: photochemistry, extraction and differences in the norp AP12 phototransduction mutant". Biochemical and Biophysical Research Communications. 59 (3): 960–966. doi:10.1016/s0006-291x(74)80073-2. PMID 4213042.
  141. ^ Ostroy SE (November 1978). "Characteristics of Drosophila rhodopsin in wild-type and norpA vision transduction mutants". The Journal of General Physiology. 72 (5): 717–732. doi:10.1085/jgp.72.5.717. PMC 2228556. PMID 105082. S2CID 5802525.
  142. ^ a b c d e f g Salcedo E, Huber A, Henrich S, Chadwell LV, Chou WH, Paulsen R, Britt SG (December 1999). "Blue- and green-absorbing visual pigments of Drosophila: ectopic expression and physiological characterization of the R8 photoreceptor cell-specific Rh5 and Rh6 rhodopsins". The Journal of Neuroscience. 19 (24): 10716–10726. doi:10.1523/jneurosci.19-24-10716.1999. PMC 6784940. PMID 10594055. S2CID 17575850.
  143. ^ Kirschfeld K, Franceschini N, Minke B (September 1977). "Evidence for a sensitising pigment in fly photoreceptors". Nature. 269 (5627): 386–390. Bibcode:1977Natur.269..386K. doi:10.1038/269386a0. PMID 909585. S2CID 28890008.
  144. ^ Minke B, Kirschfeld K (May 1979). "The contribution of a sensitizing pigment to the photosensitivity spectra of fly rhodopsin and metarhodopsin". The Journal of General Physiology. 73 (5): 517–540. doi:10.1085/jgp.73.5.517. PMC 2215190. PMID 458418. S2CID 12451748.
  145. ^ a b Kirschfeld K, Feiler R, Franceschini N (1978). "A photostable pigment within the rhabdomere of fly photoreceptors no. 7". Journal of Comparative Physiology A. 125 (3): 275–284. doi:10.1007/BF00656606. S2CID 40233531.
  146. ^ Kirschfeld K, Franceschini N (June 1977). "Photostable pigments within the membrane of photoreceptors and their possible role". Biophysics of Structure and Mechanism. 3 (2): 191–194. doi:10.1007/BF00535818. PMID 890056. S2CID 5846094.
  147. ^ Zuker CS, Montell C, Jones K, Laverty T, Rubin GM (May 1987). "A rhodopsin gene expressed in photoreceptor cell R7 of the Drosophila eye: homologies with other signal-transducing molecules". The Journal of Neuroscience. 7 (5): 1550–1557. doi:10.1523/jneurosci.07-05-01550.1987. PMC 6568820. PMID 2437266. S2CID 1490332.
  148. ^ a b c Chou WH, Hall KJ, Wilson DB, Wideman CL, Townson SM, Chadwell LV, Britt SG (December 1996). "Identification of a novel Drosophila opsin reveals specific patterning of the R7 and R8 photoreceptor cells". Neuron. 17 (6): 1101–1115. doi:10.1016/s0896-6273(00)80243-3. PMID 8982159. S2CID 18294965.
  149. ^ a b c d Feiler R, Bjornson R, Kirschfeld K, Mismer D, Rubin GM, Smith DP, et al. (October 1992). "Ectopic expression of ultraviolet-rhodopsins in the blue photoreceptor cells of Drosophila: visual physiology and photochemistry of transgenic animals". The Journal of Neuroscience. 12 (10): 3862–3868. doi:10.1523/jneurosci.12-10-03862.1992. PMC 6575971. PMID 1403087.
  150. ^ Montell C, Jones K, Zuker C, Rubin G (May 1987). "A second opsin gene expressed in the ultraviolet-sensitive R7 photoreceptor cells of Drosophila melanogaster". The Journal of Neuroscience. 7 (5): 1558–1566. doi:10.1523/JNEUROSCI.07-05-01558.1987. PMC 6568825. PMID 2952772. S2CID 17003459.
  151. ^ Huber A, Schulz S, Bentrop J, Groell C, Wolfrum U, Paulsen R (April 1997). "Molecular cloning of Drosophila Rh6 rhodopsin: the visual pigment of a subset of R8 photoreceptor cells". FEBS Letters. 406 (1–2): 6–10. doi:10.1016/s0014-5793(97)00210-x. PMID 9109375. S2CID 18368117.
  152. ^ Pollock JA, Benzer S (June 1988). "Transcript localization of four opsin genes in the three visual organs of Drosophila; RH2 is ocellus specific". Nature. 333 (6175): 779–782. Bibcode:1988Natur.333..779P. doi:10.1038/333779a0. PMID 2968518. S2CID 4303934.
  153. ^ a b Feiler R, Harris WA, Kirschfeld K, Wehrhahn C, Zuker CS (June 1988). "Targeted misexpression of a Drosophila opsin gene leads to altered visual function". Nature. 333 (6175): 737–741. Bibcode:1988Natur.333..737F. doi:10.1038/333737a0. PMID 2455230. S2CID 4248264.
  154. ^ Sakai K, Tsutsui K, Yamashita T, Iwabe N, Takahashi K, Wada A, Shichida Y (August 2017). "Drosophila melanogaster rhodopsin Rh7 is a UV-to-visible light sensor with an extraordinarily broad absorption spectrum". Scientific Reports. 7 (1): 7349. Bibcode:2017NatSR...7.7349S. doi:10.1038/s41598-017-07461-9. PMC 5544684. PMID 28779161. S2CID 3276084.
  155. ^ Ni JD, Baik LS, Holmes TC, Montell C (May 2017). "A rhodopsin in the brain functions in circadian photoentrainment in Drosophila". Nature. 545 (7654): 340–344. Bibcode:2017Natur.545..340N. doi:10.1038/nature22325. PMC 5476302. PMID 28489826. S2CID 4468254.
  156. ^ Vogt K (February 1, 1984). "The Chromophore of the Visual Pigment in Some Insect Orders". Zeitschrift für Naturforschung C. 39 (1–2): 196–197. doi:10.1515/znc-1984-1-236. S2CID 88980658.
  157. ^ Vogt K, Kirschfeld K (April 1984). "Chemical identity of the chromophores of fly visual pigment". Naturwissenschaften. 71 (4): 211–213. Bibcode:1984NW.....71..211V. doi:10.1007/BF00490436. S2CID 24205801.
  158. ^ Gühmann M, Porter ML, Bok MJ (August 2022). "The Gluopsins: Opsins without the Retinal Binding Lysine". Cells. 11 (15): 2441. doi:10.3390/cells11152441. PMC 9368030. PMID 35954284.
  159. ^ Katana R, Guan C, Zanini D, Larsen ME, Giraldo D, Geurten BR, et al. (September 2019). "Chromophore-Independent Roles of Opsin Apoproteins in Drosophila Mechanoreceptors". Current Biology. 29 (17): 2961–2969.e4. doi:10.1016/j.cub.2019.07.036. PMID 31447373. S2CID 201420079.
  160. ^ a b Leung NY, Thakur DP, Gurav AS, Kim SH, Di Pizio A, Niv MY, Montell C (April 2020). "Functions of Opsins in Drosophila Taste". Current Biology. 30 (8): 1367–1379.e6. doi:10.1016/j.cub.2020.01.068. PMC 7252503. PMID 32243853.
  161. ^ Kumbalasiri T, Rollag MD, Isoldi MC, Castrucci AM, Provencio I (March 2007). "Melanopsin triggers the release of internal calcium stores in response to light". Photochemistry and Photobiology. 83 (2): 273–279. doi:10.1562/2006-07-11-RA-964. PMID 16961436. S2CID 23060331.
  162. ^ a b Raghu P, Colley NJ, Webel R, James T, Hasan G, Danin M, et al. (May 2000). "Normal phototransduction in Drosophila photoreceptors lacking an InsP(3) receptor gene". Molecular and Cellular Neurosciences. 15 (5): 429–45. doi:10.1006/mcne.2000.0846. PMID 10833300. S2CID 23861204.
  163. ^ Hardie RC, Juusola M (October 2015). "Phototransduction in Drosophila". Current Opinion in Neurobiology. 34: 37–45. doi:10.1016/j.conb.2015.01.008. PMID 25638280. S2CID 140206989.
  164. ^ Wang T, Xu H, Oberwinkler J, Gu Y, Hardie RC, Montell C (February 2005). "Light activation, adaptation, and cell survival functions of the Na+/Ca2+ exchanger CalX". Neuron. 45 (3): 367–78. doi:10.1016/j.neuron.2004.12.046. PMID 15694324.
  165. ^ Rein K, Zöckler M, Mader MT, Grübel C, Heisenberg M (February 2002). "The Drosophila standard brain". Current Biology. 12 (3): 227–31. doi:10.1016/S0960-9822(02)00656-5. PMID 11839276. S2CID 15785406.
  166. ^ Dawkins R, Dawkins M (1976). "Hierarchical organization and postural facilitation: rules for grooming in flies". Animal Behaviour. 24 (4): 739–755. doi:10.1016/S0003-3472(76)80003-6. S2CID 53186674.
  167. ^ a b c Davis WJ (1979). "Behavioural hierarchies". Trends in Neurosciences. 2 (2): 5–7. doi:10.1016/0166-2236(79)90003-1. S2CID 53180462.
  168. ^ a b Seeds AM, Ravbar P, Chung P, Hampel S, Midgley FM, Mensh BD, Simpson JH (August 2014). "A suppression hierarchy among competing motor programs drives sequential grooming in Drosophila". eLife. 3: e02951. doi:10.7554/eLife.02951. PMC 4136539. PMID 25139955.
  169. ^ Mathis A, Mamidanna P, Cury KM, Abe T, Murthy VN, Mathis MW, Bethge M (September 2018). "DeepLabCut: markerless pose estimation of user-defined body parts with deep learning". Nature Neuroscience. 21 (9): 1281–1289. doi:10.1038/s41593-018-0209-y. PMID 30127430. S2CID 4748395.
  170. ^ Strauss R, Heisenberg M (August 1990). "Coordination of legs during straight walking and turning in Drosophila melanogaster". Journal of Comparative Physiology A. 167 (3): 403–12. doi:10.1007/BF00192575. PMID 2121965. S2CID 12965869.
  171. ^ DeAngelis BD, Zavatone-Veth JA, Clark DA (June 2019). "Drosophila". eLife. 8. doi:10.7554/eLife.46409. PMC 6598772. PMID 31250807.
  172. ^ Wosnitza A, Bockemühl T, Dübbert M, Scholz H, Büschges A (February 2013). "Inter-leg coordination in the control of walking speed in Drosophila". The Journal of Experimental Biology. 216 (Pt 3): 480–91. doi:10.1242/jeb.078139. PMID 23038731.
  173. ^ Mendes CS, Bartos I, Akay T, Márka S, Mann RS (January 2013). "Quantification of gait parameters in freely walking wild type and sensory deprived Drosophila melanogaster". eLife. 2: e00231. doi:10.7554/eLife.00231. PMC 3545443. PMID 23326642.
  174. ^ Szczecinski NS, Bockemühl T, Chockley AS, Büschges A (November 2018). "Drosophila". The Journal of Experimental Biology. 221 (Pt 22): jeb189142. doi:10.1242/jeb.189142. PMID 30274987.
  175. ^ Hooper SL (May 2012). "Body size and the neural control of movement". Current Biology. 22 (9): R318-22. doi:10.1016/j.cub.2012.02.048. PMID 22575473.
  176. ^ a b c Fry SN, Sayaman R, Dickinson MH (April 2003). (PDF). Science. 300 (5618): 495–8. Bibcode:2003Sci...300..495F. doi:10.1126/science.1081944. PMID 12702878. S2CID 40952385. Archived from the original (PDF) on September 24, 2015.
  177. ^ Hesselberg T, Lehmann FO (December 2007). "Turning behaviour depends on frictional damping in the fruit fly Drosophila". The Journal of Experimental Biology. 210 (Pt 24): 4319–34. doi:10.1242/jeb.010389. PMID 18055621.
  178. ^ Chiang AS, Lin CY, Chuang CC, Chang HM, Hsieh CH, Yeh CW, et al. (January 2011). "Three-dimensional reconstruction of brain-wide wiring networks in Drosophila at single-cell resolution". Current Biology. 21 (1): 1–11. doi:10.1016/j.cub.2010.11.056. PMID 21129968. S2CID 17155338.
  179. ^ "FlyCircuit - A Database of Drosophila Brain Neurons". Retrieved August 30, 2013.
  180. ^ Meinertzhagen IA, O'Neil SD (March 1991). "Synaptic organization of columnar elements in the lamina of the wild type in Drosophila melanogaster". The Journal of Comparative Neurology. 305 (2): 232–63. doi:10.1002/cne.903050206. PMID 1902848. S2CID 35301798.
  181. ^ Rivera-Alba M, Vitaladevuni SN, Mishchenko Y, Mischenko Y, Lu Z, Takemura SY, et al. (December 2011). "Wiring economy and volume exclusion determine neuronal placement in the Drosophila brain". Current Biology. 21 (23): 2000–5. doi:10.1016/j.cub.2011.10.022. PMC 3244492. PMID 22119527.
  182. ^ Takemura SY, Bharioke A, Lu Z, Nern A, Vitaladevuni S, Rivlin PK, et al. (August 2013). "A visual motion detection circuit suggested by Drosophila connectomics". Nature. 500 (7461): 175–81. Bibcode:2013Natur.500..175T. doi:10.1038/nature12450. PMC 3799980. PMID 23925240.
  183. ^ Takemura SY, Aso Y, Hige T, Wong A, Lu Z, Xu CS, et al. (July 2017). "Drosophila brain". eLife. 6: e26975. doi:10.7554/eLife.26975. PMC 5550281. PMID 28718765.
  184. ^ "Entire Fruit Fly Brain Imaged with Electron Microscopy". The Scientist Magazine. Retrieved July 15, 2018.
  185. ^ Zheng Z, Lauritzen JS, Perlman E, Robinson CG, Nichols M, Milkie D, et al. (July 2018). "A Complete Electron Microscopy Volume of the Brain of Adult Drosophila melanogaster". Cell. 174 (3): 730–743.e22. bioRxiv 10.1101/140905. doi:10.1016/j.cell.2018.06.019. PMC 6063995. PMID 30033368.
  186. ^ Xu CS, Januszewski M, Lu Z, Takemura SY, Hayworth K, Huang G, Shinomiya K, Maitin-Shepard J, Ackerman D, Berg S, Blakely T, et al. (2020). "A connectome of the adult Drosophila central brain". bioRxiv. Cold Spring Harbor Laboratory: 2020.01.21.911859. doi:10.1101/2020.01.21.911859. S2CID 213140797.
  187. ^ "Analysis tools for connectomics". HHMI.
  188. ^ Phelps, Jasper S.; Hildebrand, David Grant Colburn; Graham, Brett J.; Kuan, Aaron T.; Thomas, Logan A.; Nguyen, Tri M.; Buhmann, Julia; Azevedo, Anthony W.; Sustar, Anne; Agrawal, Sweta; Liu, Mingguan; Shanny, Brendan L.; Funke, Jan; Tuthill, John C.; Lee, Wei-Chung Allen (February 2, 2021). "Reconstruction of motor control circuits in adult Drosophila using automated transmission electron microscopy". Cell. 184 (3): 759–774.e18. doi:10.1016/j.cell.2020.12.013. PMC 8312698. PMID 33400916.
  189. ^ Scheffer LK, Xu CS, Januszewski M, Lu Z, Takemura SY, Hayworth KJ, Huang G, Shinomiya K, Maitlin-Shepard J, Berg S, Clements J, et al. (2020). "A Connectome and Analysis of the Adult Drosophila Central Brain". bioRxiv. Cold Spring Harbor. 9. doi:10.1101/2020.04.07.030213. PMC 7546738. PMID 32880371. S2CID 215790785.
  190. ^ Popovych, Sergiy; Macrina, Thomas; Kemnitz, Nico; Castro, Manuel; Nehoran, Barak; Jia, Zhen; Bae, J. Alexander; Mitchell, Eric; Mu, Shang; Trautman, Eric T.; Saalfeld, Stephan; Li, Kai; Seung, Sebastian (March 27, 2022). "Petascale pipeline for precise alignment of images from serial section electron microscopy". bioRxiv 10.1101/2022.03.25.485816.
  191. ^ Winding, Michael; Pedigo, Benjamin D.; Barnes, Christopher L.; Patsolic, Heather G.; Park, Youngser; Kazimiers, Tom; Fushiki, Akira; Andrade, Ingrid V.; Khandelwal, Avinash; Valdes-Aleman, Javier; Li, Feng; Randel, Nadine; Barsotti, Elizabeth; Correia, Ana; Fetter, Richard D. (March 10, 2023). "The connectome of an insect brain". Science. 379 (6636): eadd9330. doi:10.1126/science.add9330. PMC 7614541. PMID 36893230.
  192. ^ "Non pest species". Plant Health Australia. Retrieved September 19, 2017.
  193. ^ McEvey S (February 5, 2014). "Fruit Flies: A Case Of Mistaken Identity". Australian Museum. Retrieved September 19, 2017.

Further reading edit

  • Kohler RE (1994). Lords of the Fly: Drosophila genetics and the experimental life. Chicago: University of Chicago Press. ISBN 978-0-226-45063-6.
  • Gilbert SF (2000). Developmental Biology (6th ed.). Sunderland (MA): Sinauer Associates; 2000. ISBN 978-0-87893-243-6.
  • Perrimon N, Bonini NM, Dhillon P (March 2016). "Fruit flies on the front line: the translational impact of Drosophila". Disease Models & Mechanisms. 9 (3): 229–31. doi:10.1242/dmm.024810. PMC 4833334. PMID 26935101.
  • Henderson M (April 8, 2010). "Row over fruit fly Drosophila melanogaster name bugs scientists". The Times. The Australian. Retrieved September 19, 2017.

External links edit

  • "A quick and simple introduction to Drosophila melanogaster". Drosophila Virtual Library.
  • "Drosophila Genomics Resource Center" – collects, maintains and distributes Drosophila DNA clones and cell lines.
  • "Bloomington Drosophila Stock Center" – collects, maintains and distributes Drosophila melanogaster strains for research
  • "FlyBase—A Database of Drosophila Genes & Genomes".
  • "NCBI Map Viewer – Drosophila melanogaster".
  • "Drosophila Virtual Library".
  • "The Berkeley Drosophila Genome Project".
  • "FlyMove". – video resources for Drosophila development
  • . Archived from the original on October 8, 2011.
  • View the Fruitfly genome on Ensembl
  • View the dm6 genome assembly in the UCSC Genome Browser.
  • Manchester Fly Facility – for the public 2015-05-13 at the Wayback Machine from the University of Manchester
  • The droso4schools website with school-relevant resources about Drosophila
  • Part 1 of the "Small fly: BIG impact" educational videos explaining the history and importance of the model organism Drosophila.
  • Part 2 of the "Small fly: BIG impact" educational videos explaining how research is carried out in Drosophila.
  • "Inside the Fly Lab"—broadcast by WGBH and PBS, in the program series Curious, January 2008.
  • "How a Fly Detects Poison" Archived 2013-01-13 at archive.today—WhyFiles.org article describes how the fruit fly tastes a larva-killing chemical in food.

drosophila, melanogaster, species, insect, order, diptera, family, drosophilidae, species, often, referred, fruit, lesser, fruit, less, commonly, vinegar, pomace, banana, wild, melanogaster, attracted, rotting, fruit, fermenting, beverages, often, found, orcha. Drosophila melanogaster is a species of fly an insect of the order Diptera in the family Drosophilidae The species is often referred to as the fruit fly or lesser fruit fly or less commonly the vinegar fly pomace fly a 4 or banana fly 5 In the wild D melanogaster are attracted to rotting fruit and fermenting beverages and are often found in orchards kitchens and pubs Drosophila melanogasterFruit fly feeding off a bananaScientific classificationDomain EukaryotaKingdom AnimaliaPhylum ArthropodaClass InsectaOrder DipteraFamily DrosophilidaeGenus DrosophilaSubgenus SophophoraSpecies group melanogasterSpecies subgroup melanogasterSpecies complex melanogasterSpecies D melanogasterBinomial nameDrosophila melanogasterMeigen 1830 1 Starting with Charles W Woodworth s 1901 proposal of the use of this species as a model organism 6 7 D melanogaster continues to be widely used for biological research in genetics physiology microbial pathogenesis and life history evolution As of 2017 six Nobel Prizes have been awarded to drosophilists for their work using the insect 8 9 D melanogaster is typically used in research owing to its rapid life cycle relatively simple genetics with only four pairs of chromosomes and large number of offspring per generation 10 It was originally an African species with all non African lineages having a common origin 11 Its geographic range includes all continents including islands 12 D melanogaster is a common pest in homes restaurants and other places where food is served 13 Flies belonging to the family Tephritidae are also called fruit flies This can cause confusion especially in the Mediterranean Australia and South Africa where the Mediterranean fruit fly Ceratitis capitata is an economic pest Contents 1 Etymology 2 Physical appearance 3 Lifecycle and reproduction 3 1 Females 3 2 Males 3 3 Polygamy 4 Model organism in genetics 4 1 History of use in genetic analysis 4 2 Reasons for use in laboratories 4 3 Genetic markers 4 4 Classic genetic mutations 5 Genome 5 1 Similarity to humans 6 Development 6 1 Developmental plasticity 7 Sex determination 8 Immunity 8 1 JAK STAT signalling 8 2 The cellular response to infection 9 Behavioral genetics and neuroscience 9 1 Aggression 9 1 1 Acoustic communication 9 1 2 Pheromone signaling 9 1 3 Competition for food 9 1 4 Effect of sleep deprivation 9 2 Vision 9 2 1 Opsins and spectral sensitivity 9 3 Phototransduction 9 4 Grooming 9 5 Walking 9 6 Flight 10 Connectome 11 Misconceptions 12 See also 13 Notes 14 References 15 Further reading 16 External linksEtymology editThe term Drosophila meaning dew loving is a modern scientific Latin adaptation from Greek words drosos drosos dew and filia philia lover The term melanogaster meaning black belly comes from Ancient Greek melas melas black and gᾰsthr gastḗr belly Physical appearance edit nbsp Female left and male right D melanogaster nbsp View from above nbsp Front view Wild type fruit flies are yellow brown with brick red eyes and transverse black rings across the abdomen The black portions of the abdomen are the inspiration for the species name melanogaster black bellied The brick red color of the eyes of the wild type fly are due to two pigments 14 xanthommatin which is brown and is derived from tryptophan and drosopterins which are red and are derived from guanosine triphosphate 14 They exhibit sexual dimorphism females are about 2 5 mm 0 10 in long males are slightly smaller with darker backs Males are easily distinguished from females based on colour differences with a distinct black patch at the abdomen less noticeable in recently emerged flies and the sex combs a row of dark bristles on the tarsus of the first leg Furthermore males have a cluster of spiky hairs claspers surrounding the reproducing parts used to attach to the female during mating Extensive images are found at FlyBase 15 Drosophila melanogaster can be distinguished from related species by the following combination of features gena 1 10 diameter of eye at greatest vertical height wing hyaline and with costal index 2 4 male protarsus with a single row of 12 setae forming a sex comb male epandrial posterior lobe small and nearly triangular female abdominal tergite 6 with dark band running to its ventral margin female oviscapt small pale without dorsodistal depression and with 12 13 peg like outer ovisensilla 16 17 Drosophila melanogaster flies can sense air currents with the hairs on their backs Their eyes are sensitive to slight differences in light intensity and will instinctively fly away when a shadow or other movement is detected 18 Lifecycle and reproduction edit nbsp Egg of D melanogasterUnder optimal growth conditions at 25 C 77 F the D melanogaster lifespan is about 50 days from egg to death 19 The developmental period for D melanogaster varies with temperature as with many ectothermic species The shortest development time egg to adult 7 days is achieved at 28 C 82 F 20 21 Development times increase at higher temperatures 11 days at 30 C or 86 F due to heat stress Under ideal conditions the development time at 25 C 77 F is 8 5 days 20 21 22 at 18 C 64 F it takes 19 days 20 21 and at 12 C 54 F it takes over 50 days 20 21 Under crowded conditions development time increases 23 while the emerging flies are smaller 23 24 Females lay some 400 eggs embryos about five at a time into rotting fruit or other suitable material such as decaying mushrooms and sap fluxes Drosophila melanogaster is a holometabolous insect so it undergoes a full metamorphosis Their life cycle is broken down into 4 stages embryo larva pupa adult 25 The eggs which are about 0 5 mm long hatch after 12 15 hours at 25 C or 77 F 20 21 The resulting larvae grow for about 4 days at 25 C while molting twice into second and third instar larvae at about 24 and 48 h after hatching 20 21 During this time they feed on the microorganisms that decompose the fruit as well as on the sugar of the fruit itself The mother puts feces on the egg sacs to establish the same microbial composition in the larvae s guts that has worked positively for herself 26 Then the larvae encapsulate in the puparium and undergo a 4 day long metamorphosis at 25 C after which the adults eclose emerge 20 21 source source Sound of Drosophila heartbeatMales perform a sequence of five behavioral patterns to court females First males orient themselves while playing a courtship song by horizontally extending and vibrating their wings Soon after the male positions himself at the rear of the female s abdomen in a low posture to tap and lick the female genitalia Finally the male curls his abdomen and attempts copulation Females can reject males by moving away kicking and extruding their ovipositor 27 Copulation lasts around 15 20 minutes 28 during which males transfer a few hundred very long 1 76 mm sperm cells in seminal fluid to the female 29 Females store the sperm in a tubular receptacle and in two mushroom shaped spermathecae sperm from multiple matings compete for fertilization A last male precedence is believed to exist the last male to mate with a female sires about 80 of her offspring This precedence was found to occur through both displacement and incapacitation 30 The displacement is attributed to sperm handling by the female fly as multiple matings are conducted and is most significant during the first 1 2 days after copulation Displacement from the seminal receptacle is more significant than displacement from the spermathecae 30 Incapacitation of first male sperm by second male sperm becomes significant 2 7 days after copulation The seminal fluid of the second male is believed to be responsible for this incapacitation mechanism without removal of first male sperm which takes effect before fertilization occurs 30 The delay in effectiveness of the incapacitation mechanism is believed to be a protective mechanism that prevents a male fly from incapacitating his own sperm should he mate with the same female fly repetitively Sensory neurons in the uterus of female D melanogaster respond to a male protein sex peptide which is found in semen 31 This protein makes the female reluctant to copulate for about 10 days after insemination The signal pathway leading to this change in behavior has been determined The signal is sent to a brain region that is a homolog of the hypothalamus and the hypothalamus then controls sexual behavior and desire 31 Gonadotropic hormones in Drosophila maintain homeostasis and govern reproductive output via a cyclic interrelationship not unlike the mammalian estrous cycle 32 Sex peptide perturbs this homeostasis and dramatically shifts the endocrine state of the female by inciting juvenile hormone synthesis in the corpus allatum 33 D melanogaster is often used for life extension studies such as to identify genes purported to increase lifespan when mutated 34 D melanogaster is also used in studies of aging Werner syndrome is a condition in humans characterized by accelerated aging It is caused by mutations in the gene WRN that encodes a protein with essential roles in repair of DNA damage Mutations in the D melanogaster homolog of WRN also cause increased physiologic signs of aging such as shorter lifespan higher tumor incidence muscle degeneration reduced climbing ability altered behavior and reduced locomotor activity 35 Females edit nbsp Mating in captivityFemales become receptive to courting males about 8 12 hours after emergence 36 Specific neuron groups in females have been found to affect copulation behavior and mate choice One such group in the abdominal nerve cord allows the female fly to pause her body movements to copulate 31 Activation of these neurons induces the female to cease movement and orient herself towards the male to allow for mounting If the group is inactivated the female remains in motion and does not copulate Various chemical signals such as male pheromones often are able to activate the group 31 Also females exhibit mate choice copying When virgin females are shown other females copulating with a certain type of male they tend to copulate more with this type of male afterwards than naive females which have not observed the copulation of others This behavior is sensitive to environmental conditions and females copulate less in bad weather conditions 37 Males edit This section needs additional citations for verification Please help improve this article by adding citations to reliable sources in this section Unsourced material may be challenged and removed October 2015 Learn how and when to remove this template message source source source source Courtship behavior in male The male first showed wing extension stage 1 and later other steps such as abdomen bending stage 2 then frequent attempted copulation licking and even ejaculation stage 3 finally the male fell over and was on its back stage 4 D melanogaster males exhibit a strong reproductive learning curve That is with sexual experience these flies tend to modify their future mating behavior in multiple ways These changes include increased selectivity for courting only intraspecifically as well as decreased courtship times Sexually naive D melanogaster males are known to spend significant time courting interspecifically such as with D simulans flies Naive D melanogaster will also attempt to court females that are not yet sexually mature and other males D melanogaster males show little to no preference for D melanogaster females over females of other species or even other male flies However after D simulans or other flies incapable of copulation have rejected the males advances D melanogaster males are much less likely to spend time courting nonspecifically in the future This apparent learned behavior modification seems to be evolutionarily significant as it allows the males to avoid investing energy into futile sexual encounters 38 In addition males with previous sexual experience modify their courtship dance when attempting to mate with new females the experienced males spend less time courting so have lower mating latencies meaning that they are able to reproduce more quickly This decreased mating latency leads to a greater mating efficiency for experienced males over naive males 39 This modification also appears to have obvious evolutionary advantages as increased mating efficiency is extremely important in the eyes of natural selection Polygamy edit Both male and female D melanogaster flies act polygamously having multiple sexual partners at the same time 40 In both males and females polygamy results in a decrease in evening activity compared to virgin flies more so in males than females 40 Evening activity consists of those in which the flies participate other than mating and finding partners such as finding food 41 The reproductive success of males and females varies because a female only needs to mate once to reach maximum fertility 41 Mating with multiple partners provides no advantage over mating with one partner so females exhibit no difference in evening activity between polygamous and monogamous individuals 41 For males however mating with multiple partners increases their reproductive success by increasing the genetic diversity of their offspring 41 This benefit of genetic diversity is an evolutionary advantage because it increases the chance that some of the offspring will have traits that increase their fitness in their environment The difference in evening activity between polygamous and monogamous male flies can be explained with courtship For polygamous flies their reproductive success increases by having offspring with multiple partners and therefore they spend more time and energy on courting multiple females 41 On the other hand monogamous flies only court one female and expend less energy doing so 41 While it requires more energy for male flies to court multiple females the overall reproductive benefits it produces has kept polygamy as the preferred sexual choice 41 The mechanism that affects courtship behavior inDrosophila is controlled by the oscillator neurons DN1s and LNDs 42 Oscillation of the DN1 neurons was found to be effected by sociosexual interactions and is connected to mating related decrease of evening activity 42 Model organism in genetics editD melanogaster remains one of the most studied organisms in biological research particularly in genetics and developmental biology It is also employed in studies of environmental mutagenesis History of use in genetic analysis edit nbsp Alfred Sturtevant s Drosophila melanogaster genetic linkage map This was the first successful gene mapping work and provides important evidence for the chromosome theory of inheritance The map shows the relative positions of allelic characteristics on the second Drosophila chromosome The distance between the genes map units are equal to the percentage of crossing over events that occurs between different alleles D melanogaster was among the first organisms used for genetic analysis and today it is one of the most widely used and genetically best known of all eukaryotic organisms All organisms use common genetic systems therefore comprehending processes such as transcription and replication in fruit flies helps in understanding these processes in other eukaryotes including humans 43 Thomas Hunt Morgan began using fruit flies in experimental studies of heredity at Columbia University in 1910 in a laboratory known as the Fly Room The Fly Room was cramped with eight desks each occupied by students and their experiments They started off experiments using milk bottles to rear the fruit flies and handheld lenses for observing their traits The lenses were later replaced by microscopes which enhanced their observations Morgan and his students eventually elucidated many basic principles of heredity including sex linked inheritance epistasis multiple alleles and gene mapping 43 D melanogaster had historically been used in laboratories to study genetics and patterns of inheritance However D melanogaster also has importance in environmental mutagenesis research allowing researchers to study the effects of specific environmental mutagens 44 Reasons for use in laboratories edit nbsp D melanogaster multiple mutants clockwise from top brown eyes and black cuticle 2 mutations cinnabar eyes and wildtype cuticle 1 mutation sepia eyes and ebony cuticle vermilion eyes and yellow cuticle white eyes and yellow cuticle wildtype eyes and yellow cuticle There are many reasons the fruit fly is a popular choice as a model organism Its care and culture require little equipment space and expense even when using large cultures It can be safely and readily anesthetized usually with ether carbon dioxide gas by cooling or with products such as FlyNap Its morphology is easy to identify once anesthetized It has a short generation time about 10 days at room temperature so several generations can be studied within a few weeks It has a high fecundity females lay up to 100 eggs per day and perhaps 2000 in a lifetime 10 Males and females are readily distinguished and virgin females can be easily identified by their light colored translucent abdomen facilitating genetic crossing The mature larva has giant chromosomes in the salivary glands called polytene chromosomes puffs which indicate regions of transcription hence gene activity The under replication of rDNA occurs resulting in only 20 of DNA compared to the brain Compare to the 47 less rDNA in Sarcophaga barbata ovaries It has only four pairs of chromosomes three autosomes and one pair of sex chromosomes Males do not show meiotic recombination facilitating genetic studies Recessive lethal balancer chromosomes carrying visible genetic markers can be used to keep stocks of lethal alleles in a heterozygous state without recombination due to multiple inversions in the balancer The development of this organism from fertilized egg to mature adult is well understood Genetic transformation techniques have been available since 1987 One approach of inserting foreign genes into the Drosophila genome involves P elements The transposable P elements also known as transposons are segments of bacterial DNA that are transferred into the fly genome Transgenic flies have already contributed to many scientific advances e g modeling such human diseases as Parkinson s neoplasia obesity and diabetes Its complete genome was sequenced and first published in 2000 45 Sexual mosaics can be readily produced providing an additional tool for studying the development and behavior of these flies 46 Genetic markers edit See also Abdominal pigmentation in Drosophila melanogaster nbsp D melanogaster which carries the Cy allele right hence showing a characteristic phenotype of curly wings in adult flies 47 Genetic markers are commonly used in Drosophila research for example within balancer chromosomes or P element inserts and most phenotypes are easily identifiable either with the naked eye or under a microscope In the list of a few common markers below the allele symbol is followed by the name of the gene affected and a description of its phenotype Note Recessive alleles are in lower case while dominant alleles are capitalised Cy1 Curly the wings curve away from the body flight may be somewhat impaired e1 Ebony black body and wings heterozygotes are also visibly darker than wild type Sb1 Stubble bristles are shorter and thicker than wild type w1 White eyes lack pigmentation and appear white bw Brown eye color determined by various pigments combined y1 Yellow body pigmentation and wings appear yellow the fly analog of albinismClassic genetic mutations edit Drosophila genes are traditionally named after the phenotype they cause when mutated For example the absence of a particular gene in Drosophila will result in a mutant embryo that does not develop a heart Scientists have thus called this gene tinman named after the Oz character of the same name 48 Likewise changes in the Shavenbaby gene cause the loss of dorsal cuticular hairs in Drosophila sechellia larvae 49 This system of nomenclature results in a wider range of gene names than in other organisms b black The black mutation was discovered in 1910 by Thomas Hunt Morgan 50 The black mutation results in a darker colored body wings veins and segments of the fruit fly s leg 51 This occurs due to the fly s inability to create beta alanine a beta amino acid 50 The phenotypic expression of this mutation varies based on the genotype of the individual for example whether the specimen is homozygotic or heterozygotic results in a darker or less dark appearance 51 This genetic mutation is x linked recessive 52 bw brown The brown eye mutation results from inability to produce or synthesize pteridine red pigments due to a point mutation on chromosome II 53 When the mutation is homozygous the pteridine pigments are unable to be synthesized because in the beginning of the pteridine pathway a defective enzyme is being coded by homozygous recessive genes 54 unreliable source In all mutations in the pteridine pathway produces a darker eye color hence the resulting color of the biochemical defect in the pteridine pathway being brown m miniature One of the first records of the miniature mutation of wings was also made by Thomas Hunt Morgan in 1911 He described the wings as having a similar shape as the wild type phenotype However their miniature designation refers to the lengths of their wings which do not stretch beyond their body and thus are notably shorter than the wild type length He also noted its inheritance is connected to the sex of the fly and could be paired with the inheritance of other sex determined traits such as white eyes 55 The wings may also demonstrate other characteristics deviant from the wild type wing such as a duller and cloudier color 56 Miniature wings are 1 5x shorter than wild type but are believed to have the same number of cells This is due to the lack of complete flattening by these cells making the overall structure of the wing seem shorter in comparison The pathway of wing expansion is regulated by a signal receptor pathway where the neurohormone bursicon interacts with its complementary G protein coupled receptor this receptor drives one of the G protein subunits to signal further enzyme activity and results in development in the wing such as apoptosis and growth 57 se sepia The eye color of the sepia mutant is sepia a reddish brown color In wild type flies ommochromes brown and drosopterins red give the eyes the typical red color 58 59 The drosopterins are made via a pathway that involves a pyrimidodiazepine synthase 60 which is encoded on chromosome 3L The gene has a premature stop codon in sepia flies so that the flies cannot produce the pyrimidodiazepine synthase and thus no red pigment so that the eyes stay sepia 58 The sepia allele is recessive and thus offspring from sepia flies and homozygous wild type flies has red eyes The sepia phenotype does not depend on the sex of the fly 61 v vermilion The vermilion mutants cannot produce the brown ommochromes leaving the red drosopterins so that the eyes are vermilion colored a radiant red compared to a wild type D melanogaster The vermilion mutation is sex linked and recessive The gene that is defect lies on the X chromosome 62 The brown ommochromes are synthesised from kynurenine which is made from tryptophane Vermilion flies cannot convert tryptophane into kynurenine and thus cannot make ommochromes either 62 Vermilion mutants live longer than wild type flies This longer life span may be associated with the reduced amount of tryptophan converted to kynurenine in vermilion flies 63 nbsp Triple mutant male fruit fly Drosophila melanogaster exhibiting black body vestigial wings and brown eyes mutationsvg vestigial A spontaneous mutation discovered in 1919 by Thomas Morgan and Calvin Bridges Vestigial wings are those not fully developed and that have lost function Since the discovery of the vestigial gene in Drosophila melanogaster there have been many discoveries of the vestigial gene in other vertebrates and their functions within the vertebrates 64 The vestigial gene is considered to be one of the most important genes for wing formation but when it becomes over expressed the issue of ectopic wings begin to form 65 The vestigial gene acts to regulate the expression of the wing imaginal discs in the embryo and acts with other genes to regulate the development of the wings A mutated vestigial allele removes an essential sequence of the DNA required for correct development of the wings 66 w white Drosophila melanogaster wild type typically expresses a brick red eye color The white eye mutation in fruit flies is caused due to the absence of two pigments associated with red and brown eye colors peridines red and ommochromes brown 59 In January 1910 Thomas Hunt Morgan first discovered the white gene and denoted it as w The discovery of the white eye mutation by Morgan brought about the beginnings of genetic experimentation and analysis of Drosophila melanogaster Hunt eventually discovered that the gene followed a similar pattern of inheritance related to the meiotic segregation of the X chromosome He discovered that the gene was located on the X chromosome with this information This led to the discovery of sex linked genes and also to the discovery of other mutations in Drosophila melanogaster 67 The white eye mutation leads to several disadvantages in flies such as a reduced climbing ability shortened life span and lowered resistance to stress when compared to wild type flies 68 Drosophila melanogaster has a series of mating behaviors that enable them to copulate within a given environment and therefore contribute to their fitness After Morgan s discovery of the white eye mutation being sex linked a study led by Sturtevant 1915 concluded that white eyed males were less successful than wild type males in terms of mating with females 69 It was found that the greater the density in eye pigmentation the greater the success in mating for the males of Drosophila melanogaster 69 y yellow The yellow gene is a genetic mutation known as Dmel y within the widely used data base called FlyBase This mutation can be easily identified by the atypical yellow pigment observed in the cuticle of the adult flies and the mouth pieces of the larva 70 The y mutation comprises the following phenotypic classes the mutants that show a complete loss of pigmentation from the cuticle y type and other mutants that show a mosaic pigment pattern with some regions of the cuticle wild type y2 type 71 The role of the yellow gene is diverse and is responsible for changes in behaviour sex specific reproductive maturation and epigenetic reprogramming 72 The y gene is an ideal gene to study as it is visibly clear when an organism has this gene making it easier to understand the passage of DNA to offspring 72 nbsp Wild type wing left vs miniature wing right Genome editGenomic information nbsp D melanogaster chromosomes to scale with megabase pair references oriented as in the National Center for Biotechnology Information database centimorgan distances are approximate and estimated from the locations of selected mapped loci NCBI genome ID47PloidydiploidNumber of chromosomes8Year of completion2015The genome of D melanogaster sequenced in 2000 and curated at the FlyBase database 45 contains four pairs of chromosomes an X Y pair and three autosomes labeled 2 3 and 4 The fourth chromosome is relatively very small and therefore often ignored aside from its important eyeless gene The D melanogaster sequenced genome of 139 5 million base pairs has been annotated 73 and contains around 15 682 genes according to Ensemble release 73 More than 60 of the genome appears to be functional non protein coding DNA 74 involved in gene expression control Determination of sex in Drosophila occurs by the X A ratio of X chromosomes to autosomes not because of the presence of a Y chromosome as in human sex determination Although the Y chromosome is entirely heterochromatic it contains at least 16 genes many of which are thought to have male related functions 75 There are three transferrin orthologs all of which are dramatically divergent from those known in chordate models 76 Similarity to humans edit A March 2000 study by National Human Genome Research Institute comparing the fruit fly and human genome estimated that about 60 of genes are conserved between the two species 77 About 75 of known human disease genes have a recognizable match in the genome of fruit flies 78 and 50 of fly protein sequences have mammalian homologs citation needed An online database called Homophila is available to search for human disease gene homologues in flies and vice versa 79 Drosophila is being used as a genetic model for several human diseases including the neurodegenerative disorders Parkinson s Huntington s spinocerebellar ataxia and Alzheimer s disease 80 The fly is also being used to study mechanisms underlying aging and oxidative stress immunity diabetes and cancer as well as drug abuse 81 82 83 Development editMain article Drosophila embryogenesis The life cycle of this insect has four stages fertilized egg larva pupa and adult 12 Embryogenesis in Drosophila has been extensively studied as its small size short generation time and large brood size makes it ideal for genetic studies It is also unique among model organisms in that cleavage occurs in a syncytium nbsp D melanogaster oogenesisDuring oogenesis cytoplasmic bridges called ring canals connect the forming oocyte to nurse cells Nutrients and developmental control molecules move from the nurse cells into the oocyte In the figure to the left the forming oocyte can be seen to be covered by follicular support cells After fertilization of the oocyte the early embryo or syncytial embryo undergoes rapid DNA replication and 13 nuclear divisions until about 5000 to 6000 nuclei accumulate in the unseparated cytoplasm of the embryo By the end of the eighth division most nuclei have migrated to the surface surrounding the yolk sac leaving behind only a few nuclei which will become the yolk nuclei After the 10th division the pole cells form at the posterior end of the embryo segregating the germ line from the syncytium Finally after the 13th division cell membranes slowly invaginate dividing the syncytium into individual somatic cells Once this process is completed gastrulation starts 84 Nuclear division in the early Drosophila embryo happens so quickly no proper checkpoints exist so mistakes may be made in division of the DNA To get around this problem the nuclei that have made a mistake detach from their centrosomes and fall into the centre of the embryo yolk sac which will not form part of the fly The gene network transcriptional and protein interactions governing the early development of the fruit fly embryo is one of the best understood gene networks to date especially the patterning along the anteroposterior AP and dorsoventral DV axes See under morphogenesis 84 The embryo undergoes well characterized morphogenetic movements during gastrulation and early development including germ band extension formation of several furrows ventral invagination of the mesoderm and posterior and anterior invagination of endoderm gut as well as extensive body segmentation until finally hatching from the surrounding cuticle into a first instar larva During larval development tissues known as imaginal discs grow inside the larva Imaginal discs develop to form most structures of the adult body such as the head legs wings thorax and genitalia Cells of the imaginal disks are set aside during embryogenesis and continue to grow and divide during the larval stages unlike most other cells of the larva which have differentiated to perform specialized functions and grow without further cell division At metamorphosis the larva forms a pupa inside which the larval tissues are reabsorbed and the imaginal tissues undergo extensive morphogenetic movements to form adult structures Developmental plasticity edit Biotic and abiotic factors experienced during development will affect developmental resource allocation leading to phenotypic variation also referred to as developmental plasticity 85 86 As in all insects 86 environmental factors can influence several aspects of development in Drosophila melanogaster 87 88 Fruit flies reared under a hypoxia treatment experience decreased thorax length while hyperoxia produces smaller flight muscles suggesting negative developmental effects of extreme oxygen levels 89 Circadian rhythms are also subject to developmental plasticity Light conditions during development affect daily activity patterns in Drosophila melanogaster where flies raised under constant dark or light are less active as adults than those raised under a 12 hour light dark cycle 90 Temperature is one of the most pervasive factors influencing arthropod development In Drosophila melanogaster temperature induced developmental plasticity can be beneficial and or detrimental 91 92 Most often lower developmental temperatures reduce growth rates which influence many other physiological factors 93 For example development at 25 C increases walking speed thermal performance breadth and territorial success while development at 18 C increases body mass wing size all of which are tied to fitness 88 91 Moreover developing at certain low temperatures produces proportionally large wings which improve flight and reproductive performance at similarly low temperatures See acclimation 94 While certain effects of developmental temperature like body size are irreversible in ectotherms others can be reversible 86 95 When Drosophila melanogaster develop at cold temperatures they will have greater cold tolerance but if cold reared flies are maintained at warmer temperatures their cold tolerance decreases and heat tolerance increases over time 95 96 Because insects typically only mate in a specific range of temperatures their cold heat tolerance is an important trait in maximizing reproductive output 97 While the traits described above are expected to manifest similarly across sexes developmental temperature can also produce sex specific effects in D melanogaster adults Females Ovariole number is significantly affected by developmental temperature in D melanogaster 98 Egg size is also affected by developmental temperature and exacerbated when both parents develop at warm temperatures See Maternal effect 91 Under stressful temperatures these structures will develop to smaller ultimate sizes and decrease a female s reproductive output 98 91 Early fecundity total eggs laid in first 10 days post eclosion is maximized when reared at 25 C versus 17 C and 29 C regardless of adult temperature 99 Across a wide range of developmental temperatures females tend to have greater heat tolerance than males 100 Males Stressful developmental temperatures will cause sterility in D melanogaster males although the upper temperature limit can be increased by maintaining strains at high temperatures See acclimation 92 Male sterility can be reversible if adults are returned to an optimal temperature after developing at stressful temperatures 101 Male flies are smaller and more successful at defending food oviposition sites when reared at 25 C versus 18 C thus smaller males will have increased mating success and reproductive output 88 Sex determination editDrosophila flies have both X and Y chromosomes as well as autosomes Unlike humans the Y chromosome does not confer maleness rather it encodes genes necessary for making sperm Sex is instead determined by the ratio of X chromosomes to autosomes 102 Furthermore each cell decides whether to be male or female independently of the rest of the organism resulting in the occasional occurrence of gynandromorphs X Chromosomes Autosomes Ratio of X A SexXXXX AAAA 1 Normal FemaleXXX AAA 1 Normal FemaleXXY AA 1 Normal FemaleXXYY AA 1 Normal FemaleXX AA 1 Normal FemaleXY AA 0 50 Normal MaleX AA 0 50 Normal Male sterile XXX AA 1 50 MetafemaleXXXX AAA 1 33 MetafemaleXX AAA 0 66 IntersexX AAA 0 33 MetamaleThree major genes are involved in determination of Drosophila sex These are sex lethal sisterless and deadpan Deadpan is an autosomal gene which inhibits sex lethal while sisterless is carried on the X chromosome and inhibits the action of deadpan An AAX cell has twice as much deadpan as sisterless so sex lethal will be inhibited creating a male However an AAXX cell will produce enough sisterless to inhibit the action of deadpan allowing the sex lethal gene to be transcribed to create a female Later control by deadpan and sisterless disappears and what becomes important is the form of the sex lethal gene A secondary promoter causes transcription in both males and females Analysis of the cDNA has shown that different forms are expressed in males and females Sex lethal has been shown to affect the splicing of its own mRNA In males the third exon is included which encodes a stop codon causing a truncated form to be produced In the female version the presence of sex lethal causes this exon to be missed out the other seven amino acids are produced as a full peptide chain again giving a difference between males and females 103 Presence or absence of functional sex lethal proteins now go on to affect the transcription of another protein known as doublesex In the absence of sex lethal doublesex will have the fourth exon removed and be translated up to and including exon 6 DSX M ale while in its presence the fourth exon which encodes a stop codon will produce a truncated version of the protein DSX F emale DSX F causes transcription of Yolk proteins 1 and 2 in somatic cells which will be pumped into the oocyte on its production Immunity editThe D melanogaster immune system can be divided into two responses humoral and cell mediated The former is a systemic response mediated in large part through the toll and Imd pathways which are parallel systems for detecting microbes Other pathways including the stress response pathways JAK STAT and P38 nutritional signalling via FOXO and JNK cell death signalling are all involved in key physiological responses to infection D melanogaster has an organ called the fat body which is analogous to the human liver The fat body is the primary secretory organ and produces key immune molecules upon infection such as serine proteases and antimicrobial peptides AMPs AMPs are secreted into the hemolymph and bind infectious bacteria and fungi killing them by forming pores in their cell walls or inhibiting intracellular processes The cellular immune response instead refers to the direct activity of blood cells hemocytes in Drosophila which are analogous to mammalian monocytes macrophages Hemocytes also possess a significant role in mediating humoral immune responses such as the melanization reaction 104 The immune response to infection can involve up to 2 423 genes or 13 7 of the genome Although the fly s transcriptional response to microbial challenge is highly specific to individual pathogens Drosophila differentially expresses a core group of 252 genes upon infection with most bacteria This core group of genes is associated with gene ontology categories such as antimicrobial response stress response secretion neuron like reproduction and metabolism among others 105 106 Drosophila also possesses several immune mechanisms to both shape the microbiota and prevent excessive immune responses upon detection of microbial stimuli For instance secreted PGRPs with amidase activity scavenge and degrade immunostimulatory DAP type PGN in order to block Imd activation 107 Unlike mammals Drosophila have innate immunity but lack an adaptive immune response However the core elements of this innate immune response are conserved between humans and fruit flies As a result the fruit fly offers a useful model of innate immunity for disentangling genetic interactions of signalling and effector function as flies do not have to contend with interference of adaptive immune mechanisms that could confuse results Various genetic tools protocols and assays make Drosophila a classical model for studying the innate immune system 108 which has even included immune research on the international space station 109 JAK STAT signalling edit Multiple elements of the Drosophila JAK STAT signalling pathway bear direct homology to human JAK STAT pathway genes JAK STAT signalling is induced upon various organismal stresses such as heat stress dehydration or infection JAK STAT induction leads to the production of a number of stress response proteins including Thioester containing proteins TEPs 110 Turandots 111 and the putative antimicrobial peptide Listericin 112 The mechanisms through which many of these proteins act is still under investigation For instance the TEPs appear to promote phagocytosis of Gram positive bacteria and the induction of the toll pathway As a consequence flies lacking TEPs are susceptible to infection by toll pathway challenges 110 source source source source source source Drosophila hemocytes green engulfing Escherichia coli bacteria red The cellular response to infection edit Circulating hemocytes are key regulators of infection This has been demonstrated both through genetic tools to generate flies lacking hemocytes or through injecting microglass beads or lipid droplets that saturate hemocyte ability to phagocytose a secondary infection 113 114 Flies treated like this fail to phagocytose bacteria upon infection and are correspondingly susceptible to infection 115 These hemocytes derive from two waves of hematopoiesis one occurring in the early embryo and one occurring during development from larva to adult 116 However Drosophila hemocytes do not renew over the adult lifespan and so the fly has a finite number of hemocytes that decrease over the course of its lifespan 117 Hemocytes are also involved in regulating cell cycle events and apoptosis of aberrant tissue e g cancerous cells by producing Eiger a tumor necrosis factor signalling molecule that promotes JNK signalling and ultimately cell death and apoptosis 118 Behavioral genetics and neuroscience editIn 1971 Ron Konopka and Seymour Benzer published Clock mutants of Drosophila melanogaster a paper describing the first mutations that affected an animal s behavior Wild type flies show an activity rhythm with a frequency of about a day 24 hours They found mutants with faster and slower rhythms as well as broken rhythms flies that move and rest in random spurts Work over the following 30 years has shown that these mutations and others like them affect a group of genes and their products that form a biochemical or biological clock This clock is found in a wide range of fly cells but the clock bearing cells that control activity are several dozen neurons in the fly s central brain Since then Benzer and others have used behavioral screens to isolate genes involved in vision olfaction audition learning memory courtship pain and other processes such as longevity Following the pioneering work of Alfred Henry Sturtevant 119 and others Benzer and colleagues 46 used sexual mosaics to develop a novel fate mapping technique This technique made it possible to assign a particular characteristic to a specific anatomical location For example this technique showed that male courtship behavior is controlled by the brain 46 Mosaic fate mapping also provided the first indication of the existence of pheromones in this species 120 Males distinguish between conspecific males and females and direct persistent courtship preferentially toward females thanks to a female specific sex pheromone which is mostly produced by the female s tergites The first learning and memory mutants dunce rutabaga etc were isolated by William Chip Quinn while in Benzer s lab and were eventually shown to encode components of an intracellular signaling pathway involving cyclic AMP protein kinase A and a transcription factor known as CREB These molecules were shown to be also involved in synaptic plasticity in Aplysia and mammals 121 The Nobel Prize in Physiology or Medicine for 2017 was awarded to Jeffrey C Hall Michael Rosbash Michael W Young for their works using fruit flies in understanding the molecular mechanisms controlling the circadian rhythm 122 Male flies sing to the females during courtship using their wings to generate sound and some of the genetics of sexual behavior have been characterized In particular the fruitless gene has several different splice forms and male flies expressing female splice forms have female like behavior and vice versa The TRP channels nompC nanchung and inactive are expressed in sound sensitive Johnston s organ neurons and participate in the transduction of sound 123 124 Mutating the Genderblind gene also known as CG6070 alters the sexual behavior of Drosophila turning the flies bisexual 125 Flies use a modified version of Bloom filters to detect novelty of odors with additional features including similarity of novel odor to that of previously experienced examples and time elapsed since previous experience of the same odor 126 Aggression edit As with most insects aggressive behaviors between male flies commonly occur in the presence of courting a female and when competing for resources Such behaviors often involve raising wings and legs towards the opponent and attacking with the whole body 127 Thus it often causes wing damage which reduces their fitness by removing their ability to fly and mate 128 Acoustic communication edit In order for aggression to occur male flies produce sounds to communicate their intent A 2017 study found that songs promoting aggression contain pulses occurring at longer intervals 129 RNA sequencing from fly mutants displaying over aggressive behaviors found more than 50 auditory related genes important for transient receptor potentials Ca2 signaling and mechanoreceptor potentials to be upregulated in the AB neurons located in Johnston s organ 129 In addition aggression levels were reduced when these genes were knocked out via RNA interference 129 This signifies the major role of hearing as a sensory modality in communicating aggression Pheromone signaling edit Other than hearing another sensory modality that regulates aggression is pheromone signaling which operates through either the olfactory system or the gustatory system depending on the pheromone 130 An example is cVA an anti aphrodisiac pheromone used by males to mark females after copulation and to deter other males from mating 131 This male specific pheromone causes an increase in male male aggression when detected by another male s gustatory system 130 However upon inserting a mutation that makes the flies irresponsive to cVA no aggressive behaviors were seen 132 This shows how there are multiple modalities for promoting aggression in flies Competition for food edit Specifically when competing for food aggression occurs based on amount of food available and is independent of any social interactions between males 133 Specifically sucrose was found to stimulate gustatory receptor neurons which was necessary to stimulate aggression 133 However once the amount of food becomes greater than a certain amount the competition between males lowers 133 This is possibly due to an over abundance of food resources On a larger scale food was found to determine the boundaries of a territory since flies were observed to be more aggressive at the food s physical perimeter Effect of sleep deprivation edit However like most behaviors requiring arousal and wakefulness aggression was found to be impaired via sleep deprivation Specifically this occurs through the impairment of Octopamine and dopamine signaling which are important pathways for regulating arousal in insects 134 135 Due to reduced aggression sleep deprived male flies were found to be disadvantaged at mating compared to normal flies 135 However when octopamine agonists were administered upon these sleep deprived flies aggression levels were seen to be increased and sexual fitness was subsequently restored 135 Therefore this finding implicates the importance of sleep in aggression between male flies Vision edit nbsp Stereo images of the eyeThe compound eye of the fruit fly contains 760 unit eyes or ommatidia and are one of the most advanced among insects Each ommatidium contains eight photoreceptor cells R1 8 support cells pigment cells and a cornea Wild type flies have reddish pigment cells which serve to absorb excess blue light so the fly is not blinded by ambient light Eye color genes regulate cellular vesicular transport The enzymes needed for pigment synthesis are then transported to the cell s pigment granule which holds pigment precursor molecules 59 Each photoreceptor cell consists of two main sections the cell body and the rhabdomere The cell body contains the nucleus while the 100 mm long rhabdomere is made up of toothbrush like stacks of membrane called microvilli Each microvillus is 1 2 mm in length and about 60 nm in diameter 136 The membrane of the rhabdomere is packed with about 100 million opsin molecules the visual protein that absorbs light The other visual proteins are also tightly packed into the microvilli leaving little room for cytoplasm Opsins and spectral sensitivity edit nbsp The arrangement of the photoreceptor cells in an pale and yellow ommatidia of Drosophila melanogaster The top row shows two of the six outer photoreceptor cells R1 R6 and the inner R7 and R8 cells The bottom row shows the different opsins Rh1 Rh3 Rh4 Rh5 and Rh6 the cells express Figure from Sharkey et al 2020 137 nbsp Expression of the opsin Rh1 in the photoreceptor cells R1 R6The genome of Drosophila encodes seven opsins 138 five of those are expressed in the omatidia of the eye The photoreceptor cells R1 R6 express the opsin Rh1 139 which absorbs maximally blue light around 480 nm 140 141 142 however the R1 R6 cells cover a broader range of the spectrum than an opsin would allow due to a sensitising pigment 143 144 that adds two sensitivity maxima in the UV range 355 and 370 nm 142 The R7 cells come in two types with yellow and pale rhabdomeres R7y and R7p 145 146 The pale R7p cells express the opsin Rh3 147 148 which maximally absorbs UV light 345 nm 149 The R7p cells are strictly paired with the R8p cells that express Rh5 148 which maximally absorbs violet light 437 nm 142 The other the yellow R7y cells express a blue absorbing screening pigment 145 and the opsin Rh4 150 which maximally absorbs UV light 375 nm 149 The R7y cells are strictly paired with R8y cells that express Rh6 151 which maximally absorbs UV light 508 nm 142 In a subset of omatidia both R7 and R8 cells express the opsin Rh3 148 However these absorption maxima of the opsins where measured in white eyed flies without screening pigments Rh3 Rh6 149 142 or from the isolated opsin directly Rh1 140 Those pigments reduce the light that reaches the opsins depending on the wavelength Thus in fully pigmented flies the effective absorption maxima of opsins differs and thus also the sensitivity of their photoreceptor cells With screening pigment the opsin Rh3 is short wave shifted from 345 nm b to 330 nm and Rh4 from 375 nm to 355 nm Whether screening pigment is present does not make a practical difference for the opsin Rh5 435 nm and 437 nm while the opsin R6 is long wave shifted by 92 nm from 508 nm to 600 nm 137 Additionally of the opsins of the eye Drosophila has two more opsins The ocelli express the opsin Rh2 152 153 which maximally absorbs violet light 420 nm 153 And the opsin Rh7 which maximally absorbs UV light 350 nm with an unusually long wavelength tail up to 500 nm The long tail disappears if a lysine at position 90 is replaced by glutamic acid This mutant then absorbs maximally violet light 450 nm 154 The opsin Rh7 entrains with cryptochrome the circadian rhythm of Drosophila to the day night cycle in the central pacemaker neurons 155 Each Drosophila opsin binds the carotenoid chromophore 11 cis 3 hydroxyretinal via a lysine 156 157 This lysine is conserved in almost all opsins only a few opsins have lost it during evolution 158 Opsins without it are not light sensitive 159 160 161 In particular the Drosophila opsins Rh1 Rh4 and Rh7 function not only as photoreceptors but also as chemoreceptors for aristolochic acid These opsins still have the lysine like other opsins However if it is replaced by an arginine in Rh1 then Rh1 loses light sensitivity but still responds to aristolochic acid Thus the lysine is not needed for Rh1 to function as chemoreceptor 160 Spectral sensitivities of Drosophila melanogaster opsins in photoreceptor cells of white and red eyed flies 137 nbsp Spectral sensitivities of Drosophila melanogaster opsins in white eyed flies The sensitivities of Rh3 R6 are modelled with opsin templates and sensitivity estimates from Salcedo et al 1999 142 The opsin Rh1 redrawn from Salcedo et al 142 has a characteristic shape as it is coupled to a UV sensitising pigment nbsp Normalized mean spectral sensitivity curves of Drosophila melanogaster opsins Rh1 Rh3 Rh4 Rh5 and Rh6 measured in their native photoreceptor cells in red eye flies with screening pigment Each spectral curve is the average from six flies Phototransduction edit As in vertebrate vision visual transduction in invertebrates occurs via a G protein coupled pathway However in vertebrates the G protein is transducin while the G protein in invertebrates is Gq dgq in Drosophila When rhodopsin Rh absorbs a photon of light its chromophore 11 cis 3 hydroxyretinal is isomerized to all trans 3 hydroxyretinal Rh undergoes a conformational change into its active form metarhodopsin Metarhodopsin activates Gq which in turn activates a phospholipase Cb PLCb known as NorpA 162 PLCb hydrolyzes phosphatidylinositol 4 5 bisphosphate PIP2 a phospholipid found in the cell membrane into soluble inositol triphosphate IP3 and diacylglycerol DAG which stays in the cell membrane DAG a derivative of DAG or PIP2 depletion cause a calcium selective ion channel known as transient receptor potential TRP to open and calcium and sodium flows into the cell 163 IP3 is thought to bind to IP3 receptors in the subrhabdomeric cisternae an extension of the endoplasmic reticulum and cause release of calcium but this process does not seem to be essential for normal vision 162 Calcium binds to proteins such as calmodulin CaM and an eye specific protein kinase C PKC known as InaC These proteins interact with other proteins and have been shown to be necessary for shut off of the light response In addition proteins called arrestins bind metarhodopsin and prevent it from activating more Gq A sodium calcium exchanger known as CalX pumps the calcium out of the cell It uses the inward sodium gradient to export calcium at a stoichiometry of 3 Na 1 Ca 164 TRP InaC and PLC form a signaling complex by binding a scaffolding protein called InaD InaD contains five binding domains called PDZ domain proteins which specifically bind the C termini of target proteins Disruption of the complex by mutations in either the PDZ domains or the target proteins reduces the efficiency of signaling For example disruption of the interaction between InaC the protein kinase C and InaD results in a delay in inactivation of the light response Unlike vertebrate metarhodopsin invertebrate metarhodopsin can be converted back into rhodopsin by absorbing a photon of orange light 580 nm About two thirds of the Drosophila brain is dedicated to visual processing 165 Although the spatial resolution of their vision is significantly worse than that of humans their temporal resolution is around 10 times better Grooming edit Drosophila are known to exhibit grooming behaviors that are executed in a predictable manner Drosophila consistently begin a grooming sequence by using their front legs to clean the eyes then the head and antennae Using their hind legs Drosophila proceed to groom their abdomen and finally the wings and thorax Throughout this sequence Drosophila periodically rub their legs together to get rid of excess dust and debris that accumulates during the grooming process 166 Grooming behaviors have been shown to be executed in a suppression hierarchy This means that grooming behaviors that occur at the beginning of the sequence prevent those that come later in the sequence from occurring simultaneously as the grooming sequence consists of mutually exclusive behaviors 167 168 This hierarchy does not prevent Drosophila from returning to grooming behaviors that have already been accessed in the grooming sequence 167 The order of grooming behaviors in the suppression hierarchy is thought to be related to the priority of cleaning a specific body part For example the eyes and antennae are likely executed early on in the grooming sequence to prevent debris from interfering with the function of D melanogaster s sensory organs 167 168 Walking edit nbsp Top view of a walking Drosophila left with legs tracked with DeepLabCut 169 right Like many other hexapod insects Drosophila typically walk using a tripod gait 170 This means that three of the legs swing together while the other three remain stationary or in stance Specifically the middle leg moves in phase with the contralateral front and hind legs However variability around the tripod configuration exists along a continuum meaning that flies do not exhibit distinct transitions between different gaits 171 At fast walking speeds the walking configuration is mostly tripod 3 legs in stance but at slower walking speeds flies are more likely to have four tetrapod or five legs in stance wave 172 173 These transitions may help to optimize static stability 174 Because flies are so small inertial forces are negligible compared with the elastic forces of their muscles and joints or the viscous forces of the surrounding air 175 Flight edit Flies fly via straight sequences of movement interspersed by rapid turns called saccades 176 During these turns a fly is able to rotate 90 in less than 50 milliseconds 176 Characteristics of Drosophila flight may be dominated by the viscosity of the air rather than the inertia of the fly body but the opposite case with inertia as the dominant force may occur 176 However subsequent work showed that while the viscous effects on the insect body during flight may be negligible the aerodynamic forces on the wings themselves actually cause fruit flies turns to be damped viscously 177 Connectome editDrosophila is one of the few animals C elegans being another where detailed neural circuits a connectome are available A high level connectome at the level of brain compartments and interconnecting tracts of neurons exists for the full fly brain 178 A version of this is available online 179 Detailed circuit level connectomes exist for the lamina 180 181 and a medulla 182 column both in the visual system of the fruit fly and the alpha lobe of the mushroom body 183 In May 2017 a paper published in bioRxiv presented an electron microscopy image stack of the whole adult female brain at synaptic resolution The volume is available for sparse tracing of selected circuits 184 185 Since then multiple datasets have been collected including a dense connectome of half the central brain of Drosophila in 2020 186 187 and a dense connectome of the entire female adult nerve cord in 2021 188 Generally these datasets are acquired by sectioning the tissue e g the brain into thin sections on order of ten or hundreds of nanometers Each section is then imaged using an electron microscope and these images are stitched and aligned together to create a 3D image volume The methods used in reconstruction and initial analysis of the such datasets followed 189 Due to advancements in deep learning automated methods for image segmentation have made large scale reconstruction providing dense reconstructions of all the neurites within the volume 190 Furthermore the resolution of electron microscopy illuminates ultrastructural variations between neurons as well as the location of individual synapses thereby providing a wiring diagram of synaptic connectivity between all neurites within the given dataset In 2023 the complete map of a Drosophila larval brain at the synapse level and an analysis of its architecture was published The larval brain consists of 3016 neurons and 548 000 synaptic sites 191 whereas the adult brain has about 150 000 neurons and 150 million synapses Misconceptions editDrosophila is sometimes referred to as a pest due to its tendency to live in human settlements where fermenting fruit is found Flies may collect in homes restaurants stores and other locations 13 The name and behavior of this species of fly have led to the misconception that it is a biological security risk in Australia and elsewhere While other fruit fly species do pose a risk D melanogaster is attracted to fruit that is already rotting rather than causing fruit to rot 192 193 See also editAnimal testing on invertebrates Eating behavior in Insects Measurement Fruit flies in space Genetically modified insect Gynandromorphism JETLAG gene List of Drosophila databases Spatzle gene Transgenesis Zebrafish another widely used model organism in scientific researchNotes edit Vinegar fly is preferred by a handful of recent publications as being a more accurate description than fruit fly 2 3 4 Sharkey et al 137 give the absorption maximum of Rh3 as 334 nm in their result section However in the introduction and the material and methods section they give it as 345 nm For both values they cite Feiler et al who reported 345 nm only 149 Therefore this seems to be a mistake and they probably meant there 345 nm too References edit Meigen JW 1830 Systematische Beschreibung der bekannten europaischen zweiflugeligen Insekten Volume 6 PDF in German Schulz Wundermann Archived from the original PDF on February 9 2012 Drosophila insect genus Encyclopedia Britannica Retrieved October 30 2021 Vinegar Flies Penn State Extension Retrieved October 30 2021 a b Green MM September 2002 It really is not a fruit fly Genetics 162 1 1 3 doi 10 1093 genetics 162 1 1 PMC 1462251 PMID 12242218 Schilthuizen Menno April 28 2015 Semen s Chemical Cocktail Can Hijack a Mate s Brain Discover Retrieved September 11 2023 Even in the ejaculate of the lowly banana fly Drosophila melanogaster researchers have identified no fewer than 133 different kinds of proteins T H Morgan s Nobel Prize biography mentioning C W Woodworth Holden B January 1 2015 Charles W Woodworth The Remarkable Life of U C s First Entomologist 1st ed Brian Holden Publishing pp 135 137 ISBN 978 0 9864105 3 6 Nobel Prizes The Guardian October 7 2017 FruitFly ResearchGate a b Sang JH Reeve EC June 23 2001 Drosophila melanogaster The Fruit Fly Encyclopedia of genetics USA Fitzroy Dearborn Publishers I p 157 ISBN 978 1 884964 34 3 Retrieved July 1 2009 Baudry E Viginier B Veuille M August 2004 Non African populations of Drosophila melanogaster have a unique origin Molecular Biology and Evolution 21 8 1482 91 doi 10 1093 molbev msh089 PMID 15014160 a b Markow TA June 2015 The secret lives of Drosophila flies eLife 4 doi 10 7554 eLife 06793 PMC 4454838 PMID 26041333 a b Vinegar Flies Drosophila species Family Drosophilidae Department of Entomology College of Agricultural Sciences Pennsylvania State University 2017 Retrieved July 20 2017 a b Ewart GD Howells AJ January 1 1998 ABC transporters involved in transport of eye pigment precursors in Drosophila melanogaster ABC Transporters Biochemical Cellular and Molecular Aspects Methods in Enzymology Vol 292 Academic Press pp 213 24 doi 10 1016 S0076 6879 98 92017 1 ISBN 978 0 12 182193 7 PMID 9711556 FlyBase A database of Drosophila genes and genomes Genetics Society of America 2009 Archived from the original on August 15 2009 Retrieved August 11 2009 Yuzuki Keven Tidon Rosana 2020 Identification key for drosophilid species Diptera Drosophilidae exotic to the Neotropical Region and occurring in Brazil Revista Brasileira de Entomologia 64 1 doi 10 1590 1806 9665 rbent 2019 100 ISSN 1806 9665 S2CID 211570766 Miller M E Marshall S A Grimaldi D A 2017 A Review of the Species of Drosophila Diptera Drosophilidae and Genera of Drosophilidae of Northeastern North America Canadian Journal of Arthropod Identification 31 doi 10 3752 cjai 2017 31 Drosophila Melanogaster Animal Diversity Web 2000 Archived from the original on November 30 2014 Retrieved August 11 2009 Linford NJ Bilgir C Ro J Pletcher SD January 2013 Measurement of lifespan in Drosophila melanogaster Journal of Visualized Experiments 71 doi 10 3791 50068 PMC 3582515 PMID 23328955 a b c d e f g Ashburner M Thompson JN 1978 The laboratory culture of Drosophila In Ashburner M Wright TRF ed The genetics and biology ofDrosophila Vol 2A Academic Press 1 81 a b c d e f g Ashburner M Golic KG Hawley RS 2005 Drosophila A Laboratory Handbook 2nd ed Cold Spring Harbor Laboratory Press pp 162 4 ISBN 978 0 87969 706 8 Bloomington Drosophila Stock Center at Indiana University Basic Methods of Culturing Drosophila Archived 2006 09 01 at the Wayback Machine a b Chiang HC Hodson AC 1950 An analytical study of population growth in Drosophila melanogaster Ecological Monographs 20 3 173 206 Bibcode 1950EcoM 20 173C doi 10 2307 1948580 JSTOR 1948580 Bakker K 1961 An analysis of factors which determine success in competition for food among larvae of Drosophila melanogaster Archives Neerlandaises de Zoologie 14 2 200 281 doi 10 1163 036551661X00061 S2CID 85129022 Fernandez Moreno MA Farr CL Kaguni LS Garesse R 2007 Drosophila melanogaster as a Model System to Study Mitochondrial Biology Mitochondria Methods in Molecular Biology Vol 372 pp 33 49 doi 10 1007 978 1 59745 365 3 3 ISBN 978 1 58829 667 2 PMC 4876951 PMID 18314716 Blum JE Fischer CN Miles J Handelsman J November 2013 Frequent replenishment sustains the beneficial microbiome of Drosophila melanogaster mBio 4 6 e00860 13 doi 10 1128 mBio 00860 13 PMC 3892787 PMID 24194543 Cook R Connolly K 1973 Rejection Responses by Female Drosophila melanogaster Their Ontogeny Causality and Effects upon the Behaviour of the Courting Male Behaviour 44 1 2 142 166 doi 10 1163 156853973x00364 JSTOR 4533484 S2CID 85393769 Houot B Svetec N Godoy Herrera R Ferveur JF July 2010 Effect of laboratory acclimation on the variation of reproduction related characters in Drosophila melanogaster The Journal of Experimental Biology 213 Pt 13 2322 31 doi 10 1242 jeb 041566 PMID 20543131 Gilbert SF 2006 9 Fertilization in Drosophila Developmental Biology 8th ed Sinauer Associates ISBN 978 0 87893 250 4 Archived from the original on February 7 2007 a b c Price CS Dyer KA Coyne JA July 1999 Sperm competition between Drosophila males involves both displacement and incapacitation Nature 400 6743 449 52 Bibcode 1999Natur 400 449P doi 10 1038 22755 PMID 10440373 S2CID 4393369 a b c d Fruit fly research may reveal what happens in female brains during courtship mating Retrieved October 5 2014 Meiselman M Lee SS Tran RT Dai H Ding Y Rivera Perez C et al May 2017 Drosophila melanogaster Proceedings of the National Academy of Sciences of the United States of America 114 19 E3849 E3858 doi 10 1073 pnas 1620760114 PMC 5441734 PMID 28439025 Moshitzky P Fleischmann I Chaimov N Saudan P Klauser S Kubli E Applebaum SW 1996 Sex peptide activates juvenile hormone biosynthesis in the Drosophila melanogaster corpus allatum Archives of Insect Biochemistry and Physiology 32 3 4 363 74 doi 10 1002 SICI 1520 6327 1996 32 3 4 lt 363 AID ARCH9 gt 3 0 CO 2 T PMID 8756302 Carnes MU Campbell T Huang W Butler DG Carbone MA Duncan LH et al 2015 The Genomic Basis of Postponed Senescence in Drosophila melanogaster PLOS ONE 10 9 e0138569 Bibcode 2015PLoSO 1038569C doi 10 1371 journal pone 0138569 PMC 4574564 PMID 26378456 Cassidy D Epiney DG Salameh C Zhou LT Salomon RN Schirmer AE et al November 2019 Evidence for premature aging in a Drosophila model of Werner syndrome Experimental Gerontology 127 110733 doi 10 1016 j exger 2019 110733 PMC 6935377 PMID 31518666 Pitnick S 1996 Investment in testes and the cost of making long sperm in Drosophila American Naturalist 148 57 80 doi 10 1086 285911 S2CID 83654824 Dagaeff AC Pocheville A Nobel S Loyau A Isabel G Danchin E 2016 Drosophila mate copying correlates with atmospheric pressure in a speed learning situation Animal Behaviour 121 163 174 doi 10 1016 j anbehav 2016 08 022 Dukas R 2004 Male fruit flies learn to avoid interspecific courtship Behavioral Ecology 15 4 695 698 doi 10 1093 beheco arh068 Saleem S Ruggles PH Abbott WK Carney GE 2014 Sexual experience enhances Drosophila melanogaster male mating behavior and success PLOS ONE 9 5 e96639 Bibcode 2014PLoSO 996639S doi 10 1371 journal pone 0096639 PMC 4013029 PMID 24805129 a b von Haartman L 1951 Successive Polygamy Behaviour 3 1 256 273 doi 10 1163 156853951x00296 a b c d e f g Vartak VR Varma V Sharma VK February 2015 Effects of polygamy on the activity rest rhythm of male fruit flies Drosophila melanogaster Die Naturwissenschaften 102 1 2 1252 Bibcode 2015SciNa 102 3V doi 10 1007 s00114 014 1252 5 PMID 25604736 S2CID 7529509 a b Bateman AJ December 1948 Intra sexual selection in Drosophila Heredity 2 Pt 3 349 68 doi 10 1038 hdy 1948 21 PMID 18103134 a b Pierce BA 2004 Genetics A Conceptual Approach 2nd ed W H Freeman ISBN 978 0 7167 8881 2 Kilbey BJ MacDonald DJ Auerbach C Sobels FH Vogel EW June 1981 The use of Drosophila melanogaster in tests for environmental mutagens Mutation Research 85 3 141 6 doi 10 1016 0165 1161 81 90029 7 PMID 6790982 a b Adams MD Celniker SE Holt RA Evans CA Gocayne JD Amanatides PG et al March 2000 The genome sequence of Drosophila melanogaster Science 287 5461 2185 95 Bibcode 2000Sci 287 2185 CiteSeerX 10 1 1 549 8639 doi 10 1126 science 287 5461 2185 PMID 10731132 a b c Hotta Y Benzer S December 1972 Mapping of behaviour in Drosophila mosaics Nature 240 5383 527 35 Bibcode 1972Natur 240 527H doi 10 1038 240527a0 PMID 4568399 S2CID 4181921 Meiers S 2018 Exploiting emerging DNA sequencing technologies to study genomic rearrangements archiv ub uni heidelberg de doi 10 11588 heidok 00024506 Retrieved June 28 2021 Azpiazu N Frasch M July 1993 tinman and bagpipe two homeo box genes that determine cell fates in the dorsal mesoderm of Drosophila Genes amp Development 7 7B 1325 40 doi 10 1101 gad 7 7b 1325 PMID 8101173 Stern DL Frankel N December 2013 The structure and evolution of cis regulatory regions the shavenbaby story Philosophical Transactions of the Royal Society of London Series B Biological Sciences 368 1632 20130028 doi 10 1098 rstb 2013 0028 PMC 3826501 PMID 24218640 a b Phillips AM Smart R Strauss R Brembs B Kelly LE May 2005 The Drosophila black enigma the molecular and behavioural characterization of the black1 mutant allele PDF Gene 351 131 42 doi 10 1016 j gene 2005 03 013 PMID 15878647 a b FlyBase Gene Report Dmel b flybase org Retrieved March 26 2019 Sherald AF September 1981 Intergenic suppression of the black mutation of Drosophila melanogaster Molecular amp General Genetics 183 1 102 6 doi 10 1007 bf00270146 PMID 6799739 S2CID 1210971 Shoup JR May 1966 The development of pigment granules in the eyes of wild type and mutant Drosophila melanogaster The Journal of Cell Biology 29 2 223 49 doi 10 1083 jcb 29 2 223 PMC 2106902 PMID 5961338 TEACHER REFERENCE PAGES FLY EYE PIGMENTS LAB PDF Morgan TH March 1911 The Origin of Nine Wing Mutations in Drosophila Science 33 848 496 9 Bibcode 1911Sci 33 496M doi 10 1126 science 33 848 496 JSTOR 1638587 PMID 17774436 FlyBase Gene Report Dmel m flybase org Retrieved March 26 2019 Bilousov OO Katanaev VL Demydov SV Kozeretska IA March April 2013 The downregulation of the miniature gene does not replicate miniature loss of function phenotypes in Drosophila melanogaster wing to the full extent TSitologiia I Genetika 47 2 77 81 PMID 23745366 a b Kim J Suh H Kim S Kim K Ahn C Yim J September 2006 Identification and characteristics of the structural gene for the Drosophila eye colour mutant sepia encoding PDA synthase a member of the omega class glutathione S transferases The Biochemical Journal 398 3 451 60 doi 10 1042 BJ20060424 PMC 1559464 PMID 16712527 a b c Grant P Maga T Loshakov A Singhal R Wali A Nwankwo J et al October 2016 An Eye on Trafficking Genes Identification of Four Eye Color Mutations in Drosophila G3 6 10 3185 3196 doi 10 1534 g3 116 032508 PMC 5068940 PMID 27558665 Wiederrecht GJ Brown GM 1984 Purification and properties of the enzymes from Drosophila melanogaster that catalyze the conversion of dihydroneopterin triphosphate to the pyrimidodiazepine precursor of the drosopterins J Biol Chem 259 22 14121 7 doi 10 1016 S0021 9258 18 89865 9 PMID 6438092 Inheritance Patterns in Drosophila Melanogaster Retrieved March 26 2019 a b Green MM April 1952 Mutant Isoalleles at the Vermilion Locus in Drosophila Melanogaster Proceedings of the National Academy of Sciences of the United States of America 38 4 300 5 Bibcode 1952PNAS 38 300G doi 10 1073 pnas 38 4 300 PMC 1063551 PMID 16589094 Oxenkrug GF January 2010 The extended life span of Drosophila melanogaster eye color white and vermilion mutants with impaired formation of kynurenine Journal of Neural Transmission 117 1 23 26 doi 10 1007 s00702 009 0341 7 PMC 3013506 PMID 19941150 Simon E Faucheux C Zider A Theze N Thiebaud P July 2016 From vestigial to vestigial like the Drosophila gene that has taken wing Development Genes and Evolution 226 4 297 315 doi 10 1007 s00427 016 0546 3 PMID 27116603 S2CID 16651247 Tomoyasu Y Ohde T Clark Hachtel C March 14 2017 What serial homologs can tell us about the origin of insect wings F1000Research 6 268 doi 10 12688 f1000research 10285 1 PMC 5357031 PMID 28357056 Williams JA Bell JB Carroll SB December 1991 Control of Drosophila wing and haltere development by the nuclear vestigial gene product Genes amp Development 5 12B 2481 95 doi 10 1101 gad 5 12b 2481 PMID 1752439 Green MM January 2010 2010 A century of Drosophila genetics through the prism of the white gene Genetics 184 1 3 7 doi 10 1534 genetics 109 110015 PMC 2815926 PMID 20061564 Ferreiro MJ Perez C Marchesano M Ruiz S Caputi A Aguilera P et al 2018 rosophila melanogaster White Mutant w1118 Undergo Retinal Degeneration Frontiers in Neuroscience 11 732 doi 10 3389 fnins 2017 00732 PMC 5758589 PMID 29354028 a b Xiao C Qiu S Robertson RM August 2017 The white gene controls copulation success in Drosophila melanogaster Scientific Reports 7 1 7712 Bibcode 2017NatSR 7 7712X doi 10 1038 s41598 017 08155 y PMC 5550479 PMID 28794482 Gene Dmel y Flybase org The FlyBase Consortium Retrieved March 26 2019 Wittkopp PJ True JR Carroll SB April 2002 Reciprocal functions of the Drosophila yellow and ebony proteins in the development and evolution of pigment patterns Development 129 8 1849 58 doi 10 1242 dev 129 8 1849 PMID 11934851 a b Biessmann H November 1985 Molecular analysis of the yellow gene y region of Drosophila melanogaster Proceedings of the National Academy of Sciences of the United States of America 82 21 7369 73 Bibcode 1985PNAS 82 7369B doi 10 1073 pnas 82 21 7369 PMC 391346 PMID 3933004 NCBI National Center for Biotechnology Information Genome Database Retrieved November 30 2011 Halligan DL Keightley PD July 2006 Ubiquitous selective constraints in the Drosophila genome revealed by a genome wide interspecies comparison Genome Research 16 7 875 84 doi 10 1101 gr 5022906 PMC 1484454 PMID 16751341 Carvalho AB December 2002 Origin and evolution of the Drosophila Y chromosome Current Opinion in Genetics amp Development 12 6 664 8 doi 10 1016 S0959 437X 02 00356 8 PMID 12433579 Gabaldon T Koonin EV May 2013 Functional and evolutionary implications of gene orthology Nature Reviews Genetics Nature Portfolio 14 5 360 366 doi 10 1038 nrg3456 PMC 5877793 PMID 23552219 Background on Comparative Genomic Analysis US National Human Genome Research Institute December 2002 Reiter LT Potocki L Chien S Gribskov M Bier E June 2001 A systematic analysis of human disease associated gene sequences in Drosophila melanogaster Genome Research 11 6 1114 25 doi 10 1101 gr 169101 PMC 311089 PMID 11381037 Chien S Reiter LT Bier E Gribskov M January 2002 Homophila human disease gene cognates in Drosophila Nucleic Acids Research 30 1 149 51 doi 10 1093 nar 30 1 149 PMC 99119 PMID 11752278 Jaiswal M Sandoval H Zhang K Bayat V Bellen HJ 2012 Probing mechanisms that underlie human neurodegenerative diseases in Drosophila Annual Review of Genetics 46 371 96 doi 10 1146 annurev genet 110711 155456 PMC 3663445 PMID 22974305 Pick L 2017 Fly Models of Human Diseases Volume 121 of Current Topics in Developmental Biology Academic Press ISBN 978 0 12 802905 3 Buchon N Silverman N Cherry S December 2014 Immunity in Drosophila melanogaster from microbial recognition to whole organism physiology Nature Reviews Immunology 14 12 796 810 doi 10 1038 nri3763 PMC 6190593 PMID 25421701 Kaun KR Devineni AV Heberlein U June 2012 Drosophila melanogaster as a model to study drug addiction Human Genetics 131 6 959 75 doi 10 1007 s00439 012 1146 6 PMC 3351628 PMID 22350798 a b Weigmann K Klapper R Strasser T Rickert C Technau G Jackle H et al June 2003 FlyMove a new way to look at development of Drosophila Trends in Genetics 19 6 310 1 doi 10 1016 S0168 9525 03 00050 7 PMID 12801722 West Eberhard MJ May 2005 Developmental plasticity and the origin of species differences Proceedings of the National Academy of Sciences of the United States of America 102 suppl 1 6543 9 Bibcode 2005PNAS 102 6543W doi 10 1073 pnas 0501844102 PMC 1131862 PMID 15851679 a b c Abram PK Boivin G Moiroux J Brodeur J November 2017 Behavioural effects of temperature on ectothermic animals unifying thermal physiology and behavioural plasticity Biological Reviews of the Cambridge Philosophical Society 92 4 1859 1876 doi 10 1111 brv 12312 PMID 28980433 S2CID 9099834 Gibert P Huey RB Gilchrist GW January 2001 Locomotor performance of Drosophila melanogaster interactions among developmental and adult temperatures age and geography Evolution International Journal of Organic Evolution 55 1 205 9 doi 10 1111 j 0014 3820 2001 tb01286 x PMID 11263741 S2CID 2991855 a b c Zamudio KR Huey RB Crill WD 1995 Bigger isn t always better body size developmental and parental temperature and male territorial success in Drosophila melanogaster Animal Behaviour 49 3 671 677 doi 10 1016 0003 3472 95 80200 2 ISSN 0003 3472 S2CID 9124942 Harrison JF Waters JS Biddulph TA Kovacevic A Klok CJ Socha JJ April 2018 Developmental plasticity and stability in the tracheal networks supplying Drosophila flight muscle in response to rearing oxygen level Journal of Insect Physiology The limits of respiratory function external and internal constraints on insect gas exchange 106 Pt 3 189 198 doi 10 1016 j jinsphys 2017 09 006 PMID 28927826 Sheeba V Chandrashekaran MK Joshi A Sharma VK January 2002 Developmental plasticity of the locomotor activity rhythm of Drosophila melanogaster Journal of Insect Physiology 48 1 25 32 doi 10 1016 S0022 1910 01 00139 1 PMID 12770129 a b c d Crill WD Huey RB Gilchrist GW June 1996 Within and Between Generation Effects of Temperature on the Morphology and Physiology of Drosophila Melanogaster Evolution International Journal of Organic Evolution 50 3 1205 1218 doi 10 2307 2410661 JSTOR 2410661 PMID 28565273 a b David JR Araripe LO Chakir M Legout H Lemos B Petavy G et al July 2005 Male sterility at extreme temperatures a significant but neglected phenomenon for understanding Drosophila climatic adaptations Journal of Evolutionary Biology 18 4 838 46 doi 10 1111 j 1420 9101 2005 00914 x PMID 16033555 S2CID 23847613 French V Feast M Partridge L November 1998 Body size and cell size in Drosophila the developmental response to temperature Journal of Insect Physiology 44 11 1081 1089 doi 10 1016 S0022 1910 98 00061 4 PMID 12770407 Frazier MR Harrison JF Kirkton SD Roberts SP July 2008 Cold rearing improves cold flight performance in Drosophila via changes in wing morphology The Journal of Experimental Biology 211 Pt 13 2116 22 doi 10 1242 jeb 019422 PMID 18552301 a b Slotsbo S Schou MF Kristensen TN Loeschcke V Sorensen JG September 2016 Reversibility of developmental heat and cold plasticity is asymmetric and has long lasting consequences for adult thermal tolerance The Journal of Experimental Biology 219 Pt 17 2726 32 doi 10 1242 jeb 143750 PMID 27353229 Gilchrist GW Huey RB January 2001 Parental and developmental temperature effects on the thermal dependence of fitness in Drosophila melanogaster Evolution International Journal of Organic Evolution 55 1 209 14 doi 10 1111 j 0014 3820 2001 tb01287 x PMID 11263742 S2CID 1329035 Austin CJ Moehring AJ May 2013 Optimal temperature range of a plastic species Drosophila simulans The Journal of Animal Ecology 82 3 663 72 Bibcode 2013JAnEc 82 663A doi 10 1111 1365 2656 12041 PMID 23360477 a b Hodin J Riddiford LM October 2000 Different mechanisms underlie phenotypic plasticity and interspecific variation for a reproductive character in drosophilids Insecta Diptera Evolution International Journal of Organic Evolution 54 5 1638 53 doi 10 1111 j 0014 3820 2000 tb00708 x PMID 11108591 S2CID 6875815 Klepsatel P Girish TN Dircksen H Galikova M May 2019 Drosophila is maximised by optimal developmental temperature The Journal of Experimental Biology 222 Pt 10 jeb202184 doi 10 1242 jeb 202184 PMID 31064855 Schou MF Kristensen TN Pedersen A Karlsson BG Loeschcke V Malmendal A February 2017 Metabolic and functional characterization of effects of developmental temperature in Drosophila melanogaster American Journal of Physiology Regulatory Integrative and Comparative Physiology 312 2 R211 R222 doi 10 1152 ajpregu 00268 2016 PMC 5336569 PMID 27927623 Cohet Y David J January 1978 Control of the adult reproductive potential by preimaginal thermal conditions A study in Drosophila melanogaster Oecologia 36 3 295 306 Bibcode 1978Oecol 36 295C doi 10 1007 BF00348055 PMID 28309916 S2CID 12465060 Rideout EJ Narsaiya MS Grewal SS December 2015 The Sex Determination Gene transformer Regulates Male Female Differences in Drosophila Body Size PLOS Genetics 11 12 e1005683 doi 10 1371 journal pgen 1005683 PMC 4692505 PMID 26710087 Gilbert SF 2000 Developmental Biology 6th ed Sunderland MA Sinauer Associates 2000 ISBN 978 0 87893 243 6 Lemaitre B Hoffmann J 2007 The host defense of Drosophila melanogaster PDF Annual Review of Immunology 25 697 743 doi 10 1146 annurev immunol 25 022106 141615 PMID 17201680 Troha K Im JH Revah J Lazzaro BP Buchon N February 2018 Comparative transcriptomics reveals CrebA as a novel regulator of infection tolerance in D melanogaster PLOS Pathogens 14 2 e1006847 doi 10 1371 journal ppat 1006847 PMC 5812652 PMID 29394281 De Gregorio E Spellman PT Tzou P Rubin GM Lemaitre B June 2002 The Toll and Imd pathways are the major regulators of the immune response in Drosophila The EMBO Journal 21 11 2568 79 doi 10 1093 emboj 21 11 2568 PMC 126042 PMID 12032070 Paredes JC Welchman DP Poidevin M Lemaitre B November 2011 Negative regulation by amidase PGRPs shapes the Drosophila antibacterial response and protects the fly from innocuous infection PDF Immunity 35 5 770 9 doi 10 1016 j immuni 2011 09 018 PMID 22118526 Troha K Buchon N September 2019 Methods for the study of innate immunity in Drosophila melanogaster Wiley Interdisciplinary Reviews Developmental Biology 8 5 e344 doi 10 1002 wdev 344 PMID 30993906 S2CID 119527642 Gilbert R Torres M Clemens R Hateley S Hosamani R Wade W Bhattacharya S February 2020 Drosophila melanogaster infection model npj Microgravity 6 1 4 doi 10 1038 s41526 019 0091 2 PMC 7000411 PMID 32047838 a b Dostalova A Rommelaere S Poidevin M Lemaitre B September 2017 Thioester containing proteins regulate the Toll pathway and play a role in Drosophila defence against microbial pathogens and parasitoid wasps BMC Biology 15 1 79 doi 10 1186 s12915 017 0408 0 PMC 5584532 PMID 28874153 Srinivasan N Gordon O Ahrens S Franz A Deddouche S Chakravarty P et al November 2016 Drosophila melanogaster eLife 5 doi 10 7554 eLife 19662 PMC 5138034 PMID 27871362 Goto A Yano T Terashima J Iwashita S Oshima Y Kurata S May 2010 Cooperative regulation of the induction of the novel antibacterial Listericin by peptidoglycan recognition protein LE and the JAK STAT pathway The Journal of Biological Chemistry 285 21 15731 8 doi 10 1074 jbc M109 082115 PMC 2871439 PMID 20348097 Wang L Kounatidis I Ligoxygakis P January 2014 Drosophila as a model to study the role of blood cells in inflammation innate immunity and cancer Frontiers in Cellular and Infection Microbiology 3 113 doi 10 3389 fcimb 2013 00113 PMC 3885817 PMID 24409421 Neyen C Bretscher AJ Binggeli O Lemaitre B June 2014 Methods to study Drosophila immunity PDF Methods 68 1 116 28 doi 10 1016 j ymeth 2014 02 023 PMID 24631888 Hashimoto Y Tabuchi Y Sakurai K Kutsuna M Kurokawa K Awasaki T et al December 2009 Identification of lipoteichoic acid as a ligand for draper in the phagocytosis of Staphylococcus aureus by Drosophila hemocytes Journal of Immunology 183 11 7451 60 doi 10 4049 jimmunol 0901032 PMID 19890048 Holz A Bossinger B Strasser T Janning W Klapper R October 2003 The two origins of hemocytes in Drosophila Development 130 20 4955 62 doi 10 1242 dev 00702 PMID 12930778 Sanchez Bosch P Makhijani K Herboso L Gold KS Baginsky R Woodcock KJ et al December 2019 Adult Drosophila Lack Hematopoiesis but Rely on a Blood Cell Reservoir at the Respiratory Epithelia to Relay Infection Signals to Surrounding Tissues Developmental Cell 51 6 787 803 e5 doi 10 1016 j devcel 2019 10 017 PMC 7263735 PMID 31735669 Parvy JP Yu Y Dostalova A Kondo S Kurjan A Bulet P et al July 2019 Drosophila eLife 8 e45061 doi 10 7554 eLife 45061 PMC 6667213 PMID 31358113 Sturtevant AH 1929 The claret mutant type of Drosophila simulans a study of chromosome elimination and cell lineage Zeitschrift fur Wissenschaftliche Zoologie 135 323 356 Nissani M May 1975 A new behavioral bioassay for an analysis of sexual attraction and pheromones in insects The Journal of Experimental Zoology 192 2 271 5 Bibcode 1975JEZ 192 271N doi 10 1002 jez 1401920217 PMID 805823 Khan FA 2011 Biotechnology Fundamentals CRC Press p 213 ISBN 978 1 4398 2009 4 The 2017 Nobel Prize in Physiology or Medicine jointly to Jeffrey C Hall Michael Rosbash and Michael W Young for their discoveries of molecular mechanisms controlling the circadian rhythm Nobelprize org October 2 2017 Retrieved October 5 2017 Lehnert BP Baker AE Gaudry Q Chiang AS Wilson RI January 2013 Distinct roles of TRP channels in auditory transduction and amplification in Drosophila Neuron 77 1 115 28 doi 10 1016 j neuron 2012 11 030 PMC 3811118 PMID 23312520 Zhang W Yan Z Jan LY Jan YN August 2013 Sound response mediated by the TRP channels NOMPC NANCHUNG and INACTIVE in chordotonal organs of Drosophila larvae Proceedings of the National Academy of Sciences of the United States of America 110 33 13612 7 Bibcode 2013PNAS 11013612Z doi 10 1073 pnas 1312477110 PMC 3746866 PMID 23898199 Homosexuality Turned On and Off in Fruit Flies Dasgupta S Sheehan TC Stevens CF Navlakha S December 2018 A neural data structure for novelty detection Proceedings of the National Academy of Sciences of the United States of America 115 51 13093 13098 Bibcode 2018PNAS 11513093D doi 10 1073 pnas 1814448115 PMC 6304992 PMID 30509984 Zwarts L Versteven M Callaerts P January 1 2012 Genetics and neurobiology of aggression in Drosophila Fly 6 1 35 48 doi 10 4161 fly 19249 PMC 3365836 PMID 22513455 Davis SM Thomas AL Liu L Campbell IM Dierick HA January 2018 Drosophila Using a Screen for Wing Damage Genetics 208 1 273 282 doi 10 1534 genetics 117 300292 PMC 5753862 PMID 29109180 a b c Versteven M Vanden Broeck L Geurten B Zwarts L Decraecker L Beelen M et al February 2017 Drosophila aggression Proceedings of the National Academy of Sciences of the United States of America 114 8 1958 1963 doi 10 1073 pnas 1605946114 PMC 5338383 PMID 28115690 a b Sengupta S Smith DP 2014 How Drosophila Detect Volatile Pheromones Signaling Circuits and Behavior In Mucignat Caretta C ed Neurobiology of Chemical Communication Frontiers in Neuroscience CRC Press Taylor amp Francis ISBN 978 1 4665 5341 5 PMID 24830032 Retrieved May 30 2019 Laturney M Billeter JC August 2016 Drosophila melanogaster females restore their attractiveness after mating by removing male anti aphrodisiac pheromones Nature Communications 7 1 12322 Bibcode 2016NatCo 712322L doi 10 1038 ncomms12322 PMC 4976142 PMID 27484362 Wang L Han X Mehren J Hiroi M Billeter JC Miyamoto T et al June 2011 Hierarchical chemosensory regulation of male male social interactions in Drosophila Nature Neuroscience 14 6 757 62 doi 10 1038 nn 2800 PMC 3102769 PMID 21516101 a b c Lim RS Eyjolfsdottir E Shin E Perona P Anderson DJ August 27 2014 How food controls aggression in Drosophila PLOS ONE 9 8 e105626 Bibcode 2014PLoSO 9j5626L doi 10 1371 journal pone 0105626 PMC 4146546 PMID 25162609 Erion R DiAngelo JR Crocker A Sehgal A September 2012 Interaction between sleep and metabolism in Drosophila with altered octopamine signaling The Journal of Biological Chemistry 287 39 32406 14 doi 10 1074 jbc M112 360875 PMC 3463357 PMID 22829591 a b c Kayser MS Mainwaring B Yue Z Sehgal A July 2015 Griffith LC ed Sleep deprivation suppresses aggression in Drosophila eLife 4 e07643 doi 10 7554 eLife 07643 PMC 4515473 PMID 26216041 Hardie RC Raghu P September 2001 Visual transduction in Drosophila Nature 413 6852 186 93 Bibcode 2001Natur 413 186H doi 10 1038 35093002 PMID 11557987 S2CID 4415605 a b c d Sharkey CR Blanco J Leibowitz MM Pinto Benito D Wardill TJ October 2020 The spectral sensitivity of Drosophila photoreceptors Scientific Reports 10 1 18242 Bibcode 2020NatSR 1018242S doi 10 1038 s41598 020 74742 1 PMC 7588446 PMID 33106518 S2CID 215551298 nbsp Material was copied and adapted from this source which is available under a Creative Commons Attribution 4 0 International License Feuda R Goulty M Zadra N Gasparetti T Rosato E Pisani D et al August 2021 Phylogenomics of Opsin Genes in Diptera Reveals Lineage Specific Events and Contrasting Evolutionary Dynamics in Anopheles and Drosophila Genome Biology and Evolution 13 8 evab170 doi 10 1093 gbe evab170 PMC 8369074 PMID 34270718 Harris WA Stark WS Walker JA April 1976 Genetic dissection of the photoreceptor system in the compound eye of Drosophila melanogaster The Journal of Physiology 256 2 415 439 doi 10 1113 jphysiol 1976 sp011331 PMC 1309314 PMID 16992509 S2CID 45888026 a b Ostroy SE Wilson M Pak WL August 1974 Drosophila rhodopsin photochemistry extraction and differences in the norp AP12 phototransduction mutant Biochemical and Biophysical Research Communications 59 3 960 966 doi 10 1016 s0006 291x 74 80073 2 PMID 4213042 Ostroy SE November 1978 Characteristics of Drosophila rhodopsin in wild type and norpA vision transduction mutants The Journal of General Physiology 72 5 717 732 doi 10 1085 jgp 72 5 717 PMC 2228556 PMID 105082 S2CID 5802525 a b c d e f g Salcedo E Huber A Henrich S Chadwell LV Chou WH Paulsen R Britt SG December 1999 Blue and green absorbing visual pigments of Drosophila ectopic expression and physiological characterization of the R8 photoreceptor cell specific Rh5 and Rh6 rhodopsins The Journal of Neuroscience 19 24 10716 10726 doi 10 1523 jneurosci 19 24 10716 1999 PMC 6784940 PMID 10594055 S2CID 17575850 Kirschfeld K Franceschini N Minke B September 1977 Evidence for a sensitising pigment in fly photoreceptors Nature 269 5627 386 390 Bibcode 1977Natur 269 386K doi 10 1038 269386a0 PMID 909585 S2CID 28890008 Minke B Kirschfeld K May 1979 The contribution of a sensitizing pigment to the photosensitivity spectra of fly rhodopsin and metarhodopsin The Journal of General Physiology 73 5 517 540 doi 10 1085 jgp 73 5 517 PMC 2215190 PMID 458418 S2CID 12451748 a b Kirschfeld K Feiler R Franceschini N 1978 A photostable pigment within the rhabdomere of fly photoreceptors no 7 Journal of Comparative Physiology A 125 3 275 284 doi 10 1007 BF00656606 S2CID 40233531 Kirschfeld K Franceschini N June 1977 Photostable pigments within the membrane of photoreceptors and their possible role Biophysics of Structure and Mechanism 3 2 191 194 doi 10 1007 BF00535818 PMID 890056 S2CID 5846094 Zuker CS Montell C Jones K Laverty T Rubin GM May 1987 A rhodopsin gene expressed in photoreceptor cell R7 of the Drosophila eye homologies with other signal transducing molecules The Journal of Neuroscience 7 5 1550 1557 doi 10 1523 jneurosci 07 05 01550 1987 PMC 6568820 PMID 2437266 S2CID 1490332 a b c Chou WH Hall KJ Wilson DB Wideman CL Townson SM Chadwell LV Britt SG December 1996 Identification of a novel Drosophila opsin reveals specific patterning of the R7 and R8 photoreceptor cells Neuron 17 6 1101 1115 doi 10 1016 s0896 6273 00 80243 3 PMID 8982159 S2CID 18294965 a b c d Feiler R Bjornson R Kirschfeld K Mismer D Rubin GM Smith DP et al October 1992 Ectopic expression of ultraviolet rhodopsins in the blue photoreceptor cells of Drosophila visual physiology and photochemistry of transgenic animals The Journal of Neuroscience 12 10 3862 3868 doi 10 1523 jneurosci 12 10 03862 1992 PMC 6575971 PMID 1403087 Montell C Jones K Zuker C Rubin G May 1987 A second opsin gene expressed in the ultraviolet sensitive R7 photoreceptor cells of Drosophila melanogaster The Journal of Neuroscience 7 5 1558 1566 doi 10 1523 JNEUROSCI 07 05 01558 1987 PMC 6568825 PMID 2952772 S2CID 17003459 Huber A Schulz S Bentrop J Groell C Wolfrum U Paulsen R April 1997 Molecular cloning of Drosophila Rh6 rhodopsin the visual pigment of a subset of R8 photoreceptor cells FEBS Letters 406 1 2 6 10 doi 10 1016 s0014 5793 97 00210 x PMID 9109375 S2CID 18368117 Pollock JA Benzer S June 1988 Transcript localization of four opsin genes in the three visual organs of Drosophila RH2 is ocellus specific Nature 333 6175 779 782 Bibcode 1988Natur 333 779P doi 10 1038 333779a0 PMID 2968518 S2CID 4303934 a b Feiler R Harris WA Kirschfeld K Wehrhahn C Zuker CS June 1988 Targeted misexpression of a Drosophila opsin gene leads to altered visual function Nature 333 6175 737 741 Bibcode 1988Natur 333 737F doi 10 1038 333737a0 PMID 2455230 S2CID 4248264 Sakai K Tsutsui K Yamashita T Iwabe N Takahashi K Wada A Shichida Y August 2017 Drosophila melanogaster rhodopsin Rh7 is a UV to visible light sensor with an extraordinarily broad absorption spectrum Scientific Reports 7 1 7349 Bibcode 2017NatSR 7 7349S doi 10 1038 s41598 017 07461 9 PMC 5544684 PMID 28779161 S2CID 3276084 Ni JD Baik LS Holmes TC Montell C May 2017 A rhodopsin in the brain functions in circadian photoentrainment in Drosophila Nature 545 7654 340 344 Bibcode 2017Natur 545 340N doi 10 1038 nature22325 PMC 5476302 PMID 28489826 S2CID 4468254 Vogt K February 1 1984 The Chromophore of the Visual Pigment in Some Insect Orders Zeitschrift fur Naturforschung C 39 1 2 196 197 doi 10 1515 znc 1984 1 236 S2CID 88980658 Vogt K Kirschfeld K April 1984 Chemical identity of the chromophores of fly visual pigment Naturwissenschaften 71 4 211 213 Bibcode 1984NW 71 211V doi 10 1007 BF00490436 S2CID 24205801 Guhmann M Porter ML Bok MJ August 2022 The Gluopsins Opsins without the Retinal Binding Lysine Cells 11 15 2441 doi 10 3390 cells11152441 PMC 9368030 PMID 35954284 Katana R Guan C Zanini D Larsen ME Giraldo D Geurten BR et al September 2019 Chromophore Independent Roles of Opsin Apoproteins in Drosophila Mechanoreceptors Current Biology 29 17 2961 2969 e4 doi 10 1016 j cub 2019 07 036 PMID 31447373 S2CID 201420079 a b Leung NY Thakur DP Gurav AS Kim SH Di Pizio A Niv MY Montell C April 2020 Functions of Opsins in Drosophila Taste Current Biology 30 8 1367 1379 e6 doi 10 1016 j cub 2020 01 068 PMC 7252503 PMID 32243853 Kumbalasiri T Rollag MD Isoldi MC Castrucci AM Provencio I March 2007 Melanopsin triggers the release of internal calcium stores in response to light Photochemistry and Photobiology 83 2 273 279 doi 10 1562 2006 07 11 RA 964 PMID 16961436 S2CID 23060331 a b Raghu P Colley NJ Webel R James T Hasan G Danin M et al May 2000 Normal phototransduction in Drosophila photoreceptors lacking an InsP 3 receptor gene Molecular and Cellular Neurosciences 15 5 429 45 doi 10 1006 mcne 2000 0846 PMID 10833300 S2CID 23861204 Hardie RC Juusola M October 2015 Phototransduction in Drosophila Current Opinion in Neurobiology 34 37 45 doi 10 1016 j conb 2015 01 008 PMID 25638280 S2CID 140206989 Wang T Xu H Oberwinkler J Gu Y Hardie RC Montell C February 2005 Light activation adaptation and cell survival functions of the Na Ca2 exchanger CalX Neuron 45 3 367 78 doi 10 1016 j neuron 2004 12 046 PMID 15694324 Rein K Zockler M Mader MT Grubel C Heisenberg M February 2002 The Drosophila standard brain Current Biology 12 3 227 31 doi 10 1016 S0960 9822 02 00656 5 PMID 11839276 S2CID 15785406 Dawkins R Dawkins M 1976 Hierarchical organization and postural facilitation rules for grooming in flies Animal Behaviour 24 4 739 755 doi 10 1016 S0003 3472 76 80003 6 S2CID 53186674 a b c Davis WJ 1979 Behavioural hierarchies Trends in Neurosciences 2 2 5 7 doi 10 1016 0166 2236 79 90003 1 S2CID 53180462 a b Seeds AM Ravbar P Chung P Hampel S Midgley FM Mensh BD Simpson JH August 2014 A suppression hierarchy among competing motor programs drives sequential grooming in Drosophila eLife 3 e02951 doi 10 7554 eLife 02951 PMC 4136539 PMID 25139955 Mathis A Mamidanna P Cury KM Abe T Murthy VN Mathis MW Bethge M September 2018 DeepLabCut markerless pose estimation of user defined body parts with deep learning Nature Neuroscience 21 9 1281 1289 doi 10 1038 s41593 018 0209 y PMID 30127430 S2CID 4748395 Strauss R Heisenberg M August 1990 Coordination of legs during straight walking and turning in Drosophila melanogaster Journal of Comparative Physiology A 167 3 403 12 doi 10 1007 BF00192575 PMID 2121965 S2CID 12965869 DeAngelis BD Zavatone Veth JA Clark DA June 2019 Drosophila eLife 8 doi 10 7554 eLife 46409 PMC 6598772 PMID 31250807 Wosnitza A Bockemuhl T Dubbert M Scholz H Buschges A February 2013 Inter leg coordination in the control of walking speed in Drosophila The Journal of Experimental Biology 216 Pt 3 480 91 doi 10 1242 jeb 078139 PMID 23038731 Mendes CS Bartos I Akay T Marka S Mann RS January 2013 Quantification of gait parameters in freely walking wild type and sensory deprived Drosophila melanogaster eLife 2 e00231 doi 10 7554 eLife 00231 PMC 3545443 PMID 23326642 Szczecinski NS Bockemuhl T Chockley AS Buschges A November 2018 Drosophila The Journal of Experimental Biology 221 Pt 22 jeb189142 doi 10 1242 jeb 189142 PMID 30274987 Hooper SL May 2012 Body size and the neural control of movement Current Biology 22 9 R318 22 doi 10 1016 j cub 2012 02 048 PMID 22575473 a b c Fry SN Sayaman R Dickinson MH April 2003 The aerodynamics of free flight maneuvers in Drosophila PDF Science 300 5618 495 8 Bibcode 2003Sci 300 495F doi 10 1126 science 1081944 PMID 12702878 S2CID 40952385 Archived from the original PDF on September 24 2015 Hesselberg T Lehmann FO December 2007 Turning behaviour depends on frictional damping in the fruit fly Drosophila The Journal of Experimental Biology 210 Pt 24 4319 34 doi 10 1242 jeb 010389 PMID 18055621 Chiang AS Lin CY Chuang CC Chang HM Hsieh CH Yeh CW et al January 2011 Three dimensional reconstruction of brain wide wiring networks in Drosophila at single cell resolution Current Biology 21 1 1 11 doi 10 1016 j cub 2010 11 056 PMID 21129968 S2CID 17155338 FlyCircuit A Database of Drosophila Brain Neurons Retrieved August 30 2013 Meinertzhagen IA O Neil SD March 1991 Synaptic organization of columnar elements in the lamina of the wild type in Drosophila melanogaster The Journal of Comparative Neurology 305 2 232 63 doi 10 1002 cne 903050206 PMID 1902848 S2CID 35301798 Rivera Alba M Vitaladevuni SN Mishchenko Y Mischenko Y Lu Z Takemura SY et al December 2011 Wiring economy and volume exclusion determine neuronal placement in the Drosophila brain Current Biology 21 23 2000 5 doi 10 1016 j cub 2011 10 022 PMC 3244492 PMID 22119527 Takemura SY Bharioke A Lu Z Nern A Vitaladevuni S Rivlin PK et al August 2013 A visual motion detection circuit suggested by Drosophila connectomics Nature 500 7461 175 81 Bibcode 2013Natur 500 175T doi 10 1038 nature12450 PMC 3799980 PMID 23925240 Takemura SY Aso Y Hige T Wong A Lu Z Xu CS et al July 2017 Drosophila brain eLife 6 e26975 doi 10 7554 eLife 26975 PMC 5550281 PMID 28718765 Entire Fruit Fly Brain Imaged with Electron Microscopy The Scientist Magazine Retrieved July 15 2018 Zheng Z Lauritzen JS Perlman E Robinson CG Nichols M Milkie D et al July 2018 A Complete Electron Microscopy Volume of the Brain of Adult Drosophila melanogaster Cell 174 3 730 743 e22 bioRxiv 10 1101 140905 doi 10 1016 j cell 2018 06 019 PMC 6063995 PMID 30033368 Xu CS Januszewski M Lu Z Takemura SY Hayworth K Huang G Shinomiya K Maitin Shepard J Ackerman D Berg S Blakely T et al 2020 A connectome of the adult Drosophila central brain bioRxiv Cold Spring Harbor Laboratory 2020 01 21 911859 doi 10 1101 2020 01 21 911859 S2CID 213140797 Analysis tools for connectomics HHMI Phelps Jasper S Hildebrand David Grant Colburn Graham Brett J Kuan Aaron T Thomas Logan A Nguyen Tri M Buhmann Julia Azevedo Anthony W Sustar Anne Agrawal Sweta Liu Mingguan Shanny Brendan L Funke Jan Tuthill John C Lee Wei Chung Allen February 2 2021 Reconstruction of motor control circuits in adult Drosophila using automated transmission electron microscopy Cell 184 3 759 774 e18 doi 10 1016 j cell 2020 12 013 PMC 8312698 PMID 33400916 Scheffer LK Xu CS Januszewski M Lu Z Takemura SY Hayworth KJ Huang G Shinomiya K Maitlin Shepard J Berg S Clements J et al 2020 A Connectome and Analysis of the Adult Drosophila Central Brain bioRxiv Cold Spring Harbor 9 doi 10 1101 2020 04 07 030213 PMC 7546738 PMID 32880371 S2CID 215790785 Popovych Sergiy Macrina Thomas Kemnitz Nico Castro Manuel Nehoran Barak Jia Zhen Bae J Alexander Mitchell Eric Mu Shang Trautman Eric T Saalfeld Stephan Li Kai Seung Sebastian March 27 2022 Petascale pipeline for precise alignment of images from serial section electron microscopy bioRxiv 10 1101 2022 03 25 485816 Winding Michael Pedigo Benjamin D Barnes Christopher L Patsolic Heather G Park Youngser Kazimiers Tom Fushiki Akira Andrade Ingrid V Khandelwal Avinash Valdes Aleman Javier Li Feng Randel Nadine Barsotti Elizabeth Correia Ana Fetter Richard D March 10 2023 The connectome of an insect brain Science 379 6636 eadd9330 doi 10 1126 science add9330 PMC 7614541 PMID 36893230 Non pest species Plant Health Australia Retrieved September 19 2017 McEvey S February 5 2014 Fruit Flies A Case Of Mistaken Identity Australian Museum Retrieved September 19 2017 Further reading editKohler RE 1994 Lords of the Fly Drosophila geneticsand the experimental life Chicago University of Chicago Press ISBN 978 0 226 45063 6 Gilbert SF 2000 Developmental Biology 6th ed Sunderland MA Sinauer Associates 2000 ISBN 978 0 87893 243 6 Perrimon N Bonini NM Dhillon P March 2016 Fruit flies on the front line the translational impact of Drosophila Disease Models amp Mechanisms 9 3 229 31 doi 10 1242 dmm 024810 PMC 4833334 PMID 26935101 Henderson M April 8 2010 Row over fruit fly Drosophila melanogaster name bugs scientists The Times The Australian Retrieved September 19 2017 External links edit nbsp Wikispecies has information related to Drosophila melanogaster nbsp Wikimedia Commons has media related to Drosophila melanogaster nbsp Scholia has a topic profile for Drosophila melanogaster A quick and simple introduction to Drosophila melanogaster Drosophila Virtual Library Drosophila Genomics Resource Center collects maintains and distributes Drosophila DNA clones and cell lines Bloomington Drosophila Stock Center collects maintains and distributes Drosophila melanogaster strains for research FlyBase A Database of Drosophila Genes amp Genomes NCBI Map Viewer Drosophila melanogaster Drosophila Virtual Library The Berkeley Drosophila Genome Project FlyMove video resources for Drosophila development Drosophila Nomenclature naming of genes Archived from the original on October 8 2011 View the Fruitfly genome on Ensembl View the dm6 genome assembly in the UCSC Genome Browser Manchester Fly Facility for the public Archived 2015 05 13 at the Wayback Machine from the University of Manchester The droso4schools website with school relevant resources about Drosophila Part 1 of the Small fly BIG impact educational videos explaining the history and importance of the model organism Drosophila Part 2 of the Small fly BIG impact educational videos explaining how research is carried out in Drosophila Inside the Fly Lab broadcast by WGBH and PBS in the program series Curious January 2008 How a Fly Detects Poison Archived 2013 01 13 at archive today WhyFiles org article describes how the fruit fly tastes a larva killing chemical in food Retrieved from https en wikipedia org w index php title Drosophila melanogaster amp oldid 1205453851, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.