1
|
Liu ZH, Zhai Y, Xia Y, Liao Q. Mating modifies oxidative stress in the brain and confers protection against Parkinson's Disease in a Drosophila model. Biochem Biophys Res Commun 2024; 737:150911. [PMID: 39481187 DOI: 10.1016/j.bbrc.2024.150911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/26/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024]
Abstract
Mating exerts profound and multifaceted effects on the physiology of female insects, particularly influencing metabolic alterations and bolstering stress resilience. Drosophila melanogaster has emerged as an excellent model to investigate the mechanism of neurodegenerative diseases. However, interplay between mating and its impact on the Drosophila brain remains a tantalizing enigma, awaiting elucidation. Herein, we reported that mating significantly improved the climbing and jumping activity in mated females compared to the virgins in Drosophila. Mating also reduced oxidative stress in the brain. Based on the results, we found that, mated females exhibited better behavioral performance and fewer loss of dopaminergic (DA) neurons than unmated females in PINK1 RNAi flies, a well-established Parkinson's disease (PD) model. Further study demonstrated that mating led to decreased iron content in the brain, a process associated with decreased Transferrin 1 (Tsf1) and Malvolio (Mvl) and increased ferritin. Additionally, mating inhibited expression of Duox and Nox, two NADPH oxidases in Drosophila. Furthermore, Kr-h1, a transcription factor of JH, acted downstream of mating to regulate genes involved in iron metabolism and NADPH oxidases. Collectively, the findings suggested a pivotal role of mating in regulating iron metabolism and NADPH oxidases in the brain of Drosophila. Consequently, considering mating status is imperative in scientific research, particularly in the context of neurological disorders.
Collapse
Affiliation(s)
- Zhi-Hua Liu
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui, 230009, PR China.
| | - Yuyin Zhai
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui, 230009, PR China
| | - Yanzhou Xia
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui, 230009, PR China
| | - Qiaoming Liao
- Guangxi Eco-Engineering Vocational &Technical College, Liuzhou, Guangxi, 545004, PR China
| |
Collapse
|
2
|
Basu A, Singh A, Sehgal S, Madaan T, Prasad NG. Starvation increases susceptibility to bacterial infection and promotes systemic pathogen proliferation in Drosophila melanogaster females. J Invertebr Pathol 2024; 207:108209. [PMID: 39322010 DOI: 10.1016/j.jip.2024.108209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/03/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Defense against pathogens and parasites requires substantial investment of energy and resources on part of the host. This makes the host immune function dependent on availability and accessibility of resources. A resource deprived host is therefore expected to be more susceptible to infections, although empirical results do not always align with this prediction. Limiting host access to resources can additionally impact within-host pathogen numbers, either directly by altering the amount of resources available to the pathogens for proliferation or indirectly by altering the efficiency of the host immune system. We tested for the effects of host starvation (complete deprivation of resources) on susceptibility to bacterial pathogens, and within-host pathogen proliferation, in Drosophila melanogaster females. Our results show that starvation increases post-infection mortality of the host, but in a pathogen-specific manner. This increase in mortality is always accompanied by increased within-host pathogen proliferation. We therefore propose that starvation compromises host resistance to bacterial infections in Drosophila melanogaster females thereby increasing susceptibility to infections.
Collapse
Affiliation(s)
- Aabeer Basu
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India.
| | - Aparajita Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India.
| | - Suhaas Sehgal
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India; Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland(2).
| | - Tanvi Madaan
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India; Institute of Science and Technology Austria, Klosterneuburg, Austria(2).
| | - Nagaraj Guru Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India.
| |
Collapse
|
3
|
Basu A, Singh A, Ruchitha BG, Prasad NG. Experimental adaptation to pathogenic infection ameliorates negative effects of mating on host post-infection survival in Drosophila melanogaster. ZOOLOGY 2024; 166:126198. [PMID: 39173303 DOI: 10.1016/j.zool.2024.126198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 07/28/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
Sexual activity (mating) negatively affects immune function in various insect species across both sexes. In Drosophila melanogaster females, mating increases susceptibility to pathogenic challenges and encourages within-host pathogen proliferation. This effect is pathogen and host genotype dependent. We tested if mating-induced increased susceptibility to infections is more, or less, severe in hosts experimentally adapted to pathogenic infection. We selected replicate D. melanogaster populations for increased post-infection survival following infection with a bacterial pathogen, Enterococcus faecalis. We found that females from the selected populations were better at surviving a pathogenic infection compared to the females from the control populations. This was true in the case of both the pathogen used for selection and other novel pathogens (i.e., pathogens the hosts have not encountered in recent history). Additionally, the negative effect of mating on post-infection survival was limited to only the females from control populations. Therefore, we have demonstrated that experimental selection for increased post-infection survival ameliorates negative effects of mating on host susceptibility to infections.
Collapse
Affiliation(s)
- Aabeer Basu
- Indian Institute of Science Education and Research (IISER), Mohali, India.
| | - Aparajita Singh
- Indian Institute of Science Education and Research (IISER), Mohali, India.
| | - B G Ruchitha
- Indian Institute of Science Education and Research (IISER), Mohali, India.
| | | |
Collapse
|
4
|
Afkhami M. Neurobiology of egg-laying behavior in Drosophila: neural control of the female reproductive system. J Neurogenet 2024; 38:47-61. [PMID: 39250036 DOI: 10.1080/01677063.2024.2396352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Egg-laying is one of the key aspects of female reproductive behavior in insects. Egg-laying has been studied since the dawn of Drosophila melanogaster as a model organism. The female's internal state, hormones, and external factors, such as nutrition, light, and social environment, affect egg-laying output. However, only recently, neurobiological features of egg-laying behavior have been studied in detail. fruitless and doublesex, two key players in the sex determination pathway, have become focal points in identifying neurons of reproductive significance in both central and peripheral nervous systems. The reproductive tract and external terminalia house sensory neurons that carry the sensory information of egg maturation, mating and egg-laying. These sensory signals include the presence of male accessory gland products and mechanical stimuli. The abdominal neuromere houses neurons that receive information from the reproductive tract, including sex peptide abdominal ganglion neurons (SAGs), and send their information to the brain. In the brain, neuronal groups like aDNs and pC1 clusters modulate egg-laying decision-making, and other neurons like oviINs and oviDNs are necessary for egg-laying itself. Lastly, motor neurons involved in egg-laying, which are mostly octopaminergic, reside in the abdominal neuromere and orchestrate the muscle movements required for laying the egg. Egg-laying neuronal control is important in various evolutionary processes like cryptic female choice, and using different Drosophila species can provide intriguing avenues for the future of the field.
Collapse
Affiliation(s)
- Mehrnaz Afkhami
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA
| |
Collapse
|
5
|
Ye C, Ho R, Moberg KH, Zheng JQ. Adverse impact of female reproductive signaling on age-dependent neurodegeneration after mild head trauma in Drosophila. eLife 2024; 13:RP97908. [PMID: 39213032 PMCID: PMC11364438 DOI: 10.7554/elife.97908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Environmental insults, including mild head trauma, significantly increase the risk of neurodegeneration. However, it remains challenging to establish a causative connection between early-life exposure to mild head trauma and late-life emergence of neurodegenerative deficits, nor do we know how sex and age compound the outcome. Using a Drosophila model, we demonstrate that exposure to mild head trauma causes neurodegenerative conditions that emerge late in life and disproportionately affect females. Increasing age-at-injury further exacerbates this effect in a sexually dimorphic manner. We further identify sex peptide signaling as a key factor in female susceptibility to post-injury brain deficits. RNA sequencing highlights a reduction in innate immune defense transcripts specifically in mated females during late life. Our findings establish a causal relationship between early head trauma and late-life neurodegeneration, emphasizing sex differences in injury response and the impact of age-at-injury. Finally, our findings reveal that reproductive signaling adversely impacts female response to mild head insults and elevates vulnerability to late-life neurodegeneration.
Collapse
Affiliation(s)
- Changtian Ye
- Department of Cell Biology, Emory University School of MedicineAtlantaUnited States
| | - Ryan Ho
- College of Art and Science, Emory UniversityAtlantaUnited States
| | - Kenneth H Moberg
- Department of Cell Biology, Emory University School of MedicineAtlantaUnited States
| | - James Q Zheng
- Department of Cell Biology, Emory University School of MedicineAtlantaUnited States
- Department of Neurology, Emory University School of MedicineAtlantaUnited States
- Center for Neurodegenerative Diseases, Emory University School of MedicineAtlantaUnited States
| |
Collapse
|
6
|
Darby AM, Okoro DO, Aredas S, Frank AM, Pearson WH, Dionne MS, Lazzaro BP. High sugar diets can increase susceptibility to bacterial infection in Drosophila melanogaster. PLoS Pathog 2024; 20:e1012447. [PMID: 39133760 PMCID: PMC11341100 DOI: 10.1371/journal.ppat.1012447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 08/22/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Overnutrition with dietary sugar can worsen infection outcomes in diverse organisms including insects and humans, through generally unknown mechanisms. In the present study, we show that adult Drosophila melanogaster fed high-sugar diets became more susceptible to infection by the Gram-negative bacteria Providencia rettgeri and Serratia marcescens. We found that P. rettgeri and S. marcescens proliferate more rapidly in D. melanogaster fed a high-sugar diet, resulting in increased probability of host death. D. melanogaster become hyperglycemic on the high-sugar diet, and we find evidence that the extra carbon availability may promote S. marcescens growth within the host. However, we found no evidence that increased carbon availability directly supports greater P. rettgeri growth. D. melanogaster on both diets fully induce transcription of antimicrobial peptide (AMP) genes in response to infection, but D. melanogaster provided with high-sugar diets show reduced production of AMP protein. Thus, overnutrition with dietary sugar may impair host immunity at the level of AMP translation. Our results demonstrate that dietary sugar can shape infection dynamics by impacting both host and pathogen, depending on the nutritional requirements of the pathogen and by altering the physiological capacity of the host to sustain an immune response.
Collapse
Affiliation(s)
- Andrea M. Darby
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| | - Destiny O. Okoro
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| | - Sophia Aredas
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
- University of California, Irvine, Irvine, California, United States of America
- Department of Microbiology, Cornell University, Ithaca, New York, United States of America
| | - Ashley M. Frank
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Battelle, Columbus, Ohio, United States of America
| | - William H. Pearson
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Marc S. Dionne
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Brian P. Lazzaro
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
7
|
Li H, Zhang W, Zhang Y, Guo X, Hou J, Li H, Wei J, Li X. Effects of pyriproxyfen on development and hormone of the aphis, Aphis craccivora (Hemiptera: Aphididae). JOURNAL OF ECONOMIC ENTOMOLOGY 2024:toae141. [PMID: 38935064 DOI: 10.1093/jee/toae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/28/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Pyriproxyfen (PPF) has been shown to affect the pupal stage and ecdysone levels in holometabolous insects, such as silkworms and mealworms. It remains unknown whether it affects hemimetabolous insects with their hormone levels in insects lacking a pupal stage. In this laboratory study, bioassays were conducted to investigate the effects of varying doses of PPF on Aphis craccivora Koch (Hemiptera: Aphididae). Ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was used to determine the types and titers of juvenile hormone (JH) and 20-hydroxyecdysone (20E). Additionally, the effects of PPF on A. craccivora reproduction and molting, as well as its influence on relevant gene expression, were examined. The results revealed LC50 and LC90 values of 3.84 and 7.49 mg/l for PPF, respectively, after 48 h of exposure. The results demonstrated a significant reduction in the titer of JH III and a significant increase in the titer of 20E following treatment with PPF. However, there was no significant decrease observed in the titer of JH III skipped bisepoxide (JH SB3). A sublethal concentration of PPF was found to inhibit Krüppel homolog 1 (kr-h1) gene expression and reduce aphid reproduction, but it did not significantly impact ecdysone receptor expression and aphid molting. The results of this study demonstrate that PPF exhibits a lethal effect on aphids, thereby providing an effective means of control. Additionally, sublethal concentrations of PPF have been found to inhibit the JH in aphids, resulting in a decline in their reproductive ability and achieving the desired control objectives.
Collapse
Affiliation(s)
- Haolin Li
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Wenjie Zhang
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Yongheng Zhang
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Xiaxia Guo
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Jiangan Hou
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Honghong Li
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Jiguang Wei
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| | - Xuesheng Li
- Guangxi Key Laboratory of Agric-Environment and Agric-Products Safety, College of Agriculture, Guangxi University, Nanning, Guangxi 530004, People's Republic of China
| |
Collapse
|
8
|
Ye C, Ho R, Moberg KH, Zheng JQ. Sexual Dimorphism in Age-Dependent Neurodegeneration After Mild Head Trauma in Drosophila : Unveiling the Adverse Impact of Female Reproductive Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583747. [PMID: 38496515 PMCID: PMC10942469 DOI: 10.1101/2024.03.06.583747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Environmental insults, including mild head trauma, significantly increase the risk of neurodegeneration. However, it remains challenging to establish a causative connection between early-life exposure to mild head trauma and late-life emergence of neurodegenerative deficits, nor do we know how sex and age compound the outcome. Using a Drosophila model, we demonstrate that exposure to mild head trauma causes neurodegenerative conditions that emerge late in life and disproportionately affect females. Increasing age-at-injury further exacerbates this effect in a sexually dimorphic manner. We further identify Sex Peptide (SP) signaling as a key factor in female susceptibility to post-injury brain deficits. RNA sequencing highlights a reduction in innate immune defense transcripts specifically in mated females during late life. Our findings establish a causal relationship between early head trauma and late-life neurodegeneration, emphasizing sex differences in injury response and the impact of age-at-injury. Finally, our findings reveal that reproductive signaling adversely impacts female response to mild head insults and elevates vulnerability to late-life neurodegeneration.
Collapse
|
9
|
Taracena-Agarwal ML, Walter-Nuno AB, Bottino-Rojas V, Mejia APG, Xu K, Segal S, Dotson EM, Oliveira PL, Paiva-Silva GO. Juvenile Hormone as a contributing factor in establishing midgut microbiota for fecundity and fitness enhancement in adult female Aedes aegypti. Commun Biol 2024; 7:687. [PMID: 38839829 PMCID: PMC11153597 DOI: 10.1038/s42003-024-06334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 05/15/2024] [Indexed: 06/07/2024] Open
Abstract
Understanding the factors influencing mosquitoes' fecundity and longevity is important for designing better and more sustainable vector control strategies, as these parameters can impact their vectorial capacity. Here, we address how mating affects midgut growth in Aedes aegypti, what role Juvenile Hormone (JH) plays in this process, and how it impacts the mosquito's immune response and microbiota. Our findings reveal that mating and JH induce midgut growth. Additionally, the establishment of a native bacterial population in the midgut due to JH-dependent suppression of the immune response has important reproductive outcomes. Specific downregulation of AMPs with an increase in bacteria abundance in the gut results in increased egg counts and longer lifespans. Overall, these findings provide evidence of a cross-talk between JH response, gut epithelial tissue, cell cycle regulation, and the mechanisms governing the trade-offs between nutrition, immunity, and reproduction at the cellular level in the mosquito gut.
Collapse
Affiliation(s)
- Mabel L Taracena-Agarwal
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil.
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA.
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA.
| | - Ana Beatriz Walter-Nuno
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Vanessa Bottino-Rojas
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | | | - Kelsey Xu
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
| | - Steven Segal
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
| | - Ellen M Dotson
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Pedro L Oliveira
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Gabriela O Paiva-Silva
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil.
| |
Collapse
|
10
|
Gomez RA, Dallai R, Sims-West DJ, Mercati D, Sinka R, Ahmed-Braimah Y, Pitnick S, Dorus S. Proteomic diversification of spermatostyles among six species of whirligig beetles. Mol Reprod Dev 2024; 91:e23745. [PMID: 38785179 PMCID: PMC11246569 DOI: 10.1002/mrd.23745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Seminal fluid protein composition is complex and commonly assumed to be rapidly divergent due to functional interactions with both sperm and the female reproductive tract (FRT), both of which evolve rapidly. In addition to sperm, seminal fluid may contain structures, such as mating plugs and spermatophores. Here, we investigate the evolutionary diversification of a lesser-known ejaculate structure: the spermatostyle, which has independently arisen in several families of beetles and true bugs. We characterized the spermatostyle proteome, in addition to spermatostyle and FRT morphology, in six species of whirligig beetles (family Gyrinidae). Spermatostyles were enriched for proteolytic enzymes, and assays confirmed they possess proteolytic activity. Sperm-leucylaminopeptidases (S-LAPs) were particularly abundant, and their localization to spermatostyles was confirmed by immunohistochemistry. Although there was evidence for functional conservation of spermatostyle proteomes across species, phylogenetic regressions suggest evolutionary covariation between protein composition and the morphology of both spermatostyles and FRTs. We postulate that S-LAPs (and other proteases) have evolved a novel structural role in spermatostyles and discuss spermatostyles as adaptations for delivering male-derived materials to females.
Collapse
Affiliation(s)
- R. Antonio Gomez
- Department of Biology, Center for Reproductive Evolution, Syracuse University, Syracuse, New York, USA
| | - Romano Dallai
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Dylan J. Sims-West
- Department of Biology, Center for Reproductive Evolution, Syracuse University, Syracuse, New York, USA
| | - David Mercati
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Rita Sinka
- Department of Genetics, University of Szeged, Szeged, Hungary
| | - Yasir Ahmed-Braimah
- Department of Biology, Center for Reproductive Evolution, Syracuse University, Syracuse, New York, USA
| | - Scott Pitnick
- Department of Biology, Center for Reproductive Evolution, Syracuse University, Syracuse, New York, USA
| | - Steve Dorus
- Department of Biology, Center for Reproductive Evolution, Syracuse University, Syracuse, New York, USA
| |
Collapse
|
11
|
Hixson B, Huot L, Morejon B, Yang X, Nagy P, Michel K, Buchon N. The transcriptional response in mosquitoes distinguishes between fungi and bacteria but not Gram types. BMC Genomics 2024; 25:353. [PMID: 38594632 PMCID: PMC11003161 DOI: 10.1186/s12864-024-10153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/22/2024] [Indexed: 04/11/2024] Open
Abstract
Mosquitoes are prolific vectors of human pathogens, therefore a clear and accurate understanding of the organization of their antimicrobial defenses is crucial for informing the development of transmission control strategies. The canonical infection response in insects, as described in the insect model Drosophila melanogaster, is pathogen type-dependent, with distinct stereotypical responses to Gram-negative bacteria and Gram-positive bacteria/fungi mediated by the activation of the Imd and Toll pathways, respectively. To determine whether this pathogen-specific discrimination is shared by mosquitoes, we used RNAseq to capture the genome-wide transcriptional response of Aedes aegypti and Anopheles gambiae (s.l.) to systemic infection with Gram-negative bacteria, Gram-positive bacteria, yeasts, and filamentous fungi, as well as challenge with heat-killed Gram-negative, Gram-positive, and fungal pathogens. From the resulting data, we found that Ae. aegypti and An. gambiae both mount a core response to all categories of infection, and this response is highly conserved between the two species with respect to both function and orthology. When we compared the transcriptomes of mosquitoes infected with different types of bacteria, we observed that the intensity of the transcriptional response was correlated with both the virulence and growth rate of the infecting pathogen. Exhaustive comparisons of the transcriptomes of Gram-negative-challenged versus Gram-positive-challenged mosquitoes yielded no difference in either species. In Ae. aegypti, however, we identified transcriptional signatures specific to bacterial infection and to fungal infection. The bacterial infection response was dominated by the expression of defensins and cecropins, while the fungal infection response included the disproportionate upregulation of an uncharacterized family of glycine-rich proteins. These signatures were also observed in Ae. aegypti challenged with heat-killed bacteria and fungi, indicating that this species can discriminate between molecular patterns that are specific to bacteria and to fungi.
Collapse
Affiliation(s)
- Bretta Hixson
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Louise Huot
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Xiaowei Yang
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
- Current address: State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute for Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Peter Nagy
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Nicolas Buchon
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
12
|
Yamanaka N. Germ cell migration: Unexpected role of juvenile hormone before juvenile stages. Curr Biol 2024; 34:R84-R86. [PMID: 38320477 DOI: 10.1016/j.cub.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Juvenile hormone is best known for its role in maintaining juvenile-stage insects in their immature states during postembryonic development. A new study finds an unexpected role for this signaling lipid in guiding primordial germ cell migration during embryogenesis - possibly an ancestral function of isoprenoid signaling molecules.
Collapse
Affiliation(s)
- Naoki Yamanaka
- Department of Entomology, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
13
|
Jones TB, Mackey T, Juba AN, Amin K, Atyam A, McDole M, Yancy J, Thomas TC, Buhlman LM. Mild traumatic brain injury in Drosophila melanogaster alters reactive oxygen and nitrogen species in a sex-dependent manner. Exp Neurol 2024; 372:114621. [PMID: 38029809 PMCID: PMC10872660 DOI: 10.1016/j.expneurol.2023.114621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
Traumatic brain injury (TBI) is an outside force causing a modification in brain function and/or structural brain pathology that upregulates brain inducible nitric oxide synthase (iNOS), instigating increased levels of nitric oxide activity which is implicated in secondary pathology leading to behavioral deficits (Hall et al., 2012; Garry et al., 2015; Kozlov et al., 2017). In mammals, TBI-induced NO production activates an immune response and potentiates metabolic crisis through mitochondrial dysfunction coupled with vascular dysregulation; however, the direct influence on pathology is complicated by the activation of numerous secondary cascades and activation of other reactive oxygen species. Drosophila TBI models have demonstrated key features of mammalian TBI, including temporary incapacitation, disorientation, motor deficits, activation of innate immunity (inflammation), and autophagy responses observed immediately after injury (Katzenberger et al., 2013; Barekat et al., 2016; Simon et al., 2017; Anderson et al., 2018; Buhlman et al., 2021b). We hypothesized that acute behavioral phenotypes would be associated with deficits in climbing behavior and increased oxidative stress. Because flies lack mammalian-like cardiovascular and adaptive immune systems, we were able to make our observations in the absence of vascular disruption and adaptive immune system interference in a system where highly targeted interventions can be rapidly evaluated. To demonstrate the induction of injury, ten-day-old transgenic flies received an injury of increasing angles from a modified high impact trauma (HIT) device where angle-dependent increases occurred for acute neurological behavior assessments and twenty-four-hour mortality, and survival was significantly decreased. Injury caused sex-dependent effects on climbing activity and measures of oxidative stress. Specifically, after a single 60-degree HIT, female flies exhibited significant impairments in climbing activity beyond that observed in male flies. We also found that several measures of oxidative stress, including Drosophila NOS (dNOS) expression, protein nitration, and hydrogen peroxide production were significantly decreased in female flies. Interestingly, protein nitration was also decreased in males, but surpassed sham levels with a more severe injury. We also observed decreased autophagy demand in vulnerable dopaminergic neurons in female, but not male flies. In addition, mitophagy initiation was decreased in females. Collectively, our data suggest that TBI in flies induces acute behavioral phenotypes and climbing deficits that are analogous to mammalian TBI. We also observed that various indices of oxidative stress, including dNOS expression, protein tyrosine nitration, and hydrogen peroxide levels, as well as basal levels of autophagy, are altered in response to injury, an effect that is more pronounced in female flies.
Collapse
Affiliation(s)
- T Bucky Jones
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA; Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Tracy Mackey
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Amber N Juba
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA
| | - Kush Amin
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Amruth Atyam
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Madison McDole
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Jarod Yancy
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Phoenix VA Health Care System, Phoenix, AZ, USA.
| | - Lori M Buhlman
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
14
|
Hopkins BR, Angus-Henry A, Kim BY, Carlisle JA, Thompson A, Kopp A. Decoupled evolution of the Sex Peptide gene family and Sex Peptide Receptor in Drosophilidae. Proc Natl Acad Sci U S A 2024; 121:e2312380120. [PMID: 38215185 PMCID: PMC10801855 DOI: 10.1073/pnas.2312380120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/16/2023] [Indexed: 01/14/2024] Open
Abstract
Across internally fertilising species, males transfer ejaculate proteins that trigger wide-ranging changes in female behaviour and physiology. Much theory has been developed to explore the drivers of ejaculate protein evolution. The accelerating availability of high-quality genomes now allows us to test how these proteins are evolving at fine taxonomic scales. Here, we use genomes from 264 species to chart the evolutionary history of Sex Peptide (SP), a potent regulator of female post-mating responses in Drosophila melanogaster. We infer that SP first evolved in the Drosophilinae subfamily and has since followed markedly different evolutionary trajectories in different lineages. Outside of the Sophophora-Lordiphosa, SP exists largely as a single-copy gene with independent losses in several lineages. Within the Sophophora-Lordiphosa, the SP gene family has repeatedly and independently expanded. Up to seven copies, collectively displaying extensive sequence variation, are present in some species. Despite these changes, SP expression remains restricted to the male reproductive tract. Alongside, we document considerable interspecific variation in the presence and morphology of seminal microcarriers that, despite the critical role SP plays in microcarrier assembly in D. melanogaster, appears to be independent of changes in the presence/absence or sequence of SP. We end by providing evidence that SP's evolution is decoupled from that of its receptor, Sex Peptide Receptor, in which we detect no evidence of correlated diversifying selection. Collectively, our work describes the divergent evolutionary trajectories that a novel gene has taken following its origin and finds a surprisingly weak coevolutionary signal between a supposedly sexually antagonistic protein and its receptor.
Collapse
Affiliation(s)
- Ben R. Hopkins
- Department of Evolution and Ecology, University of California, Davis, CA95616
| | - Aidan Angus-Henry
- Department of Evolution and Ecology, University of California, Davis, CA95616
| | - Bernard Y. Kim
- Department of Biology, Stanford University, Stanford, CA94305
| | - Jolie A. Carlisle
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY14853
| | - Ammon Thompson
- Department of Evolution and Ecology, University of California, Davis, CA95616
| | - Artyom Kopp
- Department of Evolution and Ecology, University of California, Davis, CA95616
| |
Collapse
|
15
|
Hopkins BR, Angus-Henry A, Kim BY, Carlisle JA, Thompson A, Kopp A. Decoupled evolution of the Sex Peptide gene family and Sex Peptide Receptor in Drosophilidae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547128. [PMID: 37425821 PMCID: PMC10327216 DOI: 10.1101/2023.06.29.547128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Across internally fertilising species, males transfer ejaculate proteins that trigger wide-ranging changes in female behaviour and physiology. Much theory has been developed to explore the drivers of ejaculate protein evolution. The accelerating availability of high-quality genomes now allows us to test how these proteins are evolving at fine taxonomic scales. Here, we use genomes from 264 species to chart the evolutionary history of Sex Peptide (SP), a potent regulator of female post-mating responses in Drosophila melanogaster. We infer that SP first evolved in the Drosophilinae subfamily and has followed markedly different evolutionary trajectories in different lineages. Outside of the Sophophora-Lordiphosa, SP exists largely as a single-copy gene with independent losses in several lineages. Within the Sophophora-Lordiphosa, the SP gene family has repeatedly and independently expanded. Up to seven copies, collectively displaying extensive sequence variation, are present in some species. Despite these changes, SP expression remains restricted to the male reproductive tract. Alongside, we document considerable interspecific variation in the presence and morphology of seminal microcarriers that, despite the critical role SP plays in microcarrier assembly in D. melanogaster, appear to be independent of changes in the presence/absence or sequence of SP. We end by providing evidence that SP's evolution is decoupled from that of its receptor, SPR, in which we detect no evidence of correlated diversifying selection. Collectively, our work describes the divergent evolutionary trajectories that a novel gene has taken following its origin and finds a surprisingly weak coevolutionary signal between a supposedly sexually antagonistic protein and its receptor.
Collapse
Affiliation(s)
- Ben R. Hopkins
- Department of Evolution and Ecology, University of California – Davis, CA, USA
| | - Aidan Angus-Henry
- Department of Evolution and Ecology, University of California – Davis, CA, USA
| | | | - Jolie A. Carlisle
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Ammon Thompson
- Department of Evolution and Ecology, University of California – Davis, CA, USA
| | - Artyom Kopp
- Department of Evolution and Ecology, University of California – Davis, CA, USA
| |
Collapse
|
16
|
Tumova S, Dolezel D, Jindra M. Conserved and Unique Roles of bHLH-PAS Transcription Factors in Insects - From Clock to Hormone Reception. J Mol Biol 2023; 436:168332. [PMID: 39491146 DOI: 10.1016/j.jmb.2023.168332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
A dozen bHLH-PAS transcription factors have evolved since the dawn of the animal kingdom; nine of them have mutual orthologs between arthropods and vertebrates. These proteins are master regulators in a range of developmental processes from organogenesis, nervous system formation and functioning, to cell fate decisions defining identity of limbs or photoreceptors for color vision. Among the functionally best conserved are bHLH-PAS proteins acting in the animal circadian clock. On the other side of the spectrum are fundamental physiological mechanisms such as those underlying xenobiotic detoxification, oxygen homeostasis, and metabolic adaptation to hypoxia, infection or tumor progression. Predictably, malfunctioning of bHLH-PAS regulators leads to pathologies. Performance of the individual bHLH-PAS proteins is modulated at multiple levels including dimerization and other protein-protein interactions, proteasomal degradation, and by binding low-molecular weight ligands. Despite the vast evolutionary gap dividing arthropods and vertebrates, and the differences in their anatomy, many functions of orthologous bHLH-PAS proteins are remarkably similar, including at the molecular level. Our phylogenetic analysis shows that one bHLH-PAS protein type has been lost during vertebrate evolution. This protein has a unique function as a receptor of the sesquiterpenoid juvenile hormones of insects and crustaceans. Although some other bHLH-PAS proteins are regulated by binding small molecules, the juvenile hormone receptor presents an unprecedented case, since all other non-peptide animal hormones activate members of the nuclear receptor family. The purpose of this review is to compare and highlight parallels and differences in functioning of bHLH-PAS proteins between insects and vertebrates.
Collapse
Affiliation(s)
- Sarka Tumova
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic
| | - David Dolezel
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic
| | - Marek Jindra
- Institute of Entomology, Biology Center of the Czech Academy of Sciences, Ceske Budejovice 37005, Czech Republic.
| |
Collapse
|
17
|
Magistrado D, El-Dougdoug NK, Short SM. Sugar restriction and blood ingestion shape divergent immune defense trajectories in the mosquito Aedes aegypti. Sci Rep 2023; 13:12368. [PMID: 37524824 PMCID: PMC10390476 DOI: 10.1038/s41598-023-39067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/19/2023] [Indexed: 08/02/2023] Open
Abstract
Immune defense is comprised of (1) resistance: the ability to reduce pathogen load, and (2) tolerance: the ability to limit the disease severity induced by a given pathogen load. The study of tolerance in the field of animal immunity is fairly nascent in comparison to resistance. Consequently, studies which examine immune defense comprehensively (i.e. considering both resistance and tolerance in conjunction) are uncommon, despite their exigency in achieving a thorough understanding of immune defense. Furthermore, understanding tolerance in arthropod disease vectors is uniquely relevant, as tolerance is essential to the cyclical transmission of pathogens by arthropods. Here, we tested the effect(s) of dietary sucrose concentration and blood ingestion on resistance and tolerance to Escherichia coli infection in the yellow fever mosquito Aedes aegypti. Resistance and tolerance were measured concurrently and at multiple timepoints. We found that mosquitoes from the restricted sugar treatment displayed enhanced resistance at all timepoints post-infection compared to those from the laboratory standard sugar treatment. Blood also improved resistance, but only early post-infection. While sucrose restriction had no effect on tolerance, we show that consuming blood prior to bacterial infection ameliorates a temporal decline in tolerance that mosquitoes experience when provided with only sugar meals. Taken together, our findings indicate that different dietary components can have unique and sometimes temporally dynamic impacts on resistance and tolerance.
Collapse
Affiliation(s)
- Dom Magistrado
- Department of Entomology, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH, USA
| | - Noha K El-Dougdoug
- Department of Entomology, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH, USA
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, Egypt
| | - Sarah M Short
- Department of Entomology, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Hixson B, Huot L, Morejon B, Yang X, Nagy P, Michel K, Buchon N. The transcriptional response in mosquitoes distinguishes between fungi and bacteria but not Gram types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550663. [PMID: 37546902 PMCID: PMC10402080 DOI: 10.1101/2023.07.26.550663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Mosquitoes are prolific vectors of human pathogens; a clear and accurate understanding of the organization of their antimicrobial defenses is crucial for informing the development of transmission control strategies. The canonical infection response in insects, as described in the insect model Drosophila melanogaster , is pathogen type-dependent, with distinct stereotypical responses to Gram-negative bacteria and Gram-positive bacteria/fungi mediated by the activation of the Imd and Toll pathways, respectively. To determine whether this pathogen-specific discrimination is shared by mosquitoes, we used RNAseq to capture the genome-wide transcriptional response of Aedes aegypti and Anopheles gambiae ( s.l. ) to systemic infection with Gram-negative bacteria, Gram-positive bacteria, yeasts, and filamentous fungi, as well as challenge with heat-killed Gram-negative, Gram-positive, and fungal pathogens. From the resulting data, we found that Ae. aegypti and An. gambiae both mount a core response to all categories of infection, and this response is highly conserved between the two species with respect to both function and orthology. When we compared the transcriptomes of mosquitoes infected with different types of bacteria, we observed that the intensity of the transcriptional response was correlated with both the virulence and growth rate of the infecting pathogen. Exhaustive comparisons of the transcriptomes of Gram-negative-challenged versus Gram-positive-challenged mosquitoes yielded no difference in either species. In Ae. aegypti , however, we identified transcriptional signatures specific to bacterial infection and to fungal infection. The bacterial infection response was dominated by the expression of defensins and cecropins, while the fungal infection response included the disproportionate upregulation of an uncharacterized family of glycine-rich proteins. These signatures were also observed in Ae. aegypti challenged with heat-killed bacteria and fungi, indicating that this species can discriminate between molecular patterns that are specific to bacteria and to fungi.
Collapse
|
19
|
Abstract
Endocrine signaling networks control diverse biological processes and life history traits across metazoans. In both invertebrate and vertebrate taxa, steroid hormones regulate immune system function in response to intrinsic and environmental stimuli, such as microbial infection. The mechanisms of this endocrine-immune regulation are complex and constitute an ongoing research endeavor facilitated by genetically tractable animal models. The 20-hydroxyecdysone (20E) is the major steroid hormone in arthropods, primarily studied for its essential role in mediating developmental transitions and metamorphosis; 20E also modulates innate immunity in a variety of insect taxa. This review provides an overview of our current understanding of 20E-mediated innate immune responses. The prevalence of correlations between 20E-driven developmental transitions and innate immune activation are summarized across a range of holometabolous insects. Subsequent discussion focuses on studies conducted using the extensive genetic resources available in Drosophila that have begun to reveal the mechanisms underlying 20E regulation of immunity in the contexts of both development and bacterial infection. Lastly, I propose directions for future research into 20E regulation of immunity that will advance our knowledge of how interactive endocrine networks coordinate animals' physiological responses to environmental microbes.
Collapse
Affiliation(s)
- Scott A. Keith
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
20
|
Imrie RM, Walsh SK, Roberts KE, Lello J, Longdon B. Investigating the outcomes of virus coinfection within and across host species. PLoS Pathog 2023; 19:e1011044. [PMID: 37216391 DOI: 10.1371/journal.ppat.1011044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
Interactions between coinfecting pathogens have the potential to alter the course of infection and can act as a source of phenotypic variation in susceptibility between hosts. This phenotypic variation may influence the evolution of host-pathogen interactions within host species and interfere with patterns in the outcomes of infection across host species. Here, we examine experimental coinfections of two Cripaviruses-Cricket Paralysis Virus (CrPV), and Drosophila C Virus (DCV)-across a panel of 25 Drosophila melanogaster inbred lines and 47 Drosophilidae host species. We find that interactions between these viruses alter viral loads across D. melanogaster genotypes, with a ~3 fold increase in the viral load of DCV and a ~2.5 fold decrease in CrPV in coinfection compared to single infection, but we find little evidence of a host genetic basis for these effects. Across host species, we find no evidence of systematic changes in susceptibility during coinfection, with no interaction between DCV and CrPV detected in the majority of host species. These results suggest that phenotypic variation in coinfection interactions within host species can occur independently of natural host genetic variation in susceptibility, and that patterns of susceptibility across host species to single infections can be robust to the added complexity of coinfection.
Collapse
Affiliation(s)
- Ryan M Imrie
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| | - Sarah K Walsh
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| | - Katherine E Roberts
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| | - Joanne Lello
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Ben Longdon
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| |
Collapse
|
21
|
Ye C, Behnke JA, Hardin KR, Zheng JQ. Drosophila melanogaster as a model to study age and sex differences in brain injury and neurodegeneration after mild head trauma. Front Neurosci 2023; 17:1150694. [PMID: 37077318 PMCID: PMC10106652 DOI: 10.3389/fnins.2023.1150694] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Repetitive physical insults to the head, including those that elicit mild traumatic brain injury (mTBI), are a known risk factor for a variety of neurodegenerative conditions including Alzheimer's disease (AD), Parkinson's disease (PD), and chronic traumatic encephalopathy (CTE). Although most individuals who sustain mTBI typically achieve a seemingly full recovery within a few weeks, a subset experience delayed-onset symptoms later in life. As most mTBI research has focused on the acute phase of injury, there is an incomplete understanding of mechanisms related to the late-life emergence of neurodegeneration after early exposure to mild head trauma. The recent adoption of Drosophila-based brain injury models provides several unique advantages over existing preclinical animal models, including a tractable framework amenable to high-throughput assays and short relative lifespan conducive to lifelong mechanistic investigation. The use of flies also provides an opportunity to investigate important risk factors associated with neurodegenerative conditions, specifically age and sex. In this review, we survey current literature that examines age and sex as contributing factors to head trauma-mediated neurodegeneration in humans and preclinical models, including mammalian and Drosophila models. We discuss similarities and disparities between human and fly in aging, sex differences, and pathophysiology. Finally, we highlight Drosophila as an effective tool for investigating mechanisms underlying head trauma-induced neurodegeneration and for identifying therapeutic targets for treatment and recovery.
Collapse
Affiliation(s)
- Changtian Ye
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Joseph A. Behnke
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine R. Hardin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - James Q. Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
22
|
Amaro-Sánchez T, Ruiz-Guzmán G, Hernández-Martínez S, Krams I, Rantala MJ, Contreras-Garduño J. Effect of juvenile hormone on phenoloxidase and hemocyte number: The role of age, sex, and immune challenge. Comp Biochem Physiol B Biochem Mol Biol 2023; 265:110827. [PMID: 36610635 DOI: 10.1016/j.cbpb.2023.110827] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Hormones are key factors in determining the response of organisms to their environment. For example, the juvenile hormone (JH) coordinates the insects' development, reproduction, and survival. However, it is still unclear how the impact of juvenile hormone on insect immunity varies depending on the sex and reproductive state of the individual, as well as the type of the immune challenge (i.e., Gram-positive or Gram-negative bacteria). We used Tenebrio molitor and methoprene, a JH analog (JHa) to explore these relationships. We tested the effect of methoprene on phenoloxidase activity (PO), an important component of humoral immunity in insects, and hemocyte number. Lyophilized Gram-positive Staphylococcus aureus or Gram-negative Escherichia coli were injected for the immune challenge. The results suggest that JH did not affect the proPO, PO activity, or hemocyte number of larvae. JH and immune challenge affected the immune response and consequently, affected adult developmental stage and sex. We propose that the influence of JH on the immune response depends on age, sex, the immune response parameter, and the immune challenge, which may explain the contrasting results about the role of JH in the insect immune response.
Collapse
Affiliation(s)
- Tania Amaro-Sánchez
- Departamento de Biología, División de Ciencias Naturales y Exactas, Universidad de Guanajuato, Campus Guanajuato, Noria Alta s/n, Colonia Noria Alta, 36050 Guanajuato, Mexico
| | - Gloria Ruiz-Guzmán
- Escuela Nacional de Estudios Superiores, Unidad Morelia, Universidad Nacional Autónoma de México, Antigua Carretera a Pátzcuaro No.8701, Col. Ex-Hacienda San José de la Huerta, 58190 Morelia, Michoacán, Mexico
| | - Salvador Hernández-Martínez
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62300 Cuernavaca, Morelos, Mexico
| | - Indrikis Krams
- Institute of Ecology and Earth Sciences, University of Tartu, Vanemuise 46, 51014 Tartu, Estonia; Department of Biotechnology, Daugavpils University, Daugavpils 5401, Latvia; Department of Zoology and Animal Ecology, Faculty of Biology, University of Latvia, Riga 1004, Latvia
| | - Markus J Rantala
- Department of Biology & Turku Brain and Mind Center, University of Turku, Turku, Finland
| | - Jorge Contreras-Garduño
- Escuela Nacional de Estudios Superiores, Unidad Morelia, Universidad Nacional Autónoma de México, Antigua Carretera a Pátzcuaro No.8701, Col. Ex-Hacienda San José de la Huerta, 58190 Morelia, Michoacán, Mexico.
| |
Collapse
|
23
|
Okamoto N, Watanabe A. Interorgan communication through peripherally derived peptide hormones in Drosophila. Fly (Austin) 2022; 16:152-176. [PMID: 35499154 PMCID: PMC9067537 DOI: 10.1080/19336934.2022.2061834] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
In multicellular organisms, endocrine factors such as hormones and cytokines regulate development and homoeostasis through communication between different organs. For understanding such interorgan communications through endocrine factors, the fruit fly Drosophila melanogaster serves as an excellent model system due to conservation of essential endocrine systems between flies and mammals and availability of powerful genetic tools. In Drosophila and other insects, functions of neuropeptides or peptide hormones from the central nervous system have been extensively studied. However, a series of recent studies conducted in Drosophila revealed that peptide hormones derived from peripheral tissues also play critical roles in regulating multiple biological processes, including growth, metabolism, reproduction, and behaviour. Here, we summarise recent advances in understanding target organs/tissues and functions of peripherally derived peptide hormones in Drosophila and describe how these hormones contribute to various biological events through interorgan communications.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Watanabe
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
24
|
Ahlawat N, Geeta Arun M, Maggu K, Jigisha, Singh A, Prasad NG. Drosophila melanogaster hosts coevolving with Pseudomonas entomophila pathogen show sex-specific patterns of local adaptation. BMC Ecol Evol 2022; 22:77. [PMID: 35717176 PMCID: PMC9206745 DOI: 10.1186/s12862-022-02031-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 06/10/2022] [Indexed: 11/18/2022] Open
Abstract
Background In spatially structured populations, local adaptation improves organisms’ fitness in their native environment. Hosts and pathogens can rapidly adapt to their local antagonist. Since males and females can differ in their immunocompetence, the patterns of local adaptation can be different between the sexes. However, there is little information about sex differences in local adaptation in host–pathogen systems. Results In the current study, we experimentally coevolved four different replicate populations of Drosophila melanogaster (host) and Pseudomonas entomophila (pathogen) along with appropriate controls. We used the four host–pathogen coevolution populations to investigate the occurrence of local adaptation separately in males and females of the coevolving hosts. We also assessed local adaptation in pathogens. We set up a reciprocal infection experiment where we infected each of the four coevolving hosts with their local pathogen or non-local pathogens from the other three replicate populations. We found that overall, male and female hosts had better survivorship when infected with local pathogens, indicating that they were locally adapted. Interestingly, males were more susceptible to non-local pathogens compared to females. In addition, we found no fecundity cost in females infected with either local or non-local pathogens. We found no evidence of local adaptation among the pathogens. Conclusion Our study showed sex-specific adaptation in the coevolving hosts where female hosts had a broader response against allopatric coevolving pathogens with no cost in fecundity. Thus, our results might suggest a novel mechanism that can maintain variation in susceptibility in spatially structured populations. Supplementary Information The online version contains supplementary material available at 10.1186/s12862-022-02031-8.
Collapse
|
25
|
Xing S, Deng D, wen W, Peng W. Functional transcriptome analyses of Drosophila suzukii midgut reveal mating-dependent reproductive plasticity in females. BMC Genomics 2022; 23:726. [PMID: 36284272 PMCID: PMC9598023 DOI: 10.1186/s12864-022-08962-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Insect females undergo a huge transition in energy homeostasis after mating to compensate for nutrient investment during reproduction. To manage with this shift in metabolism, mated females experience extensive morphological, behavioral and physiological changes, including increased food intake and altered digestive processes. However, the mechanisms by which the digestive system responds to mating in females remain barely characterized. Here we performed transcriptomic analysis of the main digestive organ, the midgut, to investigate how gene expression varies with female mating status in Drosophila suzukii, a destructive and invasive soft fruit pest. RESULTS We sequenced 15,275 unique genes with an average length of 1,467 bp. In total, 652 differentially expressed genes (DEGs) were detected between virgin and mated D. suzukii female midgut libraries. The DEGs were functionally annotated utilizing the GO and KEGG pathway annotation methods. Our results showed that the major GO terms associated with the DEGs from the virgin versus mated female midgut were largely appointed to the metabolic process, response to stimulus and immune system process. We obtained a mass of protein and lipid metabolism genes which were up-regulated and carbohydrate metabolism and immune-related genes which were down-regulated at different time points after mating in female midgut by qRT-PCR. These changes in metabolism and immunity may help supply the female with the nutrients and energy required to sustain egg production. CONCLUSION Our study characterizes the transcriptional mechanisms driven by mating in the D. suzukii female midgut. Identification and characterization of the DEGs between virgin and mated females midgut will not only be crucial to better understand molecular research related to intestine plasticity during reproduction, but may also provide abundant target genes for the development of effective and ecofriendly pest control strategies against this economically important species.
Collapse
Affiliation(s)
- Shisi Xing
- grid.411427.50000 0001 0089 3695Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, HunanInternational Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha, 410081 China
| | - Dan Deng
- grid.411427.50000 0001 0089 3695Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, HunanInternational Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha, 410081 China
| | - Wen wen
- grid.411427.50000 0001 0089 3695Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, HunanInternational Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha, 410081 China
| | - Wei Peng
- grid.411427.50000 0001 0089 3695Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, HunanInternational Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University, Changsha, 410081 China
| |
Collapse
|
26
|
Arch M, Vidal M, Koiffman R, Melkie ST, Cardona PJ. Drosophila melanogaster as a model to study innate immune memory. Front Microbiol 2022; 13:991678. [PMID: 36338030 PMCID: PMC9630750 DOI: 10.3389/fmicb.2022.991678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/03/2022] [Indexed: 09/12/2023] Open
Abstract
Over the last decades, research regarding innate immune responses has gained increasing importance. A growing body of evidence supports the notion that the innate arm of the immune system could show memory traits. Such traits are thought to be conserved throughout evolution and provide a survival advantage. Several models are available to study these mechanisms. Among them, we find the fruit fly, Drosophila melanogaster. This non-mammalian model has been widely used for innate immune research since it naturally lacks an adaptive response. Here, we aim to review the latest advances in the study of the memory mechanisms of the innate immune response using this animal model.
Collapse
Affiliation(s)
- Marta Arch
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Maria Vidal
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Romina Koiffman
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- UCBL, UnivLyon, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Solomon Tibebu Melkie
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- UCBL, UnivLyon, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Pere-Joan Cardona
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, Badalona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
27
|
Wang M, Wang Y, Chang M, Wang X, Shi Z, Raikhel AS, Zou Z. Ecdysone signaling mediates the trade-off between immunity and reproduction via suppression of amyloids in the mosquito Aedes aegypti. PLoS Pathog 2022; 18:e1010837. [PMID: 36137163 PMCID: PMC9531809 DOI: 10.1371/journal.ppat.1010837] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/04/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
The balance between immunity and reproduction is essential for many key physiological functions. We report that to maintain an optimal fertility, 20-hydroxyecdysone (20E) and the ecdysone receptor (EcR) downregulate the immune deficiency (IMD) pathway during the post blood meal phase (PBM) of the Aedes aegypti reproductive cycle. RNA interference-mediated depletion of EcR elicited an increased expression of the IMD pathway components, and these mosquitoes were more resistant to infection by Gram-negative bacteria. Moreover, 20E and EcR recruit Pirk-like, the mosquito ortholog of Drosophila melanogaster Pirk. CRISPR-Cas9 knockout of Pirk-like has shown that it represses the IMD pathway by interfering with IMD-mediated formation of amyloid aggregates. 20E and EcR disruption of the amyloid formation is pivotal for maintaining normal yolk protein production and fertility. Additionally, 20E and its receptor EcR directly induce Pirk-like to interfere with cRHIM-mediated formation of amyloid. Our study highlights the vital role of 20E in governing the trade-off between immunity and reproduction. Pirk-like might be a potential target for new methods to control mosquito reproduction and pathogen transmission.
Collapse
Affiliation(s)
- Mao Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yanhong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Mengmeng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xueli Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zuokun Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Alexander S. Raikhel
- Department of Entomology and Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
- * E-mail: (ASR); (ZZ)
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- * E-mail: (ASR); (ZZ)
| |
Collapse
|
28
|
Phurealipids, produced by the entomopathogenic bacteria, Photorhabdus, mimic juvenile hormone to suppress insect immunity and immature development. J Invertebr Pathol 2022; 193:107799. [PMID: 35850258 DOI: 10.1016/j.jip.2022.107799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022]
Abstract
Phurealipids (Photorhabdus urea lipids) are synthesized from Photorhabdus bacteria that are symbiotic to entomopathogenic nematodes. Their chemical structures are similar to that of juvenile hormone (JH) and have been suspected to mimic JH signaling in immunity and the development of insects. This study investigated the physiological roles of phurealipids with respect to their contribution to bacterial pathogenicity using four natural (HB13, HB69, HB416, and HB421) and one derivative (HB27) compound. First, phurealipids like JH suppressed insect immune responses. Overall, phurealipids showed JH like immunosuppressive behavior in a lepidopteran insect Spodoptera exigua larvae. More specifically, phurealipids significantly suppressed the hemocyte spreading behavior which is a key immune response upon immune challenge. Interestingly, the methyl urea derivatives (HB13, HB27, and HB69) were more potent than the unmethylated forms (HB416 and HB421). The inhibitory activity of phurealipids prevented the cellular immune response measured by hemocytic nodule formation in response to the bacterial challenge. Phurealipids also suppressed the expression of cecropin and gallerimycin, which are two highly inducible antimicrobial peptides, in S. exigua upon immune challenge. The immunosuppressive activity of the phurealipids significantly enhanced the bacterial pathogenicity of Bacillus thuringiensis against S. exigua. Second, phurealipids like JH prevented insect metamorphosis. Especially, the methylated urea derivatives of the phurealipids showed the JH-like function by inducing the expression of S. exigua Kr-h1, a transcriptional factor. At the pupal stage, exhibiting the lowest expression of Kr-h1, phurealipid treatments elevated the expression level of Kr-h1 and delayed the pupa-to-adult metamorphosis. These results suggest that phurealipids play crucial roles in Photorhabdus pathogenicity by suppressing host immune defenses and delaying host metamorphosis.
Collapse
|
29
|
Gordon KE, Wolfner MF, Lazzaro BP. A single mating is sufficient to induce persistent reduction of immune defense in mated female Drosophila melanogaster. JOURNAL OF INSECT PHYSIOLOGY 2022; 140:104414. [PMID: 35728669 PMCID: PMC10162487 DOI: 10.1016/j.jinsphys.2022.104414] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 05/07/2023]
Abstract
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.
Collapse
Affiliation(s)
- Kathleen E Gordon
- Field of Genetics, Genomics, and Development, Cornell University, Ithaca, NY 14853, USA; Department of Entomology, Cornell University, Ithaca, NY 14853, USA.
| | - Mariana F Wolfner
- Field of Genetics, Genomics, and Development, Cornell University, Ithaca, NY 14853, USA; Department of Entomology, Cornell University, Ithaca, NY 14853, USA
| | - Brian P Lazzaro
- Field of Genetics, Genomics, and Development, Cornell University, Ithaca, NY 14853, USA; Department of Entomology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
30
|
Gupta V, Frank AM, Matolka N, Lazzaro BP. Inherent constraints on a polyfunctional tissue lead to a reproduction-immunity tradeoff. BMC Biol 2022; 20:127. [PMID: 35655304 PMCID: PMC9161490 DOI: 10.1186/s12915-022-01328-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Single tissues can have multiple functions, which can result in constraints, impaired function, and tradeoffs. The insect fat body performs remarkably diverse functions including metabolic control, reproductive provisioning, and systemic immune responses. How polyfunctional tissues simultaneously execute multiple distinct physiological functions is generally unknown. Immunity and reproduction are observed to trade off in many organisms but the mechanistic basis for this tradeoff is also typically not known. Here we investigate constraints and trade-offs in the polyfunctional insect fat body. RESULTS Using single-nucleus sequencing, we determined that the Drosophila melanogaster fat body executes diverse basal functions with heterogenous cellular subpopulations. The size and identity of these subpopulations are remarkably stable between virgin and mated flies, as well as before and after infection. However, as an emergency function, the immune response engages the entire tissue and all cellular subpopulations produce induce expression of defense genes. We found that reproductively active females who were given bacterial infection exhibited signatures of ER stress and impaired capacity to synthesize new protein in response to infection, including decreased capacity to produce antimicrobial peptides. Transient provision of a reversible translation inhibitor to mated females prior to infection rescued general protein synthesis, specific production of antimicrobial peptides, and survival of infection. CONCLUSIONS The commonly observed tradeoff between reproduction and immunity appears to be driven, in D. melanogaster, by a failure of the fat body to be able to handle simultaneous protein translation demands of reproductive provisioning and immune defense. We suggest that inherent cellular limitations in tissues that perform multiple functions may provide a general explanation for the wide prevalence of physiological and evolutionary tradeoffs.
Collapse
Affiliation(s)
- Vanika Gupta
- Department of Entomology, Cornell University, Ithaca, NY, USA.
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, USA.
| | - Ashley M Frank
- Department of Entomology, Cornell University, Ithaca, NY, USA
| | - Nick Matolka
- Department of Entomology, Cornell University, Ithaca, NY, USA
| | - Brian P Lazzaro
- Department of Entomology, Cornell University, Ithaca, NY, USA.
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
31
|
Hopkins BR, Perry JC. The evolution of sex peptide: sexual conflict, cooperation, and coevolution. Biol Rev Camb Philos Soc 2022; 97:1426-1448. [PMID: 35249265 PMCID: PMC9256762 DOI: 10.1111/brv.12849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/17/2022]
Abstract
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.
Collapse
Affiliation(s)
- Ben R. Hopkins
- Department of Evolution and Ecology University of California – Davis One Shields Avenue Davis CA 95616 U.S.A
| | - Jennifer C. Perry
- School of Biological Sciences University of East Anglia Norwich NR4 7TJ U.K
| |
Collapse
|
32
|
Zhang X, Li S, Liu S. Juvenile Hormone Studies in Drosophila melanogaster. Front Physiol 2022; 12:785320. [PMID: 35222061 PMCID: PMC8867211 DOI: 10.3389/fphys.2021.785320] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/29/2021] [Indexed: 12/02/2022] Open
Abstract
In the field of insect endocrinology, juvenile hormone (JH) is one of the most wondrous entomological terms. As a unique sesquiterpenoid hormone produced and released by the endocrine gland, corpus allatum (CA), JH is a critical regulator in multiple developmental and physiological processes, such as metamorphosis, reproduction, and behavior. Benefited from the precise genetic interventions and simplicity, the fruit fly, Drosophila melanogaster, is an indispensable model in JH studies. This review is aimed to present the regulatory factors on JH biosynthesis and an overview of the regulatory roles of JH in Drosophila. The future directions of JH studies are also discussed, and a few hot spots are highlighted.
Collapse
Affiliation(s)
- Xiaoshuai Zhang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangmeiyuan R&D Center, South China Normal University, Meizhou, China
| | - Sheng Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangmeiyuan R&D Center, South China Normal University, Meizhou, China
| | - Suning Liu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangmeiyuan R&D Center, South China Normal University, Meizhou, China
| |
Collapse
|
33
|
Geeta Arun M, Agarwala A, Syed ZA, Jigisha, Kashyap M, Venkatesan S, Chechi TS, Gupta V, Prasad NG. Experimental evolution reveals sex-specific dominance for surviving bacterial infection in laboratory populations of Drosophila melanogaster. Evol Lett 2021; 5:657-671. [PMID: 34919096 PMCID: PMC8645198 DOI: 10.1002/evl3.259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023] Open
Abstract
Males and females are subjected to distinct kinds of selection pressures, often leading to the evolution of sex‐specific genetic architecture, an example being sex‐specific dominance. Sex‐specific dominance reversals (SSDRs), where alleles at sexually antagonistic loci are at least partially dominant in the sex they benefit, have been documented in Atlantic salmon, rainbow trout, and seed beetles. Another interesting feature of many sexually reproducing organisms is the asymmetric inheritance pattern of X chromosomes, which often leads to distinct evolutionary outcomes on X chromosomes compared to autosomes. Examples include the higher efficacy of sexually concordant selection on X chromosomes, and X chromosomes being more conducive to the maintenance of sexually antagonistic polymorphisms under certain conditions. Immunocompetence is a trait that has been extensively investigated for sexual dimorphism with growing evidence for sex‐specific or sexually antagonistic variation. X chromosomes have been shown to harbor substantial immunity‐related genetic variation in the fruit fly, Drosophila melanogaster. Here, using interpopulation crosses and cytogenetic cloning, we investigated sex‐specific dominance and the role of the X chromosome in improved postinfection survivorship of laboratory populations of D. melanogaster selected against pathogenic challenge by Pseudomonas entomophila. We could not detect any contribution of the X chromosome to the evolved immunocompetence of our selected populations, as well as to within‐population variation in immunocompetence. However, we found strong evidence of sex‐specific dominance related to surviving bacterial infection. Our results indicate that alleles that confer a survival advantage to the selected populations are, on average, partially dominant in females but partially recessive in males. This could also imply an SSDR for overall fitness, given the putative evidence for sexually antagonistic selection affecting immunocompetence in Drosophila melanogaster. We also highlight sex‐specific dominance as a potential mechanism of sex differences in immunocompetence, with population‐level sex differences primarily driven by sex differences in heterozygotes.
Collapse
Affiliation(s)
- Manas Geeta Arun
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India
| | - Amisha Agarwala
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India.,Department of Biology Syracuse University Syracuse New York 13210
| | - Zeeshan Ali Syed
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India.,Department of Biology Syracuse University Syracuse New York 13210
| | - Jigisha
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India
| | - Mayank Kashyap
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India
| | - Saudamini Venkatesan
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India.,Institute of Evolutionary Biology, School of Biological Sciences, King's Buildings University of Edinburgh Edinburgh EH9 3FL United Kingdom
| | - Tejinder Singh Chechi
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India
| | - Vanika Gupta
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India.,Department of Entomology Cornell University Ithaca New York 14853
| | - Nagaraj Guru Prasad
- Department of Biological Sciences Indian Institute of Science Education and Research Mohali Mohali 140306 India
| |
Collapse
|
34
|
Rodrigues MA, Merckelbach A, Durmaz E, Kerdaffrec E, Flatt T. Transcriptomic evidence for a trade-off between germline proliferation and immunity in Drosophila. Evol Lett 2021; 5:644-656. [PMID: 34917403 PMCID: PMC8645197 DOI: 10.1002/evl3.261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 11/08/2022] Open
Abstract
Life-history theory posits that investment into reproduction might occur at the expense of investment into somatic maintenance, including immune function. If so, reduced or curtailed reproductive effort might be expected to increase immunity. In support of this notion, work in Caenorhabditis elegans has shown that worms lacking a germline exhibit improved immunity, but whether the antagonistic relation between germline proliferation and immunity also holds for other organisms is less well understood. Here, we report that transgenic ablation of germ cells in late development or early adulthood in Drosophila melanogaster causes elevated baseline expression and increased induction of Toll and Imd immune genes upon bacterial infection, as compared to fertile flies with an intact germline. We also identify immune genes whose expression after infection differs between fertile and germline-less flies in a manner that is conditional on their mating status. We conclude that germline activity strongly impedes the expression and inducibility of immune genes and that this physiological trade-off might be evolutionarily conserved.
Collapse
Affiliation(s)
| | | | - Esra Durmaz
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| | - Envel Kerdaffrec
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| | - Thomas Flatt
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| |
Collapse
|
35
|
Abstract
Horizontal gene transfer (HGT) is a driving force of microbial evolution. The gut of animals acts as a potent reservoir for the lateral transfer of virulence, fitness, and antimicrobial resistance genes through plasmids. Reduced-complexity models for the examination of host-microbe interactions involved in plasmid transfer are greatly desired. Thus, this study identifies the use of Drosophila melanogaster as a model organism for the conjugation of plasmids of various incompatibility groups in the gut. Enterobacteriaceae conjugation pairs were identified in vitro and used for oral inoculation of the Drosophila gut. Flies were enumerated for the donor, recipient, and transconjugant populations. Each donor-recipient pair was observed to persist in fly guts for the duration of the experiment. Gut concentrations of the donors and recipients were significantly different between male and female flies, with females generally demonstrating increased concentrations. Furthermore, host genetics significantly altered the concentrations of donors and recipients. However, transconjugant concentrations were not affected by host sex or genetics and were detected only in the IncPε and IncI1 plasmid groups. This study demonstrates Drosophila melanogaster as a model for gut-mediated plasmid transfer. IMPORTANCE Microbial evolution in the gut of animals due to horizontal gene transfer (HGT) is of significant interest for microbial evolution as well as within the context of human and animal health. Microbial populations evolve within the host, and factors from the bacteria and host interact to regulate this evolution. However, little is currently known about how host and bacterial factors regulate plasmid-mediated HGT in the gut. This study demonstrates the use of Drosophila and the roles of sexual dimorphism as well as plasmid incompatibility groups in HGT in the gut.
Collapse
|
36
|
Imrie RM, Roberts KE, Longdon B. Between virus correlations in the outcome of infection across host species: Evidence of virus by host species interactions. Evol Lett 2021; 5:472-483. [PMID: 34621534 PMCID: PMC8484721 DOI: 10.1002/evl3.247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Virus host shifts are a major source of outbreaks and emerging infectious diseases, and predicting the outcome of novel host and virus interactions remains a key challenge for virus research. The evolutionary relationships between host species can explain variation in transmission rates, virulence, and virus community composition between hosts, but it is unclear if correlations exist between related viruses in infection traits across novel hosts. Here, we measure correlations in viral load of four Cripavirus isolates across experimental infections of 45 Drosophilidae host species. We find positive correlations between every pair of viruses tested, suggesting that some host clades show broad susceptibility and could act as reservoirs and donors for certain types of viruses. Additionally, we find evidence of virus by host species interactions, highlighting the importance of both host and virus traits in determining the outcome of virus host shifts. Of the four viruses tested here, those that were more closely related tended to be more strongly correlated, providing tentative evidence that virus evolutionary relatedness may be a useful proxy for determining the likelihood of novel virus emergence, which warrants further research.
Collapse
Affiliation(s)
- Ryan M. Imrie
- Centre for Ecology and Conservation, Biosciences, College of Life and Environmental SciencesUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| | - Katherine E. Roberts
- Centre for Ecology and Conservation, Biosciences, College of Life and Environmental SciencesUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| | - Ben Longdon
- Centre for Ecology and Conservation, Biosciences, College of Life and Environmental SciencesUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| |
Collapse
|
37
|
Luo W, Liu S, Zhang W, Yang L, Huang J, Zhou S, Feng Q, Palli SR, Wang J, Roth S, Li S. Juvenile hormone signaling promotes ovulation and maintains egg shape by inducing expression of extracellular matrix genes. Proc Natl Acad Sci U S A 2021; 118:e2104461118. [PMID: 34544864 PMCID: PMC8488625 DOI: 10.1073/pnas.2104461118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 11/18/2022] Open
Abstract
It is well documented that the juvenile hormone (JH) can function as a gonadotropic hormone that stimulates vitellogenesis by activating the production and uptake of vitellogenin in insects. Here, we describe a phenotype associated with mutations in the Drosophila JH receptor genes, Met and Gce: the accumulation of mature eggs with reduced egg length in the ovary. JH signaling is mainly activated in ovarian muscle cells and induces laminin gene expression in these cells. Meanwhile, JH signaling induces collagen IV gene expression in the adult fat body, from which collagen IV is secreted and deposited onto the ovarian muscles. Laminin locally and collagen IV remotely contribute to the assembly of ovarian muscle extracellular matrix (ECM); moreover, the ECM components are indispensable for ovarian muscle contraction. Furthermore, ovarian muscle contraction externally generates a mechanical force to promote ovulation and maintain egg shape. This work reveals an important mechanism for JH-regulated insect reproduction.
Collapse
Affiliation(s)
- Wei Luo
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| | - Suning Liu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China;
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| | - Wenqiang Zhang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Liu Yang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jianhua Huang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Shutang Zhou
- Key Laboratory of Plant Stress Biology, State Key Laboratory of Cotton Biology, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Qili Feng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Subba Reddy Palli
- Department of Entomology, College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546
| | - Jian Wang
- Department of Entomology, University of Maryland, College Park, MD 20742
| | - Siegfried Roth
- Institute for Zoology, University of Cologne, D-50674 Cologne, Germany
| | - Sheng Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China;
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| |
Collapse
|
38
|
Reitmayer CM, Pathak AK, Harrington LC, Brindley MA, Cator LJ, Murdock CC. Sex, age, and parental harmonic convergence behavior affect the immune performance of Aedes aegypti offspring. Commun Biol 2021; 4:723. [PMID: 34117363 PMCID: PMC8196008 DOI: 10.1038/s42003-021-02236-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Harmonic convergence is a potential cue, female mosquitoes use to choose male mates. However, very little is known about the benefits this choice confers to offspring performance. Using Aedes aegypti (an important vector of human disease), we investigated whether offspring of converging parental pairs showed differences in immune competence compared to offspring derived from non-converging parental pairs. Here we show that harmonic convergence, along with several other interacting factors (sex, age, reproductive, and physiological status), significantly shaped offspring immune responses (melanization and response to a bacterial challenge). Harmonic convergence had a stronger effect on the immune response of male offspring than on female offspring. Further, female offspring from converging parental pairs disseminated dengue virus more quickly than offspring derived from non-converging parental pairs. Our results provide insight into a wide range of selective pressures shaping mosquito immune function and could have important implications for disease transmission and control.
Collapse
Affiliation(s)
- Christine M Reitmayer
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Center for Tropical and Global Emerging Diseases, University of Georgia, Athens, GA, USA
- The Pirbright Institute, Pirbright, Surrey, UK
| | - Ashutosh K Pathak
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Center for Tropical and Global Emerging Diseases, University of Georgia, Athens, GA, USA
| | - Laura C Harrington
- Department of Entomology, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
- Northeast Center for Excellence for Vector-borne Disease Research, Ithaca, NY, USA
| | - Melinda A Brindley
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Lauren J Cator
- Department of Life Sciences, Imperial College London, Ascot, UK
| | - Courtney C Murdock
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
- Center for Tropical and Global Emerging Diseases, University of Georgia, Athens, GA, USA.
- Department of Entomology, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA.
- Northeast Center for Excellence for Vector-borne Disease Research, Ithaca, NY, USA.
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
- Odum School of Ecology, University of Georgia, Athens, GA, USA.
- Center for Ecology of Infectious Diseases, Odum School of Ecology, University of Georgia, Athens, GA, USA.
- Riverbasin Center, Odum School of Ecology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
39
|
Drosophila melanogaster sex peptide regulates mated female midgut morphology and physiology. Proc Natl Acad Sci U S A 2021; 118:2018112118. [PMID: 33443193 DOI: 10.1073/pnas.2018112118] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
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.
Collapse
|
40
|
Schlamp F, Delbare SYN, Early AM, Wells MT, Basu S, Clark AG. Dense time-course gene expression profiling of the Drosophila melanogaster innate immune response. BMC Genomics 2021; 22:304. [PMID: 33902461 PMCID: PMC8074482 DOI: 10.1186/s12864-021-07593-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immune responses need to be initiated rapidly, and maintained as needed, to prevent establishment and growth of infections. At the same time, resources need to be balanced with other physiological processes. On the level of transcription, studies have shown that this balancing act is reflected in tight control of the initiation kinetics and shutdown dynamics of specific immune genes. RESULTS To investigate genome-wide expression dynamics and trade-offs after infection at a high temporal resolution, we performed an RNA-seq time course on D. melanogaster with 20 time points post Imd stimulation. A combination of methods, including spline fitting, cluster analysis, and Granger causality inference, allowed detailed dissection of expression profiles, lead-lag interactions, and functional annotation of genes through guilt-by-association. We identified Imd-responsive genes and co-expressed, less well characterized genes, with an immediate-early response and sustained up-regulation up to 5 days after stimulation. In contrast, stress response and Toll-responsive genes, among which were Bomanins, demonstrated early and transient responses. We further observed a strong trade-off with metabolic genes, which strikingly recovered to pre-infection levels before the immune response was fully resolved. CONCLUSIONS This high-dimensional dataset enabled the comprehensive study of immune response dynamics through the parallel application of multiple temporal data analysis methods. The well annotated data set should also serve as a useful resource for further investigation of the D. melanogaster innate immune response, and for the development of methods for analysis of a post-stress transcriptional response time-series at whole-genome scale.
Collapse
Affiliation(s)
- Florencia Schlamp
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| | | | - Angela M Early
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Martin T Wells
- Statistics and Data Science, Cornell University, Ithaca, NY, USA
| | - Sumanta Basu
- Statistics and Data Science, Cornell University, Ithaca, NY, USA.
| | - Andrew G Clark
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
- Statistics and Data Science, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
41
|
Ahmed-Braimah YH, Wolfner MF, Clark AG. Differences in Postmating Transcriptional Responses between Conspecific and Heterospecific Matings in Drosophila. Mol Biol Evol 2021; 38:986-999. [PMID: 33035303 PMCID: PMC7947788 DOI: 10.1093/molbev/msaa264] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In many animal species, females undergo physiological and behavioral changes after mating. Some of these changes are driven by male-derived seminal fluid proteins and are critical for fertilization success. Unfortunately, our understanding of the molecular interplay between female and male reproductive proteins remains inadequate. Here, we analyze the postmating response in a Drosophila species that has evolved strong gametic incompatibility with its sister species; Drosophila novamexicana females produce only ∼1% fertilized eggs in crosses with Drosophila americana males, compared to ∼98% produced in within-species crosses. This incompatibility is likely caused by mismatched male and female reproductive molecules. In this study, we use short-read RNA sequencing to examine the evolutionary dynamics of female reproductive genes and the postmating transcriptome response in crosses within and between species. First, we found that most female reproductive tract genes are slow-evolving compared to the genome average. Second, postmating responses in con- and heterospecific matings are largely congruent, but heterospecific matings induce expression of additional stress-response genes. Some of those are immunity genes that are activated by the Imd pathway. We also identify several genes in the JAK/STAT signaling pathway that are induced in heterospecific, but not conspecific mating. While this immune response was most pronounced in the female reproductive tract, we also detect it in the female head and ovaries. These results show that the female's postmating transcriptome-level response is determined in part by the genotype of the male, and that divergence in male reproductive genes and/or traits can have immunogenic effects on females.
Collapse
Affiliation(s)
- Yasir H Ahmed-Braimah
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 13850
- Department of Biology, Syracuse University, Syracuse, NY 13244
| | - Mariana F Wolfner
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 13850
| | - Andrew G Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 13850
| |
Collapse
|
42
|
Trade-offs between sperm viability and immune protein expression in honey bee queens (Apis mellifera). Commun Biol 2021; 4:48. [PMID: 33420325 PMCID: PMC7794525 DOI: 10.1038/s42003-020-01586-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Queens of many social hymenoptera keep sperm alive within their specialized storage organ, the spermatheca, for years, defying the typical trade-off between lifespan and reproduction. However, whether honey bee (Apis mellifera) queens experience a trade-off between reproduction and immunity is unknown, and the biochemical processes underlying sperm viability are poorly understood. Here, we survey quality metrics and viral loads of honey bee queens from nine genetic sources. Queens rated as 'failed' by beekeepers had lower sperm viability, fewer sperm, and higher levels of sacbrood virus and black queen cell virus. Quantitative proteomics on N = 123 spermathecal fluid samples shows, after accounting for sperm count, health status, and apiary effects, five spermathecal fluid proteins significantly correlating with sperm viability: odorant binding protein (OBP)14, lysozyme, serpin 88Ea, artichoke, and heat-shock protein (HSP)10. The significant negative correlation of lysozyme-a conserved immune effector-with sperm viability is consistent with a reproduction vs. immunity trade-off in honey bee queens.
Collapse
|
43
|
Chang MM, Wang YH, Yang QT, Wang XL, Wang M, Raikhel AS, Zou Z. Regulation of antimicrobial peptides by juvenile hormone and its receptor, Methoprene-tolerant, in the mosquito Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 128:103509. [PMID: 33264664 DOI: 10.1016/j.ibmb.2020.103509] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/09/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
The trade-off between reproduction and immunity has been established for a number of insect species. However, the regulatory mechanisms governing this event is not well understood. In the mosquito Aedes aegypti, the vector of dangerous human arboviral diseases, juvenile hormone (JH) is required for the female post-eclosion development and reproductive maturation. In this study, we have revealed the JH negative effect on the expression of immunity-related genes, such as antimicrobial peptides (AMPs), during the post-eclosion phase of the female mosquito gonadotrophic reproductive cycle. Mosquitoes treated with JH became more sensitive to microbial infection. Mosquitoes subjected to the RNA interference knockdown (RNAi) of the JH receptor, Methoprene-tolerant (Met), showed increased expression of several AMP genes. Met binds to the E-box-like recognition motifs in the regulatory region of the diptericin (Dpt) gene, indicating that JH can suppress the Dpt gene expression through its receptor Met. Hence, JH is involved in the modulation of immune responses during the post-eclosion phase of reproduction. The RNAi knockdown of the peptidoglycan recognition protein (PGRP-LC) led to a significant reduction of the Dpt transcript level, indicating the PGRP-LC activating role on this AMP gene. Thus, Dpt appeared to be under the dual regulation of both the JH and the immune deficiency (IMD) signaling pathways. Our study provides a better understanding of how JH regulates insect immunity in adult mosquitoes.
Collapse
Affiliation(s)
- Meng-Meng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan-Hong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Tai Yang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue-Li Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mao Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Alexander S Raikhel
- Department of Entomology and Institute for Integrative Genome Biology, University of California, Riverside, CA, 92521, USA
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, 311300, China.
| |
Collapse
|
44
|
Naim N, Amrit FRG, McClendon TB, Yanowitz JL, Ghazi A. The molecular tug of war between immunity and fertility: Emergence of conserved signaling pathways and regulatory mechanisms. Bioessays 2020; 42:e2000103. [PMID: 33169418 DOI: 10.1002/bies.202000103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
Reproduction and immunity are energy intensive, intimately linked processes in most organisms. In women, pregnancy is associated with widespread immunological adaptations that alter immunity to many diseases, whereas, immune dysfunction has emerged as a major cause for infertility in both men and women. Deciphering the molecular bases of this dynamic association is inherently challenging in mammals. This relationship has been traditionally studied in fast-living, invertebrate species, often in the context of resource allocation between life history traits. More recently, these studies have advanced our understanding of the mechanistic underpinnings of the immunity-fertility dialogue. Here, we review the molecular connections between reproduction and immunity from the perspective of human pregnancy to mechanistic discoveries in laboratory organisms. We focus particularly on recent invertebrate studies identifying conserved signaling pathways and transcription factors that regulate resource allocation and shape the balance between reproductive status and immune health.
Collapse
Affiliation(s)
- Nikki Naim
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francis R G Amrit
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - T Brooke McClendon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Judith L Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Arjumand Ghazi
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
45
|
Shpigler HY, Herb B, Drnevich J, Band M, Robinson GE, Bloch G. Juvenile hormone regulates brain-reproduction tradeoff in bumble bees but not in honey bees. Horm Behav 2020; 126:104844. [PMID: 32860832 DOI: 10.1016/j.yhbeh.2020.104844] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022]
Abstract
Gonadotropic hormones coordinate processes in diverse tissues regulating animal reproductive physiology and behavior. Juvenile hormone (JH) is the ancient and most common gonadotropin in insects, but not in advanced eusocial honey bees and some ants. To start probing the evolutionary basis of this change, we combined endocrine manipulations, transcriptomics, and behavioral analyses to study JH regulated processes in a bumble bee showing a relatively simple level of eusociality. We found that in worker fat body, more JH-regulated genes were up- rather than down-regulated, and enriched for metabolic and biosynthetic pathways. This transcriptomic pattern is consistent with earlier evidence that JH is the major gonadotropin in bumble bees. In the brain, more JH-regulated genes were down- rather than up-regulated and enriched for protein turnover pathways. Brain ribosomal protein gene expression shows a similar trend of downregulation in dominant workers, which naturally have high JH titers. In other species, similar downregulation of protein turnover is found in aging brains or under stress, associated with compromised long-term memory and health. These findings suggest a previously unknown gonadotropin-mediated tradeoff. Analysis of published data reveals no such downregulation of protein turnover pathways in the brain of honey bee workers, which exhibit more complex eusociality and in which JH is not a gonadotropin but rather regulates division of labor. These results suggest that the evolution of complex eusociality in honey bees was associated with modifications in hormonal signalling supporting extended and extremely high fertility while reducing the ancient costs of high gonadotropin titers to the brain.
Collapse
Affiliation(s)
- Hagai Y Shpigler
- Department of Ecology, Evolution, and Behavior, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Brian Herb
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jenny Drnevich
- High-Performance Biological Computing Group, Roy J Carver Biotechnology Center, The University of Illinois at Urbana-Champaign, USA
| | - Mark Band
- Roy J Carver Biotechnology Center, The University of Illinois at Urbana-Champaign, USA; Institute of Evolution, University of Haifa, Israel
| | - Gene E Robinson
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA; Entomology Department, The University of Illinois at Urbana-Champaign, USA; Neuroscience Program, The University of Illinois at Urbana-Champaign, USA
| | - Guy Bloch
- Department of Ecology, Evolution, and Behavior, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
46
|
Interactions between the microbiome and mating influence the female's transcriptional profile in Drosophila melanogaster. Sci Rep 2020; 10:18168. [PMID: 33097776 PMCID: PMC7584617 DOI: 10.1038/s41598-020-75156-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Drosophila melanogaster females undergo a variety of post-mating changes that influence their activity, feeding behavior, metabolism, egg production and gene expression. These changes are induced either by mating itself or by sperm or seminal fluid proteins. In addition, studies have shown that axenic females-those lacking a microbiome-have altered fecundity compared to females with a microbiome, and that the microbiome of the female's mate can influence reproductive success. However, the extent to which post-mating changes in transcript abundance are affected by microbiome state is not well-characterized. Here we investigated fecundity and the post-mating transcript abundance profile of axenic or control females after mating with either axenic or control males. We observed interactions between the female's microbiome and her mating status: transcripts of genes involved in reproduction and genes with neuronal functions were differentially abundant depending on the females' microbiome status, but only in mated females. In addition, immunity genes showed varied responses to either the microbiome, mating, or a combination of those two factors. We further observed that the male's microbiome status influences the fecundity of both control and axenic females, while only influencing the transcriptional profile of axenic females. Our results indicate that the microbiome plays a vital role in the post-mating switch of the female's transcriptome.
Collapse
|
47
|
Nunes C, Sucena É, Koyama T. Endocrine regulation of immunity in insects. FEBS J 2020; 288:3928-3947. [PMID: 33021015 DOI: 10.1111/febs.15581] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Organisms have constant contact with potentially harmful agents that can compromise their fitness. However, most of the times these agents fail to cause serious disease by virtue of the rapid and efficient immune responses elicited in the host that can range from behavioural adaptations to immune system triggering. The immune system of insects does not comprise the adaptive arm, making it less complex than that of vertebrates, but key aspects of the activation and regulation of innate immunity are conserved across different phyla. This is the case for the hormonal regulation of immunity as a part of the broad organismal responses to external conditions under different internal states. In insects, depending on the physiological circumstances, distinct hormones either enhance or suppress the immune response integrating individual (and often collective) responses physiologically and behaviourally. In this review, we provide an overview of our current knowledge on the endocrine regulation of immunity in insects, its mechanisms and implications on metabolic adaptation and behaviour. We highlight the importance of this multilayered regulation of immunity in survival and reproduction (fitness) and its dependence on the hormonal integration with other mechanisms and life-history traits.
Collapse
Affiliation(s)
| | - Élio Sucena
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Biwot JC, Zhang HB, Liu C, Qiao JX, Yu XQ, Wang YF. Wolbachia-induced expression of kenny gene in testes affects male fertility in Drosophila melanogaster. INSECT SCIENCE 2020; 27:869-882. [PMID: 31617302 DOI: 10.1111/1744-7917.12730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 09/18/2019] [Accepted: 09/30/2019] [Indexed: 06/10/2023]
Abstract
Wolbachia are Gram-negative endosymbionts that are known to cause embryonic lethality when infected male insects mate with uninfected females or with females carrying a different strain of Wolbachia, a situation characterized as cytoplasmic incompatibility (CI). However, the mechanism of CI is not yet fully understood, although recent studies on Drosophila melanogaster have achieved great progress. Here, we found that Wolbachia infection caused changes in the expressions of several immunity-related genes, including significant upregulation of kenny (key), in the testes of D. melanogaster. Overexpression of key in fly testes led to a significant decrease in egg hatch rates when these flies mate with wild-type females. Wolbachia-infected females could rescue this embryonic lethality. Furthermore, in key overexpressing testes terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling signal was significantly stronger than in the control testes, and the level of reactive oxygen species was significantly increased. Overexpression of key also resulted in alterations of some other immunity-related gene expressions, including the downregulation of Zn72D. Knockdown of Zn72D in fly testes also led to a significant decrease in egg hatch rates. These results suggest that Wolbachia might induce the defect in male host fertility by immunity-related pathways and thus cause an oxidative damage and cell death in male testes.
Collapse
Affiliation(s)
- John C Biwot
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Hua-Bao Zhang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Chen Liu
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Jun-Xue Qiao
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Xiao-Qiang Yu
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Yu-Feng Wang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| |
Collapse
|
49
|
Wei J, Gao H, Yang Y, Liu H, Yu H, Chen Z, Dong B. Seasonal dynamics and starvation impact on the gut microbiome of urochordate ascidian Halocynthia roretzi. Anim Microbiome 2020; 2:30. [PMID: 33499981 PMCID: PMC7807810 DOI: 10.1186/s42523-020-00048-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/05/2020] [Indexed: 02/02/2023] Open
Abstract
Background Gut microbiota plays important roles in host animal metabolism, homeostasis and environmental adaptation. However, the interplay between the gut microbiome and urochordate ascidian, the most closet relative of vertebrate, remains less explored. In this study, we characterized the gut microbial communities of urochordate ascidian (Halocynthia roretzi) across the changes of season and starvation stress using a comprehensive set of omic approaches including 16S rRNA gene amplicon sequencing, shotgun metagenomics, metabolomic profiling, and transcriptome sequencing. Results The 16S rRNA gene amplicon profiling revealed that ascidians harbor indigenous gut microbiota distinctly different to the marine microbial community and significant variations in composition and abundance of gut bacteria, with predominant bacterial orders representing each season. Depressed alpha-diversities of gut microbiota were observed across starvation stress when compared to the communities in aquafarm condition. Synechococcales involving photosynthesis and its related biosynthesis was reduced in abundance while the enrichments of Xanthomonadales and Legionellales may facilitate bile acid biosynthesis during starvation. Metabolomics analysis found that long chain fatty acids, linolenic acid, cyanoamino acid, and pigments derived from gut bacteria were upregulated, suggesting a beneficial contribution of the gut microbiome to the ascidian under starvation stress. Conclusions Our findings revealed seasonal variation of ascidian gut microbiota. Defense and energy-associated metabolites derived from gut microbiome may provide an adaptive interplay between gut microbiome and ascidian host that maintains a beneficial metabolic system across season and starvation stress. The diversity-generating metabolisms from both microbiota and host might lead to the co-evolution and environmental adaptation. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Jiankai Wei
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Hongwei Gao
- Technology Center of Qingdao Customs, Qingdao, 266002, China
| | - Yang Yang
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Haiming Liu
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Haiyan Yu
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Zigui Chen
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Bo Dong
- Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China. .,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China. .,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
50
|
Ernst DA, Fitak RR, Schmidt M, Derby CD, Johnsen S, Lohmann KJ. Pulse magnetization elicits differential gene expression in the central nervous system of the Caribbean spiny lobster, Panulirus argus. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2020; 206:725-742. [PMID: 32607762 DOI: 10.1007/s00359-020-01433-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 05/18/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022]
Abstract
Diverse animals use Earth's magnetic field to guide their movements, but the neural and molecular mechanisms underlying the magnetic sense remain enigmatic. One hypothesis is that particles of the mineral magnetite (Fe3O4) provide the basis of magnetoreception. Here we examined gene expression in the central nervous system of a magnetically sensitive invertebrate, the Caribbean spiny lobster (Panulirus argus), after applying a magnetic pulse known to alter magnetic orientation behavior. Numerous genes were differentially expressed in response to the pulse, including 647 in the brain, 1256 in the subesophageal ganglion, and 712 in the thoracic ganglia. Many such genes encode proteins linked to iron regulation, oxidative stress, and immune response, consistent with possible impacts of a magnetic pulse on magnetite-based magnetoreceptors. Additionally, however, altered expression also occurred for numerous genes with no apparent link to magnetoreception, including genes encoding proteins linked to photoreception, carbohydrate and hormone metabolism, and other physiological processes. Overall, the results are consistent with the magnetite hypothesis of magnetoreception, yet also reveal that in spiny lobsters, a strong pulse altered expression of > 10% of all expressed genes, including many seemingly unrelated to sensory processes. Thus, caution is required when interpreting the effects of magnetic pulses on animal behavior.
Collapse
Affiliation(s)
- David A Ernst
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Department of Biological Sciences, University of Arkansas, Fayetteville, AR, 72701, USA.
| | - Robert R Fitak
- Genomics and Bioinformatics Cluster, Department of Biology, University of Central Florida, Orlando, FL, 32816, USA
| | - Manfred Schmidt
- Neuroscience Institute and Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Charles D Derby
- Neuroscience Institute and Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Sönke Johnsen
- Department of Biology, Duke University, Durham, NC, 27708, USA
| | - Kenneth J Lohmann
- Department of Biology, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|