Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gen Comp Endocrinol ; 353: 114521, 2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38621462

RESUMEN

Myoinhibitory peptides (MIPs) affect various physiological functions, including juvenile hormone signaling, muscle contraction, larval development, and reproduction in invertebrates. Although MIPs are ligands for MIP and/or sex peptide receptors (MIP/SPRs) in diverse arthropods and model organisms belonging to Lophotrochozoa, the MIP signaling system has not yet been fully investigated in mollusks. In this study, we identified the MIP signaling system in the Pacific abalone Haliotis discus hannai (Hdh). Similar to the invertebrate MIPs, a total of eight paracopies of MIPs (named Hdh-MIP1 to Hdh-MIP8), harboring a WX5-7Wamide motif, except for Hdh-MIP2, were found in the Hdh-MIP precursor. Furthermore, we characterized a functional Hdh-MIPR, which responded to the Hdh-MIPs, except for Hdh-MIP2, possibly linked with the PKC/Ca2+ and PKA/cAMP signaling pathways. Hdh-MIPs delayed larval metamorphosis but increased the spawning behavior. These results suggest that the Hdh-MIP signaling system provides insights into the unique function of MIP in invertebrates.


Asunto(s)
Gastrópodos , Larva , Metamorfosis Biológica , Transducción de Señal , Animales , Metamorfosis Biológica/fisiología , Larva/crecimiento & desarrollo , Larva/metabolismo , Transducción de Señal/fisiología , Gastrópodos/crecimiento & desarrollo , Gastrópodos/metabolismo , Gastrópodos/fisiología , Péptidos , Reproducción/fisiología
2.
Genetics ; 227(1)2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38551457

RESUMEN

Across diverse insect taxa, the behavior and physiology of females dramatically changes after mating-processes largely triggered by the transfer of seminal proteins from their mates. In the vinegar fly Drosophila melanogaster, the seminal protein sex peptide (SP) decreases the likelihood of female flies remating and causes additional behavioral and physiological changes that promote fertility including increasing egg production. Although SP is only found in the Drosophila genus, its receptor, sex peptide receptor (SPR), is the widely conserved myoinhibitory peptide (MIP) receptor. To test the functional role of SPR in mediating postmating responses in a non-Drosophila dipteran, we generated 2 independent Spr-knockout alleles in the yellow fever mosquito, Aedes aegypti. Although SPR is needed for postmating responses in Drosophila and the cotton bollworm Helicoverpa armigera, Spr mutant Ae. aegypti show completely normal postmating decreases in remating propensity and increases in egg laying. In addition, injection of synthetic SP or accessory gland homogenate from D. melanogaster into virgin female mosquitoes did not elicit these postmating responses. Our results demonstrate that Spr is not required for these canonical postmating responses in Ae. aegypti, indicating that other, as yet unknown, signaling pathways are likely responsible for these behavioral switches in this disease vector.


Asunto(s)
Aedes , Proteínas de Insectos , Oviposición , Receptores de Péptidos de Invertebrados , Animales , Femenino , Masculino , Aedes/genética , Aedes/fisiología , Drosophila melanogaster/fisiología , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Receptores de Péptidos de Invertebrados/metabolismo , Receptores de Péptidos de Invertebrados/genética , Conducta Sexual Animal
3.
Sci Rep ; 14(1): 6411, 2024 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-38494531

RESUMEN

Physiological status can influence social behavior, which in turn can affect physiology and health. Previously, we reported that tumor growth in Drosophila virgin females depends on the social context, but did not investigate the underlying physiological mechanisms. Here, we sought to characterize the signal perceived between tumorous flies, ultimately discovering that the tumor suppressive effect varies depending on reproductive status. Firstly, we show that the tumor suppressive effect is neither dependent on remnant pheromone-like products nor on the microbiota. Transcriptome analysis of the heads of these tumorous flies reveals social-dependent gene-expression changes related to nervous-system activity, suggesting that a cognitive-like relay might mediate the tumor suppressive effect. The transcriptome also reveals changes in the expression of genes related to mating behavior. Surprisingly, we observed that this social-dependent tumor-suppressive effect is lost in fertilized females. After mating, Drosophila females change their behavior-favoring offspring survival-in response to peptides transferred via the male ejaculate, a phenomenon called "male manipulation". Remarkably, the social-dependent tumor suppressive effect is restored in females mated by sex-peptide deficient males. Since male manipulation has likely been selected to favor male gene transmission, our findings indicate that this evolutionary trait impedes social-dependent tumor growth slowdown.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Femenino , Masculino , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Conducta Sexual Animal/fisiología , Péptidos/metabolismo , Reproducción
4.
Mol Biol Evol ; 41(4)2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38518286

RESUMEN

Post-mating responses play a vital role in successful reproduction across diverse species. In fruit flies, sex peptide binds to the sex peptide receptor, triggering a series of post-mating responses. However, the origin of sex peptide receptor predates the emergence of sex peptide. The evolutionary origins of the interactions between sex peptide and sex peptide receptor and the mechanisms by which they interact remain enigmatic. In this study, we used ancestral sequence reconstruction, AlphaFold2 predictions, and molecular dynamics simulations to study sex peptide-sex peptide receptor interactions and their origination. Using AlphaFold2 and long-time molecular dynamics simulations, we predicted the structure and dynamics of sex peptide-sex peptide receptor interactions. We show that sex peptide potentially binds to the ancestral states of Diptera sex peptide receptor. Notably, we found that only a few amino acid changes in sex peptide receptor are sufficient for the formation of sex peptide-sex peptide receptor interactions. Ancestral sequence reconstruction and molecular dynamics simulations further reveal that sex peptide receptor interacts with sex peptide through residues that are mostly involved in the interaction interface of an ancestral ligand, myoinhibitory peptides. We propose a potential mechanism whereby sex peptide-sex peptide receptor interactions arise from the preexisting myoinhibitory peptides-sex peptide receptor interface as well as early chance events both inside and outside the preexisting interface that created novel sex peptide-specific sex peptide-sex peptide receptor interactions. Our findings provide new insights into the origin and evolution of sex peptide-sex peptide receptor interactions and their relationship with myoinhibitory peptides-sex peptide receptor interactions.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Péptidos/química , Drosophila/metabolismo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo
5.
Proc Natl Acad Sci U S A ; 121(10): e2310841121, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38412134

RESUMEN

Connectomics research has made it more feasible to explore how neural circuits can generate multiple outputs. Female sexual drive provides a good model for understanding reversible, long-term functional changes in motivational circuits. After emerging, female flies avoid male courtship, but they become sexually receptive over 2 d. Mating causes females to reject further mating for several days. Here, we report that pC1 neurons, which process male courtship and regulate copulation behavior, exhibit increased CREB (cAMP response element binding protein) activity during sexual maturation and decreased CREB activity after mating. This increased CREB activity requires the neuropeptide Dh44 (Diuretic hormone 44) and its receptors. A subset of the pC1 neurons secretes Dh44, which stimulates CREB activity and increases expression of the TRP channel Pyrexia (Pyx) in more pC1 neurons. This, in turn, increases pC1 excitability and sexual drive. Mating suppresses pyx expression and pC1 excitability. Dh44 is orthologous to the conserved corticotrophin-releasing hormone family, suggesting similar roles in other species.


Asunto(s)
Proteínas de Drosophila , Neuropéptidos , Animales , Masculino , Femenino , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Neuropéptidos/metabolismo , Copulación/fisiología , Cortejo , Hormonas , Conducta Sexual Animal/fisiología
6.
Proc Natl Acad Sci U S A ; 120(5): e2214883120, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36706221

RESUMEN

Sex peptide (SP), a seminal fluid protein of Drosophila melanogaster males, has been described as driving a virgin-to-mated switch in females, through eliciting an array of responses including increased egg laying, activity, and food intake and a decreased remating rate. While it is known that SP achieves this, at least in part, by altering neuronal signaling in females, the genetic architecture and temporal dynamics of the female's response to SP remain elusive. We used a high-resolution time series RNA-sequencing dataset of female heads at 10 time points within the first 24 h after mating to learn about the genetic architecture, at the gene and exon levels, of the female's response to SP. We find that SP is not essential to trigger early aspects of a virgin-to-mated transcriptional switch, which includes changes in a metabolic gene regulatory network. However, SP is needed to maintain and diversify metabolic changes and to trigger changes in a neuronal gene regulatory network. We further find that SP alters rhythmic gene expression in females and suggests that SP's disruption of the female's circadian rhythm might be key to its widespread effects.


Asunto(s)
Relojes Circadianos , Proteínas de Drosophila , Animales , Masculino , Femenino , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Espermatozoides/metabolismo , Relojes Circadianos/genética , Factores de Tiempo , Péptidos/metabolismo , Perfilación de la Expresión Génica , Conducta Sexual Animal/fisiología
7.
BMC Biol ; 20(1): 279, 2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36514080

RESUMEN

BACKGROUND: Male-derived seminal fluid proteins (SFPs) that enter female fruitflies during mating induce a myriad of physiological and behavioral changes, optimizing fertility of the mating pair. Some post-mating changes in female Drosophila melanogaster persist for ~10-14 days. Their long-term persistence is because the seminal protein that induces these particular changes, the Sex Peptide (SP), is retained long term in females by binding to sperm, with gradual release of its active domain from sperm. Several other "long-term response SFPs" (LTR-SFPs) "prime" the binding of SP to sperm. Whether female factors play a role in this process is unknown, though it is important to study both sexes for a comprehensive physiological understanding of SFP/sperm interactions and for consideration in models of sexual conflict. RESULTS: We report here that sperm in male ejaculates bind SP more weakly than sperm that have entered females. Moreover, we show that the amount of SP, and other SFPs, bound to sperm increases with time and transit of individual seminal proteins within the female reproductive tract (FRT). Thus, female contributions are needed for maximal and appropriate binding of SP, and other SFPs, to sperm. Towards understanding the source of female molecular contributions, we ablated spermathecal secretory cells (SSCs) and/or parovaria (female accessory glands), which contribute secretory proteins to the FRT. We found no dramatic change in the initial levels of SP bound to sperm stored in mated females with ablated or defective SSCs and/or parovaria, indicating that female molecules that facilitate the binding of SP to sperm are not uniquely derived from SSCs and parovaria. However, we observed higher levels of SP (and sperm) retention long term in females whose SSCs and parovaria had been ablated, indicating secretions from these female tissues are necessary for the gradual release of Sex Peptide's active region from stored sperm. CONCLUSION: This study reveals that the SP-sperm binding pathway is not entirely male-derived and that female contributions are needed to regulate the levels of SP associated with sperm stored in their storage sites.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Masculino , Femenino , Drosophila melanogaster/fisiología , Proteínas de Drosophila/metabolismo , Semen/metabolismo , Espermatozoides/fisiología , Conducta Sexual Animal/fisiología , Péptidos/metabolismo
8.
Insects ; 13(7)2022 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-35886799

RESUMEN

Mating initiates broad physiological changes encompassing multiple organ systems in females. Elucidating the complex inter- and intra-organ signaling events that coordinate these physiological changes is an important goal in the field of reproductive biology. Further characterization of these complex molecular and physiological interactions is key to understanding how females meet the energetic demands of offspring production. Many recent studies of the fruit fly, Drosophila melanogaster, have described the mechanisms of post-mating changes within the female reproductive tract and digestive system. Additionally, other studies have described post-mating signaling crosstalk between these systems. Interestingly, male seminal fluid proteins have been linked to post-mating responses within the female reproductive tract and gut, and to signaling events between the two organ systems. However, information about the hormonal and neuronal signaling pathways underlying the post-mating signaling events within and between the reproductive tract and digestive systems that are triggered by seminal fluid proteins has yet to be combined into a single view. In this article, we summarize and integrate these studies into a single "network schematic" of the known signaling events within and between the reproductive and digestive systems downstream of male seminal fluid proteins. This synthesis also draws attention to the incomplete parts of these pathways, so that outstanding questions may be addressed in future studies.

9.
J Insect Physiol ; 140: 104414, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35728669

RESUMEN

In many species, female reproductive investment comes at a cost to immunity and resistance to infection. Mated Drosophila melanogaster females are more susceptible to bacterial infection than unmated females. Transfer of the male seminal fluid protein Sex Peptide reduces female post-mating immune defense. Sex Peptide is known to cause both short- and long-term changes to female physiology and behavior. While previous studies showed that females were less resistant to bacterial infection as soon as 2.5 h and as long as 26.5 h after mating, it is unknown whether this is a binary switch from mated to unmated state or whether females can recover to unmated levels of immunity. It is additionally unknown whether repeated mating causes progressive reduction in defense capacity. We compared the immune defense of mated females when infected at 2, 4, 7, or 10 days after mating to that of unmated females and saw no recovery of immune capacity regardless of the length of time between mating and infection. Because D. melanogaster females can mate multiply, we additionally tested whether a second mating, and therefore a second transfer of seminal fluids, caused deeper reduction in immune performance. We found that females mated either once or twice before infection survived at equal proportions, both with significantly lower probability than unmated females. We conclude that a single mating event is sufficient to persistently suppress the female immune system. Interestingly, we observed that induced levels of expression of genes encoding antimicrobial peptides (AMPs) decreased with age in both experiments, partially obscuring the effects of mating. Collectively, the data indicate that being reproductively active versus reproductively inactive are alternative binary states with respect to female D. melanogaster immunity. The establishment of a suppressed immune status in reproductively active females can inform our understanding of the regulation of immune defense and the mechanisms of physiological trade-offs.


Asunto(s)
Drosophila melanogaster , Reproducción , Animales , Drosophila melanogaster/fisiología , Femenino , Masculino , Péptidos/metabolismo , Reproducción/fisiología , Conducta Sexual Animal/fisiología
10.
Biol Rev Camb Philos Soc ; 97(4): 1426-1448, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35249265

RESUMEN

A central paradigm in evolutionary biology is that the fundamental divergence in the fitness interests of the sexes ('sexual conflict') can lead to both the evolution of sex-specific traits that reduce fitness for individuals of the opposite sex, and sexually antagonistic coevolution between the sexes. However, clear examples of traits that evolved in this way - where a single trait in one sex demonstrably depresses the fitness of members of the opposite sex, resulting in antagonistic coevolution - are rare. The Drosophila seminal protein 'sex peptide' (SP) is perhaps the most widely cited example of a trait that appears to harm females while benefitting males. Transferred in the ejaculate by males during mating, SP triggers profound and wide-ranging changes in female behaviour and physiology. Early studies reported that the transfer of SP enhances male fitness while depressing female fitness, providing the foundations for the widespread view that SP has evolved to manipulate females for male benefit. Here, we argue that this view is (i) a simplification of a wider body of contradictory empirical research, (ii) narrow with respect to theory describing the origin and maintenance of sexually selected traits, and (iii) hard to reconcile with what we know of the evolutionary history of SP's effects on females. We begin by charting the history of thought regarding SP, both at proximate (its production, function, and mechanism of action) and ultimate (its fitness consequences and evolutionary history) levels, reviewing how studies of SP were central to the development of the field of sexual conflict. We describe a prevailing paradigm for SP's evolution: that SP originated and continues to evolve to manipulate females for male benefit. In contrast to this view, we argue on three grounds that the weight of evidence does not support the view that receipt of SP decreases female fitness: (i) results from studies of SP's impact on female fitness are mixed and more often neutral or positive, with fitness costs emerging only under nutritional extremes; (ii) whether costs from SP are appreciable in wild-living populations remains untested; and (iii) recently described confounds in genetic manipulations of SP raise the possibility that measures of the costs and benefits of SP have been distorted. Beyond SP's fitness effects, comparative and genetic data are also difficult to square with the idea that females suffer fitness costs from SP. Instead, these data - from functional and evolutionary genetics and the neural circuitry of female responses to SP - suggest an evolutionary history involving the evolution of a dedicated SP-sensing apparatus in the female reproductive tract that is likely to have evolved because it benefits females, rather than harms them. We end by exploring theory and evidence that SP benefits females by functioning as a signal of male quality or of sperm receipt and storage (or both). The expanded view of the evolution of SP that we outline recognises the context-dependent and fluctuating roles played by both cooperative and antagonistic selection in the origin and maintenance of reproductive traits.


Asunto(s)
Semen , Conducta Sexual Animal , Animales , Evolución Biológica , Femenino , Masculino , Péptidos/genética , Péptidos/metabolismo , Reproducción/fisiología , Conducta Sexual Animal/fisiología , Espermatozoides
11.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33495334

RESUMEN

Seminal fluid plays an essential role in promoting male reproductive success and modulating female physiology and behavior. In the fruit fly, Drosophila melanogaster, Sex Peptide (SP) is the best-characterized protein mediator of these effects. It is secreted from the paired male accessory glands (AGs), which, like the mammalian prostate and seminal vesicles, generate most of the seminal fluid contents. After mating, SP binds to spermatozoa and is retained in the female sperm storage organs. It is gradually released by proteolytic cleavage and induces several long-term postmating responses, including increased ovulation, elevated feeding, and reduced receptivity to remating, primarily signaling through the SP receptor (SPR). Here, we demonstrate a previously unsuspected SPR-independent function for SP. We show that, in the AG lumen, SP and secreted proteins with membrane-binding anchors are carried on abundant, large neutral lipid-containing microcarriers, also found in other SP-expressing Drosophila species. These microcarriers are transferred to females during mating where they rapidly disassemble. Remarkably, SP is a key microcarrier assembly and disassembly factor. Its absence leads to major changes in the seminal proteome transferred to females upon mating. Males expressing nonfunctional SP mutant proteins that affect SP's binding to and release from sperm in females also do not produce normal microcarriers, suggesting that this male-specific defect contributes to the resulting widespread abnormalities in ejaculate function. Our data therefore reveal a role for SP in formation of seminal macromolecular assemblies, which may explain the presence of SP in Drosophila species that lack the signaling functions seen in Dmelanogaster.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lípidos/química , Microesferas , Semen/química , Animales , Proteínas de Drosophila/genética , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Mutación/genética , Proteoma/metabolismo , Conducta Sexual Animal , Especificidad de la Especie
12.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443193

RESUMEN

Drosophila melanogaster females experience a large shift in energy homeostasis after mating to compensate for nutrient investment in egg production. To cope with this change in metabolism, mated females undergo widespread physiological and behavioral changes, including increased food intake and altered digestive processes. The mechanisms by which the female digestive system responds to mating remain poorly characterized. Here, we demonstrate that the seminal fluid protein Sex Peptide (SP) is a key modulator of female post-mating midgut growth and gene expression. SP is both necessary and sufficient to trigger post-mating midgut growth in females under normal nutrient conditions, and likely acting via its receptor, Sex Peptide Receptor (SPR). Moreover, SP is responsible for almost the totality of midgut transcriptomic changes following mating, including up-regulation of protein and lipid metabolism genes and down-regulation of carbohydrate metabolism genes. These changes in metabolism may help supply the female with the nutrients required to sustain egg production. Thus, we report a role for SP in altering female physiology to enhance reproductive output: Namely, SP triggers the switch from virgin to mated midgut state.


Asunto(s)
Sistema Digestivo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fertilidad/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores de Péptidos/metabolismo , Reproducción/fisiología , Transcriptoma/genética , Animales , Copulación , Sistema Digestivo/anatomía & histología , Sistema Digestivo/crecimiento & desarrollo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Fertilidad/genética , Ontología de Genes , Intestinos/anatomía & histología , Intestinos/crecimiento & desarrollo , Intestinos/fisiología , Masculino , Receptores de Péptidos/genética , Reproducción/genética , Semen/metabolismo , Conducta Sexual Animal/fisiología , Transcriptoma/fisiología
13.
Evolution ; 74(11): 2483-2493, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32944943

RESUMEN

Females of many animal species mate several times with different males (polyandry), whereas females of some species mate with a single male (monandry) only once. Little is known about the mechanisms by which these different mating systems evolve. Females of Drosophila prolongata mate serially, unlike Drosophila melanogaster females that refuse to remate for several days after their first mating (remating suppression [RS]). Nevertheless, interestingly, nonvirgin D. prolongata females refuse to remate with males that are prohibited from performing their species-specific courtship behavior, leg vibration (LV), suggesting that LV overrides RS making it cryptic in D. prolongata. In this study, we examined how long this cryptic RS persists. Surprisingly, it was sustained for at least 2 weeks, showing that RS is substantially augmented in D. prolongata compared to that of D. melanogaster. The two most closely related species to D. prolongata, Drosophila rhopaloa and Drosophila carrolli, do not perform LV and showed augmented RS, supporting the idea that augmented RS could have evolved before LV was acquired. These results suggested that D. prolongata females are intrinsically monandrous, whereas the newly evolved courtship behavior makes them polyandrous. This is a rare case in which a proximate mechanism of polyandry evolution from monandry is demonstrated.


Asunto(s)
Evolución Biológica , Drosophila/genética , Conducta Sexual Animal , Animales , Femenino , Masculino
14.
Insect Biochem Mol Biol ; 127: 103472, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32971207

RESUMEN

Drosophila melanogaster sex peptide receptor (DrmSPR) is a G protein-coupled receptor (GPCR) with 'dual ligand selectivity' towards sex peptide (SP) and myoinhibitory peptides (MIPs), which are only remotely related to one another. SPR is conserved in almost all the sequenced lophotrochozoan and ecdysozoan genomes. SPRs from non-drosophilid taxa, such as those from the mosquitoes Aedes aegypti (AeaSPR), Anopheles gambiae (AngSPR), and the sea slug Aplysia californica (ApcSPR), are highly sensitive to MIP, but not to SP. To understand how Drosophila SPRs evolved their SP sensitivity while maintaining MIP sensitivity, we examined ligand selectivity in a series of chimeric GPCRs that combine domains from the SP-sensitive DrmSPR and the SP-insensitive AeaSPR. We found replacement of Pro 238 (P238) in DrmSPR with the corresponding residue from AeaSPR (L310) reduced its SP sensitivity 2.7 fold without altering its MIP sensitivity. The P238 residue located in the third extracellular loop (ECL3) is conserved in Drosophila SPRs and in SPR from the moth Bombyx mori (BomSPR), which is considerably more sensitive to SP than AeaSPR, AngSPR, or ApcSPR. We found, however, that rather than improving AeaSPR's sensitivity to SP, replacement of L310 in AeaSPR with Pro significantly reduces its MIP sensitivity. Thus, our identification of a single amino acid residue critical for SP sensitivity, but not for MIP sensitivity is an important step in clarifying how DrmSPR evolved the ability to detect SP.


Asunto(s)
Aedes/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Receptores de Péptidos/genética , Aedes/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ligandos , Receptores de Péptidos/química , Receptores de Péptidos/metabolismo , Alineación de Secuencia
15.
Insect Biochem Mol Biol ; 124: 103414, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32589920

RESUMEN

In Drosophila melanogaster mating triggers profound changes in the behaviour and reproductive physiology of the female. Many of these post-mating effects are elicited by sex peptide (SP), a 36-mer pheromone made in the male accessory gland and passed to the female in the seminal fluid. The peptide comprises several structurally and functionally distinct domains, one of which consists of five 4-hydroxyprolines and induces a female immune response. The SP gene predicts an isoleucine (Ile14) sandwiched between two of the hydroxyprolines of the mature secreted peptide, but the identity of this residue was not established by peptide sequencing and amino acid analysis, presumably because of modification of the side chain. Here we have used matrix-assisted laser desorption ionisation mass spectrometry together with Fourier-transform ion cyclotron resonance mass spectrometry to show that Ile14 is modified by oxidation of the side chain - a very unusual post-translational modification. Mass spectrometric analysis of glands from different geographical populations of male D. melanogaster show that SP with six hydroxylated side chains is the most common form of the peptide, but that a sub-strain of Canton-S flies held at Leeds only has two or three hydroxylated prolines and an unmodified Ile14. The D. melanogaster genome has remarkably 17 putative hydroxylase genes that are strongly and almost exclusively expressed in the male accessory gland, suggesting that the gland is a powerhouse of protein oxidation. Strain variation in the pattern of sex peptide hydroxylation might be explained by differences in the expression of individual hydroxylase genes.


Asunto(s)
Proteínas de Drosophila/química , Drosophila melanogaster/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Genes de Insecto , Variación Genética , Hidroxilación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isoleucina/metabolismo , Oxigenasas de Función Mixta/genética , Atractivos Sexuales/química , Atractivos Sexuales/metabolismo , Conducta Sexual Animal/fisiología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos
16.
Arch Insect Biochem Physiol ; 104(2): e21665, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32091155

RESUMEN

Insect pests can cause crop damage in yield or quality, resulting in profit losses for farmers. The primary approach to control them is still the use of chemical pesticides resulting in significant hazards to the environment and human health. Biological control and the sterile insect technique are alternative strategies to improve agriculture protection. However, both strategies have significant limitations. A newly introduced approach that could be both effective and species-specific is the RNA interference mechanism. One key point for the success of this strategy is the delivery method of double-strand RNA (dsRNA) to the insects. A method of dsRNA delivery to insects with potential use in the field is the oral delivery, feeding the insects engineered microorganisms that produce dsRNA. Here, we present the first protocol for dsRNA feeding using modified bacteria, in the olive fruit fly, the most important insect pest of cultivated olives. We chose to target the sex peptide receptor gene. The sex peptide receptor interacts with the sex peptide, a peptide that is responsible for the postmating behavior in the model organism, Drosophila melanogaster. Feeding the female olive fruit fly with bacteria that produced dsRNA for the sex peptide receptor gene resulted in the development of female insects with significantly lower oviposition rates. Administration of dsRNA producing bacteria in insect diet against target genes that lead to genetic sexing or female-specific lethality could be added in the armory of control methods.


Asunto(s)
Proteínas de Insectos/genética , Oviposición/efectos de los fármacos , ARN Bicatenario/farmacología , Receptores de Péptidos/genética , Tephritidae/fisiología , Animales , Proteínas de Insectos/metabolismo , Receptores de Péptidos/metabolismo , Tephritidae/genética
17.
J Evol Biol ; 33(5): 629-641, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31991034

RESUMEN

Successful reproduction depends on interactions between numerous proteins beyond those involved directly in gamete fusion. Although such reproductive proteins evolve in response to sexual selection pressures, how networks of interacting proteins arise and evolve as reproductive phenotypes change remains an open question. Here, we investigated the molecular evolution of the 'sex peptide network' of Drosophila melanogaster, a functionally well-characterized reproductive protein network. In this species, the peptide hormone sex peptide (SP) and its interacting proteins cause major changes in female physiology and behaviour after mating. In contrast, females of more distantly related Drosophila species do not respond to SP. In spite of these phenotypic differences, we detected orthologs of all network proteins across 22 diverse Drosophila species and found evidence that most orthologs likely function in reproduction throughout the genus. Within SP-responsive species, we detected the recurrent, adaptive evolution of several network proteins, consistent with sexual selection acting to continually refine network function. We also found some evidence for adaptive evolution of several proteins along two specific phylogenetic lineages that correspond with increased expression of the SP receptor in female reproductive tracts or increased sperm length, respectively. Finally, we used gene expression profiling to examine the likely degree of functional conservation of the paralogs of an SP network protein that arose via gene duplication. Our results suggest a dynamic history for the SP network in which network members arose before the onset of robust SP-mediated responses and then were shaped by both purifying and positive selection.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Evolución Molecular , Péptidos y Proteínas de Señalización Intercelular/genética , Receptores de Péptidos/genética , Selección Sexual , Adaptación Biológica , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Femenino , Duplicación de Gen , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Receptores de Péptidos/metabolismo , Reproducción/genética , Serina Proteasas/genética , Serina Proteasas/metabolismo
18.
Neuron ; 102(5): 1025-1036.e6, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31072787

RESUMEN

Female behavior changes profoundly after mating. In Drosophila, the mechanisms underlying the long-term changes led by seminal products have been extensively studied. However, the effect of the sensory component of copulation on the female's internal state and behavior remains elusive. We pursued this question by dissociating the effect of coital sensory inputs from those of male ejaculate. We found that the sensory inputs of copulation cause a reduction of post-coital receptivity in females, referred to as the "copulation effect." We identified three layers of a neural circuit underlying this phenomenon. Abdominal neurons expressing the mechanosensory channel Piezo convey the signal of copulation to female-specific ascending neurons, LSANs, in the ventral nerve cord. LSANs relay this information to neurons expressing myoinhibitory peptides in the brain. We hereby provide a neural mechanism by which the experience of copulation facilitates females encoding their mating status, thus adjusting behavior to optimize reproduction.


Asunto(s)
Encéfalo/metabolismo , Copulación/fisiología , Proteínas de Drosophila/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Neuronas/metabolismo , Abdomen , Animales , Encéfalo/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster , Femenino , Ganglios Sensoriales/metabolismo , Ganglios Sensoriales/fisiología , Canales Iónicos/fisiología , Vías Nerviosas , Neuronas/fisiología , Conducta Sexual Animal/fisiología
19.
J Neurogenet ; 33(2): 44-51, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31084242

RESUMEN

G protein-coupled receptors (GPCRs) represent a family of seven-pass transmembrane protein receptors whose ligands include neuropeptides and small-molecule neuromodulators such as dopamine and serotonin. These neurotransmitters act at long distances and are proposed to define the ground state of the nervous system. The Drosophila genome encodes approximately 50 neuropeptides and their functions in physiology and behavior are now under intensive studies. Key information currently lacking in the field is the spatiotemporal activation patterns of endogenous GPCRs. Here we report application of the Tango system, a reporter assay to detect GPCR activity, to endogenous GPCRs in the fly genome. We developed a method to integrate the sensor component of the Tango system to the C-terminus of endogenous genes by using genome editing techniques. We demonstrate that Tango sensors in the Sex-peptide receptor (SPR) locus allow sensitive detection of mating-dependent SPR activity in the female reproductive organ. The method is easily applicable to any GPCR and will provide a way to systematically characterize GPCRs in the fly brain.


Asunto(s)
Proteínas de Drosophila/fisiología , Genes Reporteros , Técnicas Genéticas , Receptores de Péptidos/fisiología , Animales , Animales Modificados Genéticamente , Drosophila , Femenino , Masculino
20.
Neurochem Res ; 44(6): 1508-1516, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30661229

RESUMEN

Sleep is a highly conserved state in animals, but its regulation and physiological function is poorly understood. Drosophila melanogaster is an excellent model for studying sleep regulation and has been used to investigate how sex and social interactions can influence wake-sleep profiles. Previously we have shown that copulation has a profound effect on day time activity and quiescence (siesta sleep) of individual post-mated females. Here we have the studied the effect of mating and the transfer of the 36 amino acid sex peptide in the seminal fluid on the behavior of mated female Drosophila populations, where there will be on-going social interactions. The locomotor activity and sleep patterns of virgin and post-mated female D. melanogaster from three laboratory strains (Oregon-R, Canton-S and Dahomey) were recorded in social groups of 20 individuals in a 12-12 h light-dark cycle. Virgin female populations from all three fly strains displayed consolidated periods of low activity in between two sharp peaks of activity, corresponding to lights-on and lights-off. Similar light-correlated peaks were recorded for the mated female populations, however, the low afternoon activity and siesta seen in virgin populations was abolished after mating in all three strains. In contrast, night activity appeared unaffected. This post-mating effect was sustained for several days and was dependent on the male SP acting as a pheromone. Evidence from mixed populations of virgin and mated females suggests that the siesta of non-mated females is not easily disturbed by the presence of highly active post-mated females.


Asunto(s)
Conducta Animal/fisiología , Copulación/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Péptidos/fisiología , Sueño/fisiología , Conducta Social , Animales , Ritmo Circadiano/fisiología , Femenino , Péptidos y Proteínas de Señalización Intercelular , Locomoción/fisiología , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...