Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Heredity (Edinb) ; 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39369145

ABSTRACT

Mother's curse refers to male-biased deleterious mutations that may accumulate on mitochondria due to its strict maternal inheritance. If these mutations persist, males should ideally compensate through mutations on Y-chromosomes given its strict paternal inheritance. Previous work addressed this hypothesis by comparing coevolved and non-coevolved Y-mitochondria pairs placed alongside completely foreign autosomal backgrounds, expecting males with coevolved pairs to exhibit greater fitness due to Y-compensation. To date, no evidence for Y-compensation has been found. That experimental design assumes Y-chromosomes compensate via direct interaction with mitochondria and/or coevolved autosomes are unimportant in its function or elucidation. If Y-chromosomes instead compensate by modifying autosomal targets (or its elucidation requires coevolved autosomes), then this design could fail to detect Y-compensation. Here we address if Y-chromosomes ameliorate mitochondrial mutations affecting male lifespan in Drosophila melanogaster. Using three disparate populations we compared lifespan among males with coevolved and non-coevolved Y-mitochondria pairs placed alongside autosomal backgrounds coevolved with mitochondria. We found coevolved pairs exhibited lower mortality risk relative to non-coevolved pairs. In contrast, no such pattern was observed when coevolved and non-coevolved pairs were placed alongside non-coevolved autosomes, as with previous studies. These data are consistent with Y-compensation and highlight the importance of autosomes in this capacity. However, we cannot fully exclude the possibility that Y-autosomal coevolution independent of mitochondrial mutations contributed to our results. Regardless, modern practices in medicine, conservation, and agriculture that introduce foreign Y-chromosomes into non-coevolved backgrounds should be used with caution, as they may disrupt Y-autosome coadaptation and/or inadvertently unbridle mother's curse.

2.
Heredity (Edinb) ; 130(4): 236-241, 2023 04.
Article in English | MEDLINE | ID: mdl-36759734

ABSTRACT

How natural selection facilitates sexually dimorphic evolution despite a shared genome is unclear. The patrilineal inheritance of Y-chromosomes makes them an appealing solution. However, they have largely been dismissed due to their gene-poor, heterochromatic nature and because the additive genetic variation necessary for adaptive evolution is theoretically difficult to maintain. Further, previous empirical work has revealed mostly Y-linked sign epistatic variance segregating within populations, which can often impede adaptive evolution. To assess the evolutionary impact of Y-linked variation, we established replicate populations in Drosophila simulans containing multiple Y-chromosomes (YN populations) or a single Y-chromosome variant (Y1 populations) drawn from a single population. We estimated male and female heritabilities for several traits known to be influenced by Y-chromosomes, including the number of sternopleural bristles, abdominal bristles, sex comb teeth, and tibia length. A decrease in YN heritabilities compared with Y1 would be consistent with Y-chromosome variation being sign epistatic. A decrease in Y1 heritabilities would be consistent with Y-chromosome variation being additive, though additive-by-additive epistatic variation cannot be entirely dismissed. Female heritability estimates served as controls and were not expected to differ. We found male Y1 populations exhibited lower heritabilities for all traits except tibia length; consistent with Y-linked additivity (on average YN trait heritabilities were 25% greater than Y1). Female estimates showed no difference. These data suggest Y-chromosomes should play an important role in male trait evolution and may even influence sexually dimorphic evolution by shaping traits shared by both sexes.


Subject(s)
Drosophila melanogaster , Y Chromosome , Animals , Male , Female , Drosophila melanogaster/genetics , Phenotype , Genome , Epistasis, Genetic
3.
BMC Evol Biol ; 18(1): 204, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30587105

ABSTRACT

BACKGROUND: Variation in the non-coding regions of Y-chromosomes have been shown to influence gene regulation throughout the genome in some systems; a phenomenon termed Y-linked regulatory variation (YRV). This type of sex-specific genetic variance could have important implications for the evolution of male and female traits. If YRV contributes to the additive genetic variation of an autosomally coded trait shared between the sexes (e.g. body size), then selection could facilitate sexually dimorphic evolution via the Y-chromosome. In contrast, if YRV is entirely non-additive (i.e. interacts epistatically with other chromosomes), then Y-chromosomes could constrain trait evolution in both sexes whenever they are selected for the same trait value. The ability for this phenomenon to influence such fundamental evolutionary dynamics remains unexplored. RESULTS: Here we address the evolutionary contribution of Y-linked variance by selecting for improved male geotaxis in populations possessing multiple Y-chromosomes (i.e. possessed Y-linked additive and/or epistatic variation) or a single Y-chromosome variant (i.e. possessed no Y-linked variation). We found that males from populations possessing Y-linked variation did not significantly respond to selection; however, males from populations with no Y-linked variation did respond. These patterns suggest the presence of a large quantity of Y-linked epistatic variance in the multi-Y population that dramatically slowed its response. CONCLUSIONS: Our results imply that YRV is unlikely to facilitate the evolution of sexually dimorphic traits (at least for the trait examined here), but can interfere with the rate of trait evolution in both males and females. This result could have real biological implications as it suggests that YRV can affect how quickly a population responds to new selective pressures (e.g. invasive species, novel pathogens, or climate change). Considering that YRV influences hundreds of genes and is likely typical of other independently-evolved hemizygous chromosomes, YRV-like phenomena may represent common and significant costs to hemizygous sex determination.


Subject(s)
Drosophila melanogaster/genetics , Epistasis, Genetic , Y Chromosome , Animals , Drosophila melanogaster/physiology , Female , Gene Expression Regulation , Genetic Variation , Genome , Male , Sex Characteristics
4.
Proc Biol Sci ; 282(1820): 20151301, 2015 12 07.
Article in English | MEDLINE | ID: mdl-26631557

ABSTRACT

Sexually dimorphic phenotypes arise from the differential expression of male and female shared genes throughout the genome. Unfortunately, the underlying molecular mechanisms by which dimorphic regulation manifests and evolves are unclear. Recent work suggests that Y-chromosomes may play an important role, given that Drosophila melanogaster Ys were shown to influence the regulation of hundreds of X and autosomal genes. For Y-linked regulatory variation (YRV) to facilitate sexually dimorphic evolution, however, it must exist within populations (where selection operates) and influence male fitness. These criteria have seldom been investigated, leaving the potential for dimorphic evolution via YRV unclear. Interestingly, male and female D. melanogaster differ in immune gene regulation. Furthermore, immune gene regulation appears to be influenced by the Y-chromosome, suggesting it may contribute to dimorphic immune evolution. We address this possibility by introgressing Y-chromosomes from a single wild population into an isogenic background (to create Y-lines) and assessing immune gene regulation and bacterial defence. We found that Y-line males differed in their immune gene regulation and their ability to defend against Serratia marcescens. Moreover, gene expression and bacterial defence were positively genetically correlated. These data indicate that the Y-chromosome has the potential to shape the evolution of sexually dimorphic immunity in this system.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Y Chromosome/genetics , Animals , Biological Evolution , Drosophila melanogaster/microbiology , Female , Gene Expression Regulation , Male , Serratia marcescens/physiology , Sex Characteristics
5.
J Exp Biol ; 217(Pt 20): 3664-9, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25147243

ABSTRACT

As temperatures change, insects alter the amount of melanin in their cuticle to improve thermoregulation. However, melanin is also central to insect immunity, suggesting that thermoregulatory strategy may indirectly impact immune defense by altering the abundance of melanin pathway components (a hypothesis we refer to as thermoregulatory-dependent immune investment). This may be the case in the cricket Allonemobius socius, where warm environments (both seasonal and geographical) produced crickets with lighter cuticles and increased pathogen susceptibility. Unfortunately, the potential for thermoregulatory strategy to influence insect immunity has not been widely explored. Here we address the relationships between temperature, thermoregulatory strategy and immunity in the fruit fly Drosophila melanogaster. To this end, flies from two separate Canadian populations were reared in either a summer- or autumn-like environment. Shortly after adult eclosion, flies were moved to a common environment where their cuticle color and susceptibility to a bacterial pathogen (Pseudomonas aeruginosa) were measured. As with A. socius, individuals from summer-like environments exhibited lighter cuticles and increased pathogen susceptibility, suggesting that the thermoregulatory-immunity relationship is evolutionarily conserved across the hemimetabolous and holometabolous clades. If global temperatures continue to rise as expected, then thermoregulation might play an important role in host infection and mortality rates in systems that provide critical ecosystem services (e.g. pollination), or influence the prevalence of insect-vectored disease (e.g. malaria).


Subject(s)
Body Temperature Regulation/immunology , Disease Susceptibility , Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Immunity, Innate , Temperature , Animals , Canada , Drosophila melanogaster/physiology , Female , Male , Melanins/metabolism , Phenotype , Pigmentation , Pseudomonas aeruginosa/physiology , Seasons
6.
J Exp Biol ; 216(Pt 21): 4005-10, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23868839

ABSTRACT

To improve thermoregulation in colder environments, insects are expected to darken their cuticles with melanin via the phenoloxidase cascade, a phenomenon predicted by the thermal melanin hypothesis. However, the phenoloxidase cascade also plays a significant role in insect immunity, leading to the additional hypothesis that the thermal environment indirectly shapes immune function via direct selection on cuticle color. Support for the latter hypothesis comes from the cricket Allonemobius socius, where cuticle darkness and immune-related phenoloxidase activity increase with latitude. However, thermal environments vary seasonally as well as geographically, suggesting that seasonal plasticity in immunity may also exist. Although seasonal fluctuations in vertebrate immune function are common (because of flux in breeding or resource abundance), seasonality in invertebrate immunity has not been widely explored. We addressed this possibility by rearing crickets in simulated summer and fall environments and assayed their cuticle color and immune function. Prior to estimating immunity, crickets were placed in a common environment to minimize metabolic rate differences. Individuals reared under fall-like conditions exhibited darker cuticles, greater phenoloxidase activity and greater resistance to the bacteria Serratia marcescens. These data support the hypothesis that changes in the thermal environment modify cuticle color, which indirectly shapes immune investment through pleiotropy. This hypothesis may represent a widespread mechanism governing immunity in numerous systems, considering that most insects operate in seasonally and geographically variable thermal environments.


Subject(s)
Gryllidae/immunology , Gryllidae/microbiology , Immunity, Innate , Animals , Female , Gryllidae/physiology , Hemolymph/metabolism , Host-Pathogen Interactions , Male , Melanins/metabolism , Monophenol Monooxygenase/blood , Phenotype , Pigmentation , Random Allocation , Seasons , Serratia marcescens/physiology , South Carolina , Temperature
7.
Proc Biol Sci ; 279(1742): 3577-83, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22696524

ABSTRACT

All animals are under the constant threat of pathogenic infection. However, little is known regarding the influence of acute infection on sperm viability, particularly in female insects. This information is crucial for our understanding of mating and immune system coevolution, considering that females store sperm and serve as the site of sperm competition. Using the fruitfly, Drosophila melanogaster, we examined the influence of infection on sperm viability and storage. Twenty-four hours after haemocoel inoculation with a pathogen mimic (peptidoglycan, PGN) both sexes exhibited reduced sperm viability, indicating that systemic immune activation played a significant role in gamete survival. Surprisingly, sperm death did not appear to result from a reproductive-immune system trade-off, considering that sperm survived 24 h in vitro once removed from their somatic resources. Instead, our results are most consistent with death owing to immune effector collateral damage. We also examined the potential for sexually transmitted pathogens to influence sperm storage. Females mated with 'infected' males (created by dipping genitalia into a PGN solution) exhibited a higher proportion of empty sperm stores 48 h after mating compared to their controls. Remarkably, these data indicate that females may increase their fitness by removing 'infected' ejaculates from storage over time.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Pseudomonas aeruginosa/physiology , Animals , Cell Survival , Drosophila melanogaster/physiology , Escherichia coli , Female , Genitalia, Male/microbiology , Male , Peptidoglycan/administration & dosage , Pseudomonas aeruginosa/immunology , Random Allocation , Reproduction , Spermatozoa/cytology , Spermatozoa/physiology
8.
Proc Biol Sci ; 279(1748): 4740-6, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23034704

ABSTRACT

In recent years, studies have shown that reproductive effort decelerates in response to pathogenic infection. If infection substantially reduces a host's residual reproductive value (RRV), however, then an acceleration of effort may instead occur (e.g. terminal investment). Reproductive acceleration would theoretically allow hosts to maintain or exaggerate their sexual signal upon infection. This would create a deceptive message from the perspective of the chooser, who may unwittingly copulate with an infected mate to their detriment. Using the cricket Allonemobius socius, we assessed the potential for reduced RRV to accelerate male reproductive effort and create a dishonest signal. RRV was manipulated through male age and simulated pathogenic insult. Reproductive effort was measured as calling song energetics, mating success, latency to mate and nuptial gift size. We show that males adopted either an accelerated or decelerated reproductive strategy upon infection, and that this decision was probably mediated by RRV. Moreover, males who accelerated their effort produced a dishonest signal by increasing their song energetics while providing fewer paternal resources (i.e. smaller gifts). Our study is one of the few to document the existence of dishonest signals and relate dishonesty to a potential reduction in female fitness, underscoring the conflict inherent in sexual reproduction.


Subject(s)
Gryllidae/microbiology , Gryllidae/physiology , Sexual Behavior, Animal/physiology , Age Factors , Animals , Copulation , Dose-Response Relationship, Drug , Female , Gryllidae/drug effects , Lipopolysaccharides/pharmacology , Male , Sexual Behavior, Animal/drug effects , Vocalization, Animal
9.
Nature ; 429(6987): 65-7, 2004 May 06.
Article in English | MEDLINE | ID: mdl-15129280

ABSTRACT

Indirect-benefit models of sexual selection assert that females gain heritable offspring advantages through a mating bias for males of superior genetic quality. This has generally been tested by associating a simple morphological quality indicator (for example, bird tail length) with offspring viability. However, selection acts simultaneously on many characters, limiting the ability to detect significant associations, especially if the simple indicator is weakly correlated to male fitness. Furthermore, recent conceptual developments suggest that the benefits gained from such mating biases may be sex-specific because of sexually antagonistic genes that differentially influence male and female reproductive ability. A more suitable test of the indirect-benefit model would examine associations between an aggregate quality indicator (such as male mating success) and gender-specific adult fitness components, under the expectation that these components may trade off. Here, we show that a father's mating success in the cricket, Allonemobius socius, is positively genetically correlated with his son's mating success but negatively with his daughter's reproductive success. This provides empirical evidence that a female mating bias can result in sexually antagonistic offspring fitness.


Subject(s)
Gryllidae/physiology , Reproduction/physiology , Sexual Behavior, Animal/physiology , Animals , Body Constitution , Female , Gryllidae/genetics , Gryllidae/growth & development , Male , Models, Biological , Phenotype , Reproduction/genetics , Selection, Genetic , Sex Characteristics , Time Factors
10.
Proc Biol Sci ; 276(1659): 1109-17, 2009 Mar 22.
Article in English | MEDLINE | ID: mdl-19129115

ABSTRACT

Ecological immunology attempts to explain variation in immune function. Much of this work makes predictions about how potential hosts should invest in overall immunity. However, this 'overall' perspective under-emphasizes other critical aspects, such as the specificity, inducibility and timing of an immune response. Here, we investigate these aspects by examining gene regulation across several immune system components in both male and female Drosophila melanogaster prior to and after mating. To elucidate potentially important temporal dynamics, we also assayed several genes over time. We found that males and females emphasized different components of their immune system, however overall investment was similar. Specifically, the sexes emphasized different gene paralogues within major gene families, and males tended to invest more in gram-negative defence. By contrast, the inducibility of the immune response was both transient (lasting approx. 24 hours) and equal between the sexes. Furthermore, mating tended to induce humoral gene upregulation, while cell-mediated genes were unaffected. Within the humoral system, gram-negative bacterial defence genes exhibited a greater inducibility than those associated with fungal or gram-positive bacterial defence. Our results suggest that variation in the effectiveness of the immune response between the sexes may be driven by differences in emphasis rather than overall investment.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/physiology , Sex Characteristics , Sexual Behavior, Animal/physiology , Animals , Bacteria/immunology , Female , Fungi/immunology , Gene Expression Regulation , Male , Reproduction/immunology , Time Factors
11.
Proc Biol Sci ; 274(1614): 1211-7, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17311779

ABSTRACT

Reproductive costs are an essential component of evolutionary theory. For instance, an increase in reproduction is generally coupled with a decrease in immunocompetence shortly after mating. However, recent work in Drosophila melanogaster suggests that the potential to mount an immune response, as measured by the levels of immune gene expression, increases after mating. These data are in contrast to previous studies, which suggest that mating can reduce a fly's ability to survive an actual bacterial challenge (realized immunity). This pattern may be driven by some aspect of mating, independent of resource limitation, which reduces immune function by inhibiting the effective deployment of immune gene products. Though several studies have examined both the potential and the realized immunity after mating, none have examined these immune measures simultaneously. Here, we examined the link between the potential and the realized immunity in a sterile mutant of D. melanogaster. Shortly after mating, we found that female immune gene expression was high, but survival against infection was low. Surprisingly, this pattern was reversed within 24 h. Thus, estimates of immunity based on gene expression do not appear to reflect an actual ability to defend against pathogens in the hours following copulation. We discuss the possible mechanisms that may account for this pattern.


Subject(s)
Biological Evolution , Drosophila melanogaster/immunology , Gene Expression Regulation/immunology , Sexual Behavior, Animal/physiology , Animals , Drosophila melanogaster/microbiology , Drosophila melanogaster/physiology , Female , Pseudomonas aeruginosa/immunology , Survival Analysis , Time Factors
12.
J Insect Physiol ; 93-94: 36-41, 2016.
Article in English | MEDLINE | ID: mdl-27530304

ABSTRACT

Altering one's temperature preference (e.g. behavioral fever or behavioral chill) is a common immune defense among ectotherms that is likely to be evolutionarily conserved. However, the temperature chosen by an infected host may not be optimal for pathogen defense, causing preference to be inefficient. Here we examined the efficiency of temperature preference in Drosophila melanogaster infected with an LD50 of the gram negative bacteria Pseudomonas aeruginosa. To this end, we estimated the host's uninfected and infected temperature preferences as well as their optimal survival temperature. We found that flies decreased their preference from 26.3°C to 25.2°C when infected, and this preference was stable over 48h. Furthermore, the decrease in temperature preference was associated with an increased chance of surviving the infection. Nevertheless, the infected temperature preference did not coincide with the optimum temperature for infection survival, which lies at or below 21.4°C. These data suggest that the behavioral response to P. aeruginosa infection is considerably inefficient, and the mechanisms that may account for this pattern are discussed. Future studies of infected temperature preferences should document its efficiency, as this understudied aspect of behavioral immunity can provide important insight into preference evolution.


Subject(s)
Drosophila melanogaster/microbiology , Drosophila melanogaster/physiology , Pseudomonas aeruginosa/physiology , Animals , Cold Temperature , Female , Longevity
13.
Evolution ; 56(3): 590-6, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11989688

ABSTRACT

In many insect systems, males donate nuptial gifts to insure an effective copulation or as a form of paternal investment. However, if gift magnitude is both body size-limited and positively related to fitness, then the opportunity exists for the gift to promote the evolution of large male size. In the striped ground cricket, Allonemobius socius, males transfer a body size-limited, somatic nuptial gift that is comprised primarily of hemolymph. To address the implications of this gift on male size evolution, we quantified the intensity and direction of natural (fecundity) and sexual (mating success) selection over multiple generations. We found that male size was under strong positive sexual selection throughout the breeding season. This pattern of selection was similar in successive generations spanning multiple years. Male size was also under strong natural selection, with the largest males siring the most offspring. However, multivariate selection gradients indicated that gift size, and not male size, was the best predictor of female fecundity. In other words, direct fecundity selection for larger gifts placed indirect positive selection on male body size, supporting the hypothesis that nuptial gifts can influence the evolution of male body size in this system. Although female size was also under strong selection due to a size related fecundity advantage, it did not exceed selection on male size. The implications of these results with regard to the maintenance of the female-biased size dimorphic system are discussed.


Subject(s)
Biological Evolution , Body Constitution/genetics , Gryllidae/anatomy & histology , Analysis of Variance , Animals , Body Weight , Genomic Imprinting , Gryllidae/genetics , Gryllidae/physiology , In Vitro Techniques , Reproduction , Seasons , Wings, Animal/anatomy & histology
14.
Evolution ; 58(11): 2478-85, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15612291

ABSTRACT

One of the most common life history trade-offs in animals is the reduction in survivorship with increasing reproductive effort. Despite the prevalence of this pattern, its underlying physiological mechanisms are not well understood. Here we test the hypothesis that immune suppression mediates this phenotypic trade-off by manipulating reproductive effort and measuring immune function and mortality rates in the striped ground cricket, Allonemobius socius. Because A. socius males provide females with a hemolymph-based nuptial gift during copulation, and many structural components of immunity reside in the hemolymph, we also predicted that sexual selection may differentially affect how disease resistance evolves in males and females. We found that an increased mating effort resulted in a reduced immune ability, coupled with an increased rate in age-specific mortality for both sexes. Thus, immune suppression appears to be a link between reproductive effort and cost in this system. In addition, males and females appeared to differentially invest in several aspects of immunity prior to mating, with males exhibiting a higher concentration of circulating hemocytes and a superior bacterial defense capability. This pattern may be the result of previously established positive selection on gift size due to its affect on female fecundity. In short, female choice for larger gifts may lead to a sexually dimorphic immune ability.


Subject(s)
Gryllidae/immunology , Gryllidae/physiology , Hemolymph/immunology , Immunity/physiology , Longevity/physiology , Sexual Behavior, Animal/physiology , Age Factors , Animals , Female , Male , Melanins/metabolism , Monophenol Monooxygenase/metabolism , Muramidase/metabolism , Reproduction/physiology , Selection, Genetic , South Carolina
15.
J Insect Physiol ; 57(6): 778-83, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21419131

ABSTRACT

Sperm viability has been associated with the degree of promiscuity across species, as well as the degree of reproductive success within species. Thus, sperm survival within the female reproductive tract likely plays a key role in how mating systems evolve. In the fruit fly, Drosophila melanogaster, however, the extent and cause of sperm death has been the subject of recent debate. Here, we assess sperm death within the female reproductive tract of D. melanogaster following single and multiple matings in order to elucidate the extent of death and its potential mechanisms, including an acute female response to mating, female age and/or sperm senescence. We found no evidence that sperm viability was influenced by an acute female response or female age. We also found that rival ejaculates did not influence viability, supporting recent work in the system. Instead, the majority of death appears to be due to the aging of male gametes within the female, and that at least some dead resident sperm remain in the female after multiple mating. In contrast to earlier in vivo work, we found that overall sperm death was minimal (8.7%), indicating viability should have a negligible influence on female remating rates.


Subject(s)
Drosophila melanogaster/physiology , Sexual Behavior, Animal , Spermatozoa/cytology , Aging , Animals , Cell Survival , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Female , Male , Reproduction
16.
Evolution ; 65(2): 584-90, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21271997

ABSTRACT

Sperm competition is a potent postcopulatory selective force where sperm from rival males compete to fertilize a limited set of ova. Considering that sperm production is costly, we expect males to strategically allocate sperm in accordance with the level of competition. Accordingly, previous work has examined a male's strategic allocation in terms of sperm number. However, the seminal fluid proteins (Sfps) transferred along with sperm may also play a crucial role in competition. Surprisingly, the strategic allocation of Sfps has remained largely unexplored. Using Drosophila melanogaster, we examined the expression of three seminal fluid and four spermatogenesis genes in response to perceived sperm competition intensity by manipulating male density in a pre-mating and courtship environment. In the pre-mating environment, we found that males modified Sfp ratios by reducing the production of two spfs when potential rivals were present, while one Sfp and all spermatogenesis genes remained unaltered. In the courtship environment, males did not modify spermatogenesis or Sfp production in response to either rival males or female presence. Our data suggest that perceived competition in the pre-mating environment places a significant influence on Sfp allocation, which may be a general trend in promiscuous animal systems with internal fertilization.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Semen/chemistry , Seminal Plasma Proteins/metabolism , Spermatozoa/physiology , Animals , Competitive Behavior , Courtship , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Female , Male , Semen/physiology
17.
J Evol Biol ; 18(6): 1515-22, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16313464

ABSTRACT

In many animal systems, females exhibit a localized immune response to insemination that helps defend against sexually transmitted disease. However, this response may also kill sperm, reducing a male's reproductive potential. If males could suppress this response, they may be able to increase their sperm's representation in the female's reproductive tract, thereby increasing their fitness. Here we address the hypothesis that, under conditions of sperm competition, males interfere with female immunity. To test our hypothesis, we manipulated levels of female mating frequency (single vs. multiply mated) and seminal diversity (monandrous vs. polyandrous) in the cricket, Allonemobius socius and measured female immune response. As mating frequency increased, female hemocyte load decreased, indicating a general reproductive cost. As seminal diversity increased, phenoloxidase (PO) activity (in vitro measure of 'potential' macroparasitic defense) increased and encapsulation ability (in vivo measure of 'realized' macroparasitic defense) decreased in polyandrous females. These results suggest that males may manipulate female immunity by interrupting the pro-PO cascade, which begins with the activation of PO and ends in the encapsulation of invading foreign bodies. In other words, female immune function may serve as a battleground over which a sexual conflict is fought.


Subject(s)
Gryllidae/immunology , Gryllidae/physiology , Sexual Behavior, Animal/physiology , Spermatozoa/immunology , Animals , Female , Hemocytes/immunology , Male , Monophenol Monooxygenase/metabolism , Muramidase/metabolism , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL