1
|
Kumar S, D'Souza LC, Shaikh FH, Rathor P, Ratnasekhar CH, Sharma A. Multigenerational immunotoxicity assessment: A three-generation study in Drosophila melanogaster upon developmental exposure to triclosan. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 370:125860. [PMID: 39954761 DOI: 10.1016/j.envpol.2025.125860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Triclosan (TCS) is widely used as an antibacterial agent, nevertheless, its presence in different environmental matrices and its persistent environmental nature pose a significant threat to the organism, including humans. Numerous studies showed that TCS exposure could lead to multiple toxicities, including immune dysfunction. However, whether parental TCS exposure could impair the offspring's immune response remains limited. Maintaining the immune homeostasis is imperative to neutralize the pathogen and crucial for tissue repair and the organism's survival. Thus, this study aimed to assess the multigenerational immune response of TCS using Drosophila melanogaster. TCS was administered to organisms (1.0, 10, and 100.0 μg/mL) over three generations during their developing phases, and its effect on the immunological response of the unexposed progeny was evaluated. Total circulatory hemocyte (immune cells) count, crystal cell count, phagocytic activity, clotting time, gene expression related to immune response and epigenetics, ROS generation, and cell death were assessed in the offspring. A concentration-dependent decline in total hemocytes, crystal cells, phagocytic activity, and increased clotting time in the subsequent generations was observed. Furthermore, parental TCS exposure enhanced the ROS levels, induced cell death, and altered the expression of antimicrobial peptides drosomycin, diptericin, and inflammatory genes upd1, upd2, and upd3, in the offspring's hemocytes across successive generations. The upregulation of reaper hid, and grim suggests that TCS promotes apoptotic death in the offspring's hemocytes. Notably, the increased mRNA expression of epigenetic regulators dnmt2 and g9a in the hemocytes of the offspring indicates epigenetic modifications. Further, we also observed that the antioxidant N-acetylcysteine (NAC) supplementation to the parents alleviated TCS toxicity and improved immunological functions in the progeny, indicating the role of ROS in the TCS-induced multigenerational immune toxicity. This finding provides valuable insights into the potential immune risk of prenatal TCS exposure to their offspring in the higher organism.
Collapse
Affiliation(s)
- Sandeep Kumar
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Leonard Clinton D'Souza
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Faiz Hanif Shaikh
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Priya Rathor
- Metabolomics Lab, Council of Scientific and Industrial Research (CSIR)-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - C H Ratnasekhar
- Metabolomics Lab, Council of Scientific and Industrial Research (CSIR)-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Anurag Sharma
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Department of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
2
|
Perdomo HD, Khorramnejad A, Cham NM, Kropf A, Sogliani D, Bonizzoni M. Prolonged exposure to heat enhances mosquito tolerance to viral infection. Commun Biol 2025; 8:168. [PMID: 39901025 PMCID: PMC11790978 DOI: 10.1038/s42003-025-07617-8] [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: 09/27/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
How and to what extent mosquito-virus interaction is influenced by climate change is a complex question of ecological and epidemiological relevance. We worked at the intersection between thermal biology and vector immunology and studied shifts in tolerance and resistance to the cell fusing agent virus (CFAV), a prominent component of the mosquito virome known to contribute to shaping mosquito vector competence, in warm-acclimated and warm-evolved Aedes albopictus mosquitoes. We show that the length of the thermal challenge influences the outcome of the infection with warm-evolved mosquitoes being more tolerant to CFAV infection, while warm-acclimated mosquitoes being more resistant and suffering from extensive fitness costs. These results highlight the importance of considering fluctuations in vector immunity in relation to the length of a thermal challenge to understand natural variation in vector response to viruses and frame realistic transmission models.
Collapse
Affiliation(s)
- Hugo D Perdomo
- Department of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Ayda Khorramnejad
- Department of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Nfamara M Cham
- Department of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Alida Kropf
- Department of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Davide Sogliani
- Department of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy
| | - Mariangela Bonizzoni
- Department of Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100, Pavia, Italy.
| |
Collapse
|
3
|
Lai Y, Wang S. Epigenetic Regulation in Insect-Microbe Interactions. ANNUAL REVIEW OF ENTOMOLOGY 2025; 70:293-311. [PMID: 39374433 DOI: 10.1146/annurev-ento-022724-010640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Insects have evolved diverse interactions with a variety of microbes, such as pathogenic fungi, bacteria, and viruses. The immune responses of insect hosts, along with the dynamic infection process of microbes in response to the changing host environment and defenses, require rapid and fine-tuned regulation of gene expression programs. Epigenetic mechanisms, including DNA methylation, histone modifications, and noncoding RNA regulation, play important roles in regulating the expression of genes involved in insect immunity and microbial pathogenicity. This review highlights recent discoveries and insights into epigenetic regulatory mechanisms that modulate insect-microbe interactions. A deeper understanding of these regulatory mechanisms underlying insect-microbe interactions holds promise for the development of novel strategies for biological control of insect pests and mitigation of vector-borne diseases.
Collapse
Affiliation(s)
- Yiling Lai
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, China;
| | - Sibao Wang
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, China;
| |
Collapse
|
4
|
Jia D, Luo G, Guan H, Yu T, Sun X, Du Y, Wang Y, Chen H, Wei T. Arboviruses antagonize insect Toll antiviral immune signaling to facilitate the coexistence of viruses with their vectors. PLoS Pathog 2024; 20:e1012318. [PMID: 38865374 PMCID: PMC11198909 DOI: 10.1371/journal.ppat.1012318] [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: 01/24/2024] [Revised: 06/25/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
Many plant arboviruses are persistently transmitted by piercing-sucking insect vectors. However, it remains largely unknown how conserved insect Toll immune response exerts antiviral activity and how plant viruses antagonize it to facilitate persistent viral transmission. Here, we discover that southern rice black-streaked dwarf virus (SRBSDV), a devastating planthopper-transmitted rice reovirus, activates the upstream Toll receptors expression but suppresses the downstream MyD88-Dorsal-defensin cascade, resulting in the attenuation of insect Toll immune response. Toll pathway-induced the small antibacterial peptide defensin directly interacts with viral major outer capsid protein P10 and thus binds to viral particles, finally blocking effective viral infection in planthopper vector. Furthermore, viral tubular protein P7-1 directly interacts with and promotes RING E3 ubiquitin ligase-mediated ubiquitinated degradation of Toll pathway adaptor protein MyD88 through the 26 proteasome pathway, finally suppressing antiviral defensin production. This virus-mediated attenuation of Toll antiviral immune response to express antiviral defensin ensures persistent virus infection without causing evident fitness costs for the insects. E3 ubiquitin ligase also is directly involved in the assembly of virus-induced tubules constructed by P7-1 to facilitate viral spread in planthopper vector, thereby acting as a pro-viral factor. Together, we uncover a previously unknown mechanism used by plant arboviruses to suppress Toll immune response through the ubiquitinated degradation of the conserved adaptor protein MyD88, thereby facilitating the coexistence of arboviruses with their vectors in nature.
Collapse
Affiliation(s)
- Dongsheng Jia
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Guozhong Luo
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Heran Guan
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Tingting Yu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Xinyan Sun
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Yu Du
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Yiheng Wang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Hongyan Chen
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Taiyun Wei
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| |
Collapse
|
5
|
Hédelin L, Thiébaut A, Huang J, Li X, Lemoine A, Haas G, Meignin C, Cai H, Waterhouse RM, Martins N, Imler JL. Investigating the Evolution of Drosophila STING-Dependent Antiviral Innate Immunity by Multispecies Comparison of 2'3'-cGAMP Responses. Mol Biol Evol 2024; 41:msae032. [PMID: 38377349 PMCID: PMC10917227 DOI: 10.1093/molbev/msae032] [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: 10/13/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Viruses represent a major threat to all animals, which defend themselves through induction of a large set of virus-stimulated genes that collectively control the infection. In vertebrates, these genes include interferons that play a critical role in the amplification of the response to infection. Virus- and interferon-stimulated genes include restriction factors targeting the different steps of the viral replication cycle, in addition to molecules associated with inflammation and adaptive immunity. Predictably, antiviral genes evolve dynamically in response to viral pressure. As a result, each animal has a unique arsenal of antiviral genes. Here, we exploit the capacity to experimentally activate the evolutionarily conserved stimulator of IFN genes (STING) signaling pathway by injection of the cyclic dinucleotide 2'3'-cyclic guanosine monophosphate-adenosine monophosphate into flies to define the repertoire of STING-regulated genes in 10 Drosophila species, spanning 40 million years of evolution. Our data reveal a set of conserved STING-regulated factors, including STING itself, a cGAS-like-receptor, the restriction factor pastel, and the antiviral protein Vago, but also 2 key components of the antiviral RNA interference pathway, Dicer-2, and Argonaute2. In addition, we identify unknown species- or lineage-specific genes that have not been previously associated with resistance to viruses. Our data provide insight into the core antiviral response in Drosophila flies and pave the way for the characterization of previously unknown antiviral effectors.
Collapse
Affiliation(s)
- Léna Hédelin
- CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Antonin Thiébaut
- Department of Ecology and Evolution, SIB Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Jingxian Huang
- School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Li
- School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Aurélie Lemoine
- CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Gabrielle Haas
- CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Carine Meignin
- CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Hua Cai
- School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Robert M Waterhouse
- Department of Ecology and Evolution, SIB Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Nelson Martins
- CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - Jean-Luc Imler
- CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
- School of Basic Medical Science, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Song C, Kim MY, Cho JY. The Role of Protein Methyltransferases in Immunity. Molecules 2024; 29:360. [PMID: 38257273 PMCID: PMC10819338 DOI: 10.3390/molecules29020360] [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: 11/26/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The immune system protects our body from bacteria, viruses, and toxins and removes malignant cells. Activation of immune cells requires the onset of a network of important signaling proteins. Methylation of these proteins affects their structure and biological function. Under stimulation, T cells, B cells, and other immune cells undergo activation, development, proliferation, differentiation, and manufacture of cytokines and antibodies. Methyltransferases alter the above processes and lead to diverse outcomes depending on the degree and type of methylation. In the previous two decades, methyltransferases have been reported to mediate a great variety of immune stages. Elucidating the roles of methylation in immunity not only contributes to understanding the immune mechanism but is helpful in the development of new immunotherapeutic strategies. Hence, we review herein the studies on methylation in immunity, aiming to provide ideas for new approaches.
Collapse
Affiliation(s)
- Chaoran Song
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| |
Collapse
|
7
|
Romanov SE, Shloma VV, Maksimov DA, Koryakov DE. SetDB1 and Su(var)3-9 are essential for late stages of larval development of Drosophila melanogaster. Chromosome Res 2023; 31:35. [PMID: 38099968 DOI: 10.1007/s10577-023-09743-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023]
Abstract
Methylation of H3K9 histone residue is a marker of gene silencing in eukaryotes. Three enzymes responsible for adding this modification - G9a, SetDB1/Egg, and Su(var)3-9 - are known in Drosophila. To understand how simultaneous mutations of SetDB1 and Su(var)3-9 may affect the fly development, appropriate combinations were obtained. Double mutants egg; Su(var)3-9 displayed pronounced embryonic lethality, slower larval growth and died before or during metamorphosis. Analysis of transcription in larval salivary glands and wing imaginal disks indicated that the effect of double mutation is tissue-specific. In salivary gland chromosomes, affected genes display low H3K9me2 enrichment and are rarely bound by SetDB1 or Su(var)3-9. We suppose that each of these enzymes directly or indirectly controls its own set of gene targets in different organs, and double mutation results in an imbalanced developmental program. This also indicates that SetDB1 and Su(var)3-9 may affect transcription via H3K9-independent mechanisms. Unexpectedly, in double and triple mutants, amount of di- and tri-methylated H3K9 is drastically reduced, but not completely absent. We hypothesize that this residual methylation implies the existence of additional H3K9-specific methyltransferase in Drosophila.
Collapse
Affiliation(s)
- Stanislav E Romanov
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, 630090, Russia
| | - Viktor V Shloma
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, 630090, Russia
| | - Daniil A Maksimov
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, 630090, Russia
| | - Dmitry E Koryakov
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, 630090, Russia.
| |
Collapse
|
8
|
Prakash A, Monteith KM, Bonnet M, Vale PF. Duox and Jak/Stat signalling influence disease tolerance in Drosophila during Pseudomonas entomophila infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104756. [PMID: 37302730 DOI: 10.1016/j.dci.2023.104756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/18/2023] [Accepted: 06/09/2023] [Indexed: 06/13/2023]
Abstract
Disease tolerance describes an infected host's ability to maintain health independently of the ability to clear microbe loads. The Jak/Stat pathway plays a pivotal role in humoral innate immunity by detecting tissue damage and triggering cellular renewal, making it a candidate tolerance mechanism. Here, we find that in Drosophila melanogaster infected with Pseudomonas entomophila disrupting ROS-producing dual oxidase (duox) or the negative regulator of Jak/Stat Socs36E, render male flies less tolerant. Another negative regulator of Jak/Stat, G9a - which has previously been associated with variable tolerance of viral infections - did not affect the rate of mortality with increasing microbe loads compared to flies with functional G9a, suggesting it does not affect tolerance of bacterial infection as in viral infection. Our findings highlight that ROS production and Jak/Stat signalling influence the ability of flies to tolerate bacterial infection sex-specifically and may therefore contribute to sexually dimorphic infection outcomes in Drosophila.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, UK.
| | - Katy M Monteith
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, UK
| | - Mickael Bonnet
- UFR De Biologie, Campus Universitaire Des Cezeaux, France
| | - Pedro F Vale
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, UK.
| |
Collapse
|
9
|
Hagedorn E, Bunnell D, Henschel B, Smith DL, Dickinson S, Brown AW, De Luca M, Turner AN, Chtarbanova S. RNA virus-mediated changes in organismal oxygen consumption rate in young and old Drosophila melanogaster males. Aging (Albany NY) 2023; 15:1748-1767. [PMID: 36947702 PMCID: PMC10085608 DOI: 10.18632/aging.204593] [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: 10/20/2022] [Accepted: 02/20/2023] [Indexed: 03/24/2023]
Abstract
Aging is accompanied by increased susceptibility to infections including with viral pathogens resulting in higher morbidity and mortality among the elderly. Significant changes in host metabolism can take place following virus infection. Efficient immune responses are energetically costly, and viruses divert host molecular resources to promote their own replication. Virus-induced metabolic reprogramming could impact infection outcomes, however, how this is affected by aging and impacts organismal survival remains poorly understood. RNA virus infection of Drosophila melanogaster with Flock House virus (FHV) is an effective model to study antiviral responses with age, where older flies die faster than younger flies due to impaired disease tolerance. Using this aged host-virus model, we conducted longitudinal, single-fly respirometry studies to determine if metabolism impacts infection outcomes. Analysis using linear mixed models on Oxygen Consumption Rate (OCR) following the first 72-hours post-infection showed that FHV modulates respiration, but age has no significant effect on OCR. However, the longitudinal assessment revealed that OCR in young flies progressively and significantly decreases, while OCR in aged flies remains constant throughout the three days of the experiment. Furthermore, we found that the OCR signature at 24-hours varied in response to both experimental treatment and survival status. FHV-injected flies that died prior to 48- or 72-hours measurements had a lower OCR compared to survivors at 48-hours. Our findings suggest the host's metabolic profile could influence the outcome of viral infections.
Collapse
Affiliation(s)
- Eli Hagedorn
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35401, USA
- Present Address: Indiana University School of Medicine-Indianapolis, Medical Scientist Training Program, Indianapolis, IN 46202, USA
| | - Dean Bunnell
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35401, USA
| | - Beate Henschel
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Biostatistics Consulting Center, Bloomington, IN 47405, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie Dickinson
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Biostatistics Consulting Center, Bloomington, IN 47405, USA
| | - Andrew W. Brown
- Department of Applied Health Sciences, Indiana University, School of Public Health-Bloomington, Bloomington, IN 47405, USA
- Present Address: University of Arkansas for Medical Sciences and Arkansas Children’s Research Institute, Little Rock, AR 72202, USA
| | - Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ashley N. Turner
- Department of Biology, Jacksonville State University, Jacksonville, AL 36265, USA
| | - Stanislava Chtarbanova
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35401, USA
- Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Convergent Bioscience and Medicine, University of Alabama, Tuscaloosa, AL 35401, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL 35401, USA
| |
Collapse
|
10
|
Kutzer MAM, Gupta V, Neophytou K, Doublet V, Monteith KM, Vale PF. Intraspecific genetic variation in host vigour, viral load and disease tolerance during Drosophila C virus infection. Open Biol 2023; 13:230025. [PMID: 36854375 PMCID: PMC9974301 DOI: 10.1098/rsob.230025] [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: 03/02/2023] Open
Abstract
Genetic variation for resistance and disease tolerance has been described in a range of species. In Drosophila melanogaster, genetic variation in mortality following systemic Drosophila C virus (DCV) infection is driven by large-effect polymorphisms in the restriction factor pastrel (pst). However, it is unclear if pst contributes to disease tolerance. We investigated systemic DCV challenges spanning nine orders of magnitude, in males and females of 10 Drosophila Genetic Reference Panel lines carrying either a susceptible (S) or resistant (R) pst allele. We find among-line variation in fly survival, viral load and disease tolerance measured both as the ability to maintain survival (mortality tolerance) and reproduction (fecundity tolerance). We further uncover novel effects of pst on host vigour, as flies carrying the R allele exhibited higher survival and fecundity even in the absence of infection. Finally, we found significant genetic variation in the expression of the JAK-STAT ligand upd3 and the epigenetic regulator of JAK-STAT G9a. However, while G9a has been previously shown to mediate tolerance of DCV infection, we found no correlation between the expression of either upd3 or G9a on fly tolerance or resistance. Our work highlights the importance of both resistance and tolerance in viral defence.
Collapse
Affiliation(s)
- Megan A. M. Kutzer
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, UK
| | - Vanika Gupta
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, UK
| | - Kyriaki Neophytou
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, UK
| | - Vincent Doublet
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, UK
| | - Katy M. Monteith
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, UK
| | - Pedro F. Vale
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, UK
| |
Collapse
|
11
|
Huang Z, Wang W, Xu P, Gong S, Hu Y, Liu Y, Su F, Anjum KM, Deng WM, Yang S, Liu J, Jiao R, Chen J. Drosophila Ectoderm-expressed 4 modulates JAK/STAT pathway and protects flies against Drosophila C virus infection. Front Immunol 2023; 14:1135625. [PMID: 36817462 PMCID: PMC9937023 DOI: 10.3389/fimmu.2023.1135625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Sterile alpha and HEAT/Armadillo motif-containing protein (SARM) is conserved in evolution and negatively regulates TRIF-dependent Toll signaling in mammals. The SARM protein from Litopenaeus vannamei and its Drosophila orthologue Ectoderm-expressed (Ect4) are also involved in immune defense against pathogen infection. However, the functional mechanism of the protective effect remains unclear. In this study, we show that Ect4 is essential for the viral load in flies after a Drosophila C virus (DCV) infection. Viral load is increased in Ect4 mutants resulting in higher mortality rates than wild-type. Overexpression of Ect4 leads to a suppression of virus replication and thus improves the survival rate of the animals. Ect4 is required for the viral induction of STAT-responsive genes, TotA and TotM. Furthermore, Ect4 interacts with Stat92E, affecting the tyrosine phosphorylation and nuclear translocation of Stat92E in S2 cells. Altogether, our study identifies the adaptor protein Ect4 of the Toll pathway contributes to resistance to viral infection and regulates JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Zongliang Huang
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China,Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Wang
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China,Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Pengpeng Xu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shangyu Gong
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yingshan Hu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Liu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fang Su
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Khalid Mahmood Anjum
- Department of Wildlife and Ecology, University of Veterinary and Animal Sciences, Lahore, Punjab, Pakistan
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Suping Yang
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Jiyong Liu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Jiyong Liu, ; Renjie Jiao, ; Jianming Chen,
| | - Renjie Jiao
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Jiyong Liu, ; Renjie Jiao, ; Jianming Chen,
| | - Jianming Chen
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China,Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Jiyong Liu, ; Renjie Jiao, ; Jianming Chen,
| |
Collapse
|
12
|
Ferguson JM, González-González A, Kaiser JA, Winzer SM, Anast JM, Ridenhour B, Miura TA, Parent CE. Hidden variable models reveal the effects of infection from changes in host survival. PLoS Comput Biol 2023; 19:e1010910. [PMID: 36812266 PMCID: PMC9987815 DOI: 10.1371/journal.pcbi.1010910] [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: 02/14/2022] [Revised: 03/06/2023] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
The impacts of disease on host vital rates can be demonstrated using longitudinal studies, but these studies can be expensive and logistically challenging. We examined the utility of hidden variable models to infer the individual effects of infectious disease from population-level measurements of survival when longitudinal studies are not possible. Our approach seeks to explain temporal deviations in population-level survival after introducing a disease causative agent when disease prevalence cannot be directly measured by coupling survival and epidemiological models. We tested this approach using an experimental host system (Drosophila melanogaster) with multiple distinct pathogens to validate the ability of the hidden variable model to infer per-capita disease rates. We then applied the approach to a disease outbreak in harbor seals (Phoca vituline) that had data on observed strandings but no epidemiological data. We found that our hidden variable modeling approach could successfully detect the per-capita effects of disease from monitored survival rates in both the experimental and wild populations. Our approach may prove useful for detecting epidemics from public health data in regions where standard surveillance techniques are not available and in the study of epidemics in wildlife populations, where longitudinal studies can be especially difficult to implement.
Collapse
Affiliation(s)
- Jake M. Ferguson
- Department of Biology, University of Hawaiʻi at Mānoa, Honolulu, Hawaii, United States of America
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
| | - Andrea González-González
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Johnathan A. Kaiser
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Sara M. Winzer
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Justin M. Anast
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Ben Ridenhour
- Department of Mathematics, University of Idaho, Moscow, Idaho, United States of America
| | - Tanya A. Miura
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Christine E. Parent
- Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Institute for Interdisciplinary Data Sciences, University of Idaho, Moscow, Idaho, United States of America
| |
Collapse
|
13
|
Rai S, Tapadia MG. Hsc70-4 aggravates PolyQ-mediated neurodegeneration by modulating NF-κB mediated immune response in Drosophila. Front Mol Neurosci 2022; 15:857257. [PMID: 36425218 PMCID: PMC9678916 DOI: 10.3389/fnmol.2022.857257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 10/11/2022] [Indexed: 10/06/2023] Open
Abstract
Huntington's disease occurs when the stretch of CAG repeats in exon 1 of the huntingtin (htt) gene crosses the permissible limit, causing the mutated protein (mHtt) to form insoluble aggregates or inclusion bodies. These aggregates are non-typically associated with various essential proteins in the cells, thus disrupting cellular homeostasis. The cells try to bring back normalcy by synthesizing evolutionary conserved cellular chaperones, and Hsp70 is one of the families of heat shock proteins that has a significant part in this, which comprises of heat-inducible and cognate forms. Here, we demonstrate that the heat shock cognate (Hsc70) isoform, Hsc70-4/HSPA8, has a distinct role in polyglutamate (PolyQ)-mediated pathogenicity, and its expression is enhanced in the polyQ conditions in Drosophila. Downregulation of hsc70-4 rescues PolyQ pathogenicity with a notable improvement in the ommatidia arrangement and near-normal restoration of optic neurons leading to improvement in phototaxis response. Reduced hsc70-4 also attenuates the augmented immune response by decreasing the expression of NF-κB and the antimicrobial peptides, along with that JNK overactivation is also restored. These lead to the rescue of the photoreceptor cells, indicating a decrease in the caspase activity, thus reverting the PolyQ pathogenicity. At the molecular level, we show the interaction between Hsc70-4, Polyglutamine aggregates, and NF-κB, which may be responsible for the dysregulation of signaling molecules in polyQ conditions. Thus, the present data provides a functional link between Hsc70-4 and NF-κB under polyQ conditions.
Collapse
Affiliation(s)
| | - Madhu G. Tapadia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
14
|
Elucidating Mechanisms of Tolerance to Salmonella Typhimurium across Long-Term Infections Using the Collaborative Cross. mBio 2022; 13:e0112022. [PMID: 35880881 PMCID: PMC9426527 DOI: 10.1128/mbio.01120-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Understanding the molecular mechanisms underlying resistance and tolerance to pathogen infection may present the opportunity to develop novel interventions. Resistance is the absence of clinical disease with a low pathogen burden, while tolerance is minimal clinical disease with a high pathogen burden. Salmonella is a worldwide health concern. We studied 18 strains of collaborative cross mice that survive acute Salmonella Typhimurium (STm) infections. We infected these strains orally and monitored them for 3 weeks. Five strains cleared STm (resistant), six strains maintained a bacterial load and survived (tolerant), while seven strains survived >7 days but succumbed to infection within the study period and were called “delayed susceptible.” Tolerant strains were colonized in the Peyer’s patches, mesenteric lymph node, spleen, and liver, while resistant strains had significantly reduced bacterial colonization. Tolerant strains had lower preinfection core body temperatures and had disrupted circadian patterns of body temperature postinfection sooner than other strains. Tolerant strains had higher circulating total white blood cells than resistant strains, driven by increased numbers of neutrophils. Tolerant strains had more severe tissue damage and higher circulating levels of monocyte chemoattractant protein 1 (MCP-1) and interferon gamma (IFN-γ), but lower levels of epithelial neutrophil-activating protein 78 (ENA-78) than resistant strains. Quantitative trait locus (QTL) analysis revealed one significant association and six suggestive associations. Gene expression analysis identified 22 genes that are differentially regulated in tolerant versus resistant animals that overlapped these QTLs. Fibrinogen genes (Fga, Fgb, and Fgg) were found across the QTL, RNA, and top canonical pathways, making them the best candidate genes for differentiating tolerance and resistance.
Collapse
|
15
|
Mongelli V, Lequime S, Kousathanas A, Gausson V, Blanc H, Nigg J, Quintana-Murci L, Elena SF, Saleh MC. Innate immune pathways act synergistically to constrain RNA virus evolution in Drosophila melanogaster. Nat Ecol Evol 2022; 6:565-578. [PMID: 35273366 PMCID: PMC7612704 DOI: 10.1038/s41559-022-01697-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 12/14/2021] [Indexed: 02/05/2023]
Abstract
Host-pathogen interactions impose recurrent selective pressures that lead to constant adaptation and counter-adaptation in both competing species. Here, we sought to study this evolutionary arms-race and assessed the impact of the innate immune system on viral population diversity and evolution, using Drosophila melanogaster as model host and its natural pathogen Drosophila C virus (DCV). We isogenized eight fly genotypes generating animals defective for RNAi, Imd and Toll innate immune pathways as well as pathogen-sensing and gut renewal pathways. Wild-type or mutant flies were then orally infected with DCV and the virus was serially passaged ten times via reinfection in naive flies. Viral population diversity was studied after each viral passage by high-throughput sequencing and infection phenotypes were assessed at the beginning and at the end of the evolution experiment. We found that the absence of any of the various immune pathways studied increased viral genetic diversity while attenuating virulence. Strikingly, these effects were observed in a range of host factors described as having mainly antiviral or antibacterial functions. Together, our results indicate that the innate immune system as a whole and not specific antiviral defence pathways in isolation, generally constrains viral diversity and evolution.
Collapse
Affiliation(s)
- Vanesa Mongelli
- Viruses and RNA Interference Unit, Institut Pasteur, CNRS, Paris, France
| | - Sebastian Lequime
- Cluster of Microbial Ecology, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | | | - Valérie Gausson
- Viruses and RNA Interference Unit, Institut Pasteur, CNRS, Paris, France
| | - Hervé Blanc
- Viruses and RNA Interference Unit, Institut Pasteur, CNRS, Paris, France
| | - Jared Nigg
- Viruses and RNA Interference Unit, Institut Pasteur, CNRS, Paris, France
| | - Lluis Quintana-Murci
- Human Evolutionary Genetic Unit, Institut Pasteur, CNRS, Paris, France
- Human Genomics and Evolution, Collège de France, Paris, France
| | - Santiago F Elena
- Instituto de Biología Integrativa de Sistemas (CSIC-Universitat de València), València, Spain.
- The Santa Fe Institute, Santa Fe, NM, USA.
| | - Maria-Carla Saleh
- Viruses and RNA Interference Unit, Institut Pasteur, CNRS, Paris, France.
| |
Collapse
|
16
|
Roy M, Viginier B, Mayeux CA, Ratinier M, Fablet M. Infections by Transovarially Transmitted DMelSV in Drosophila Have No Impact on Ovarian Transposable Element Transcripts but Increase Their Amounts in the Soma. Genome Biol Evol 2021; 13:evab207. [PMID: 34498066 PMCID: PMC8459167 DOI: 10.1093/gbe/evab207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Transposable elements (TEs) are genomic parasites, which activity is tightly controlled in germline cells. Using Sindbis virus, it was recently demonstrated that viral infections affect TE transcript amounts in somatic tissues. However, the strongest evolutionary impacts are expected in gonads, because that is where the genomes of the next generations lie. Here, we investigated this aspect using the Drosophila melanogaster Sigma virus. It is particularly relevant in the genome/TE interaction given its tropism to ovaries, which is the organ displaying the more sophisticated TE control pathways. Our results in Drosophila simulans flies allowed us to confirm the existence of a strong homeostasis of the TE transcriptome in ovaries upon infection, which, however, rely on TE-derived small RNA modulations. In addition, we performed a meta-analysis of RNA-seq data and propose that the immune pathway that is triggered upon viral infection determines the direction of TE transcript modulation in somatic tissues.
Collapse
Affiliation(s)
- Marlène Roy
- Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, Université Lyon 1, CNRS, UMR 5558, Villeurbanne, France
- EPHE, PSL Research University, INRA, Université de Lyon, Université Claude Bernard Lyon1, UMR754, IVPC, Lyon, France
| | - Barbara Viginier
- EPHE, PSL Research University, INRA, Université de Lyon, Université Claude Bernard Lyon1, UMR754, IVPC, Lyon, France
| | - Camille A Mayeux
- Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, Université Lyon 1, CNRS, UMR 5558, Villeurbanne, France
| | - Maxime Ratinier
- EPHE, PSL Research University, INRA, Université de Lyon, Université Claude Bernard Lyon1, UMR754, IVPC, Lyon, France
| | - Marie Fablet
- Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, Université Lyon 1, CNRS, UMR 5558, Villeurbanne, France
| |
Collapse
|
17
|
Ren X, Wang R, Yu XT, Cai B, Guo F. Regulation of histone H3 lysine 9 methylation in inflammation. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1931477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Xin Ren
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Rong Wang
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Xiao-ting Yu
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Bo Cai
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Fei Guo
- Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
18
|
Rosendo Machado S, van der Most T, Miesen P. Genetic determinants of antiviral immunity in dipteran insects - Compiling the experimental evidence. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104010. [PMID: 33476667 DOI: 10.1016/j.dci.2021.104010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 06/12/2023]
Abstract
The genetic basis of antiviral immunity in dipteran insects is extensively studied in Drosophila melanogaster and advanced technologies for genetic manipulation allow a better characterization of immune responses also in non-model insect species. Especially, immunity in vector mosquitoes is recently in the spotlight, due to the medical impact that these insects have by transmitting viruses and other pathogens. Here, we review the current state of experimental evidence that supports antiviral functions for immune genes acting in different cellular pathways. We discuss the well-characterized RNA interference mechanism along with the less well-defined JAK-STAT, Toll, and IMD signaling pathways. Furthermore, we highlight the initial evidence for antiviral activity observed for the autophagy pathway, transcriptional pausing, as well as piRNA production from endogenous viral elements. We focus our review on studies from Drosophila and mosquito species from the lineages Aedes, Culex, and Anopheles, which contain major vector species responsible for virus transmission.
Collapse
Affiliation(s)
- Samara Rosendo Machado
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Tom van der Most
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands.
| |
Collapse
|
19
|
Riahi H, Fenckova M, Goruk KJ, Schenck A, Kramer JM. The epigenetic regulator G9a attenuates stress-induced resistance and metabolic transcriptional programs across different stressors and species. BMC Biol 2021; 19:112. [PMID: 34030685 PMCID: PMC8142638 DOI: 10.1186/s12915-021-01025-0] [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: 05/31/2020] [Accepted: 04/14/2021] [Indexed: 01/07/2023] Open
Abstract
Background Resistance and tolerance are two coexisting defense strategies for fighting infections. Resistance is mediated by signaling pathways that induce transcriptional activation of resistance factors that directly eliminate the pathogen. Tolerance refers to adaptations that limit the health impact of a given pathogen burden, without targeting the infectious agent. The key players governing immune tolerance are largely unknown. In Drosophila, the histone H3 lysine 9 (H3K9) methyltransferase G9a was shown to mediate tolerance to virus infection and oxidative stress (OS), suggesting that abiotic stresses like OS may also evoke tolerance mechanisms. In response to both virus and OS, stress resistance genes were overinduced in Drosophila G9a mutants, suggesting an intact but overactive stress response. We recently demonstrated that G9a promotes tolerance to OS by maintaining metabolic homeostasis and safeguarding energy availability, but it remained unclear if this mechanism also applies to viral infection, or is conserved in other species and stress responses. To address these questions, we analyzed publicly available datasets from Drosophila, mouse, and human in which global gene expression levels were measured in G9a-depleted conditions and controls at different time points upon stress exposure. Results In all investigated datasets, G9a attenuates the transcriptional stress responses that confer resistance against the encountered stressor. Comparative analysis of conserved G9a-dependent stress response genes suggests that G9a is an intimate part of the design principles of stress resistance, buffering the induction of promiscuous stress signaling pathways and stress-specific resistance factors. Importantly, we find stress-dependent downregulation of metabolic genes to also be dependent on G9a across all of the tested datasets. Conclusions These results suggest that G9a sets the balance between activation of resistance genes and maintaining metabolic homeostasis, thereby ensuring optimal organismal performance during exposure to diverse types of stress across different species. We therefore propose G9a as a potentially conserved master regulator underlying the widely important, yet poorly understood, concept of stress tolerance. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01025-0.
Collapse
Affiliation(s)
- Human Riahi
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kayla J Goruk
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Jamie M Kramer
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
20
|
Leite THJF, Ferreira ÁGA, Imler JL, Marques JT. Distinct Roles of Hemocytes at Different Stages of Infection by Dengue and Zika Viruses in Aedes aegypti Mosquitoes. Front Immunol 2021; 12:660873. [PMID: 34093550 PMCID: PMC8169962 DOI: 10.3389/fimmu.2021.660873] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
Aedes aegypti mosquitoes are vectors for arboviruses of medical importance such as dengue (DENV) and Zika (ZIKV) viruses. Different innate immune pathways contribute to the control of arboviruses in the mosquito vector including RNA interference, Toll and Jak-STAT pathways. However, the role of cellular responses mediated by circulating macrophage-like cells known as hemocytes remains unclear. Here we show that hemocytes are recruited to the midgut of Ae. aegypti mosquitoes in response to DENV or ZIKV. Blockade of the phagocytic function of hemocytes using latex beads induced increased accumulation of hemocytes in the midgut and a reduction in virus infection levels in this organ. In contrast, inhibition of phagocytosis by hemocytes led to increased systemic dissemination and replication of DENV and ZIKV. Hence, our work reveals a dual role for hemocytes in Ae. aegypti mosquitoes, whereby phagocytosis is not required to control viral infection in the midgut but is essential to restrict systemic dissemination. Further understanding of the mechanism behind this duality could help the design of vector-based strategies to prevent transmission of arboviruses.
Collapse
Affiliation(s)
- Thiago H J F Leite
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Álvaro G A Ferreira
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou - Fiocruz, Belo Horizonte, Brazil
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, Inserm U1257, Strasbourg, France
| | - João T Marques
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Université de Strasbourg, CNRS UPR9022, Inserm U1257, Strasbourg, France
| |
Collapse
|
21
|
Sciambra N, Chtarbanova S. The Impact of Age on Response to Infection in Drosophila. Microorganisms 2021; 9:microorganisms9050958. [PMID: 33946849 PMCID: PMC8145649 DOI: 10.3390/microorganisms9050958] [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: 03/30/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/26/2023] Open
Abstract
This review outlines the known cellular pathways and mechanisms involved in Drosophila age-dependent immunity to pathogenic microorganisms such as bacteria and fungi. We discuss the implication of host signaling pathways such as the Toll, Immune Deficiency (IMD), Janus kinase signal transducer and activator of transcription (JAK/STAT), and Insulin/Insulin Growth Factor/Target of Rapamycin (IIS/TOR) on immune function with aging. Additionally, we review the effects that factors such as sexual dimorphism, environmental stress, and cellular physiology exert on age-dependent immunity in Drosophila. We discuss potential tradeoffs between heightened immune function and longevity in the absence of infection, and we provide detailed tables outlining the various assays and pathogens used in the cited studies, as well as the age, sex, and strains of Drosophila used. We also discuss the overlapping effects these pathways and mechanisms have on one another. We highlight the great utility of Drosophila as a model organism and the importance of a greater focus on age-dependent antiviral immunity for future studies.
Collapse
|
22
|
Sheffield L, Sciambra N, Evans A, Hagedorn E, Goltz C, Delfeld M, Kuhns H, Fierst JL, Chtarbanova S. Age-dependent impairment of disease tolerance is associated with a robust transcriptional response following RNA virus infection in Drosophila. G3-GENES GENOMES GENETICS 2021; 11:6219303. [PMID: 33836060 PMCID: PMC8495950 DOI: 10.1093/g3journal/jkab116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022]
Abstract
Advanced age in humans is associated with greater susceptibility to and higher mortality rates from infections, including infections with some RNA viruses. The underlying innate immune mechanisms, which represent the first line of defense against pathogens, remain incompletely understood. Drosophila melanogaster is able to mount potent and evolutionarily conserved innate immune defenses against a variety of microorganisms including viruses and serves as an excellent model organism for studying host–pathogen interactions. With its relatively short lifespan, Drosophila also is an organism of choice for aging studies. Despite numerous advantages that this model offers, Drosophila has not been used to its full potential to investigate the response of the aged host to viral infection. Here, we show that, in comparison to younger flies, aged Drosophila succumb more rapidly to infection with the RNA-containing Flock House virus due to an age-dependent defect in disease tolerance. Relative to younger individuals, we find that older Drosophila mount transcriptional responses characterized by differential regulation of more genes and genes regulated to a greater extent. We show that loss of disease tolerance to Flock House virus with age associates with a stronger regulation of genes involved in apoptosis, some genes of the Drosophila immune deficiency NF-kB pathway, and genes whose products function in mitochondria and mitochondrial respiration. Our work shows that Drosophila can serve as a model to investigate host–virus interactions during aging and furthermore sets the stage for future analysis of the age-dependent mechanisms that govern survival and control of virus infections at older age.
Collapse
Affiliation(s)
- Lakbira Sheffield
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA.,Graduate Biomedical Sciences program, University of Alabama at Birmingham, Birmingham, AL- 35294, USA
| | - Noah Sciambra
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Alysa Evans
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Eli Hagedorn
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Casey Goltz
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Megan Delfeld
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Haley Kuhns
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Janna L Fierst
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Stanislava Chtarbanova
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| |
Collapse
|
23
|
Schneider J, Imler JL. Sensing and signalling viral infection in drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 117:103985. [PMID: 33358662 DOI: 10.1016/j.dci.2020.103985] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 06/12/2023]
Abstract
The fruitfly Drosophila melanogaster is a valuable model to unravel mechanisms of innate immunity, in particular in the context of viral infections. RNA interference, and more specifically the small interfering RNA pathway, is a major component of antiviral immunity in drosophila. In addition, the contribution of inducible transcriptional responses to the control of viruses in drosophila and other invertebrates is increasingly recognized. In particular, the recent discovery of a STING-IKKβ-Relish signalling cassette in drosophila has confirmed that NF-κB transcription factors play an important role in the control of viral infections, in addition to bacterial and fungal infections. Here, we review recent developments in the field, which begin to shed light on the mechanisms involved in sensing of viral infections and in signalling leading to production of antiviral effectors.
Collapse
Affiliation(s)
- Juliette Schneider
- Université de Strasbourg, CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
24
|
Li-Byarlay H, Boncristiani H, Howell G, Herman J, Clark L, Strand MK, Tarpy D, Rueppell O. Transcriptomic and Epigenomic Dynamics of Honey Bees in Response to Lethal Viral Infection. Front Genet 2020; 11:566320. [PMID: 33101388 PMCID: PMC7546774 DOI: 10.3389/fgene.2020.566320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/17/2020] [Indexed: 12/28/2022] Open
Abstract
Honey bees (Apis mellifera L.) suffer from many brood pathogens, including viruses. Despite considerable research, the molecular responses and dynamics of honey bee pupae to viral pathogens remain poorly understood. Israeli Acute Paralysis Virus (IAPV) is emerging as a model virus since its association with severe colony losses. Using worker pupae, we studied the transcriptomic and methylomic consequences of IAPV infection over three distinct time points after inoculation. Contrasts of gene expression and 5 mC DNA methylation profiles between IAPV-infected and control individuals at these time points - corresponding to the pre-replicative (5 h), replicative (20 h), and terminal (48 h) phase of infection - indicate that profound immune responses and distinct manipulation of host molecular processes accompany the lethal progression of this virus. We identify the temporal dynamics of the transcriptomic response to with more genes differentially expressed in the replicative and terminal phases than in the pre-replicative phase. However, the number of differentially methylated regions decreased dramatically from the pre-replicative to the replicative and terminal phase. Several cellular pathways experienced hyper- and hypo-methylation in the pre-replicative phase and later dramatically increased in gene expression at the terminal phase, including the MAPK, Jak-STAT, Hippo, mTOR, TGF-beta signaling pathways, ubiquitin mediated proteolysis, and spliceosome. These affected biological functions suggest that adaptive host responses to combat the virus are mixed with viral manipulations of the host to increase its own reproduction, all of which are involved in anti-viral immune response, cell growth, and proliferation. Comparative genomic analyses with other studies of viral infections of honey bees and fruit flies indicated that similar immune pathways are shared. Our results further suggest that dynamic DNA methylation responds to viral infections quickly, regulating subsequent gene activities. Our study provides new insights of molecular mechanisms involved in epigenetic that can serve as foundation for the long-term goal to develop anti-viral strategies for honey bees, the most important commercial pollinator.
Collapse
Affiliation(s)
- Hongmei Li-Byarlay
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
| | - Humberto Boncristiani
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Gary Howell
- High Performance Cluster, Office of Information Technology, North Carolina State University, Raleigh, NC, United States
| | - Jake Herman
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Lindsay Clark
- High Performance Computing in Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Micheline K. Strand
- Army Research Office, Army Research Laboratory, Research Triangle Park, NC, United States
| | - David Tarpy
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States
| | - Olav Rueppell
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC, United States
| |
Collapse
|
25
|
Sui B, Chen D, Liu W, Wu Q, Tian B, Li Y, Hou J, Liu S, Xie J, Jiang H, Luo Z, Lv L, Huang F, Li R, Zhang C, Tian Y, Cui M, Zhou M, Chen H, Fu ZF, Zhang Y, Zhao L. A novel antiviral lncRNA, EDAL, shields a T309 O-GlcNAcylation site to promote EZH2 lysosomal degradation. Genome Biol 2020; 21:228. [PMID: 32873321 PMCID: PMC7465408 DOI: 10.1186/s13059-020-02150-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The central nervous system (CNS) is vulnerable to viral infection, yet few host factors in the CNS are known to defend against invasion by neurotropic viruses. Long noncoding RNAs (lncRNAs) have been revealed to play critical roles in a wide variety of biological processes and are highly abundant in the mammalian brain, but their roles in defending against invasion of pathogens into the CNS remain unclear. RESULTS We report here that multiple neurotropic viruses, including rabies virus, vesicular stomatitis virus, Semliki Forest virus, and herpes simplex virus 1, elicit the neuronal expression of a host-encoded lncRNA EDAL. EDAL inhibits the replication of these neurotropic viruses in neuronal cells and rabies virus infection in mouse brains. EDAL binds to the conserved histone methyltransferase enhancer of zest homolog 2 (EZH2) and specifically causes EZH2 degradation via lysosomes, reducing the cellular H3K27me3 level. The antiviral function of EDAL resides in a 56-nt antiviral substructure through which its 18-nt helix-loop intimately contacts multiple EZH2 sites surrounding T309, a known O-GlcNAcylation site. EDAL positively regulates the transcription of Pcp4l1 encoding a 10-kDa peptide, which inhibits the replication of multiple neurotropic viruses. CONCLUSIONS Our findings show that a neuronal lncRNA can exert an effective antiviral function via blocking a specific O-GlcNAcylation that determines EZH2 lysosomal degradation, rather than the traditional interferon-dependent pathway.
Collapse
Affiliation(s)
- Baokun Sui
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Dong Chen
- Center for Genome analysis, ABLife Inc., Wuhan, 430075, China
- Center for Genome analysis and Laboratory for Genome Regulation and Human Health, ABLife Inc., Wuhan, 430075, China
| | - Wei Liu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiong Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bin Tian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yingying Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jing Hou
- Center for Genome analysis, ABLife Inc., Wuhan, 430075, China
- Center for Genome analysis and Laboratory for Genome Regulation and Human Health, ABLife Inc., Wuhan, 430075, China
| | - Shiyong Liu
- School of Physics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Juan Xie
- School of Physics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, China
| | - Zhaochen Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lei Lv
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Fei Huang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ruiming Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chengguang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuling Tian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Min Cui
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhen F Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Department of Pathology, University of Georgia, Athens, GA, 30602, USA
| | - Yi Zhang
- Center for Genome analysis, ABLife Inc., Wuhan, 430075, China.
- Center for Genome analysis and Laboratory for Genome Regulation and Human Health, ABLife Inc., Wuhan, 430075, China.
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China.
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
26
|
González-González A, Wayne ML. Immunopathology and immune homeostasis during viral infection in insects. Adv Virus Res 2020; 107:285-314. [PMID: 32711732 DOI: 10.1016/bs.aivir.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Organisms clear infections by mounting an immune response that is normally turned off once the pathogens have been cleared. However, sometimes this immune response is not properly or timely arrested, resulting in the host damaging itself. This immune dysregulation may be referred to as immunopathology. While our knowledge of immune and metabolic pathways in insects, particularly in response to viral infections, is growing, little is known about the mechanisms that regulate this immune response and hence little is known about immunopathology in this important and diverse group of organisms. In this chapter we focus both on documenting the molecular mechanisms described involved in restoring immune homeostasis in insects after viral infections and on identifying potential mechanisms for future investigation. We argue that learning about the immunopathological consequences of an improperly regulated immune response in insects will benefit both insect and human health.
Collapse
Affiliation(s)
| | - Marta L Wayne
- Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
27
|
Abstract
Transposable elements (TEs) are genomic parasites that are found in all genomes, some of which display sequence similarity to certain viruses. In insects, TEs are controlled by the Piwi-interacting small interfering RNA (piRNA) pathway in gonads, while the small interfering RNA (siRNA) pathway is dedicated to TE somatic control and defense against viruses. So far, these two small interfering RNA pathways are considered to involve distinct molecular effectors and are described as independent. Using Sindbis virus (SINV) in Drosophila, here we show that viral infections affect TE transcript amounts via modulations of the piRNA and siRNA repertoires, with the clearest effects in somatic tissues. These results suggest that viral acute or chronic infections may impact TE activity and, thus, the tempo of genetic diversification. In addition, these results deserve further evolutionary considerations regarding potential benefits to the host, the virus, or the TEs.
Collapse
|
28
|
Oliveira JH, Bahia AC, Vale PF. How are arbovirus vectors able to tolerate infection? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103514. [PMID: 31585195 DOI: 10.1016/j.dci.2019.103514] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
One of the defining features of mosquito vectors of arboviruses such as Dengue and Zika is their ability to tolerate high levels of virus proliferation without suffering significant pathology. This adaptation is central to vector competence and disease spread. The molecular mechanisms, pathways, cellular and metabolic adaptations responsible for mosquito disease tolerance are still largely unknown and may represent effective ways to control mosquito populations and prevent arboviral diseases. In this review article, we describe the key link between disease tolerance and pathogen transmission, and how vector control methods may benefit by focusing efforts on dissecting the mechanisms underlying mosquito tolerance of arboviral infections. We briefly review recent work investigating tolerance mechanisms in other insects, describe the state of the art regarding the mechanisms of disease tolerance in mosquitos, and highlight the emerging role of gut microbiota in mosquito immunity and disease tolerance.
Collapse
Affiliation(s)
- José Henrique Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil.
| | - Ana Cristina Bahia
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Pedro F Vale
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
29
|
Merkling SH, Raquin V, Dabo S, Henrion-Lacritick A, Blanc H, Moltini-Conclois I, Frangeul L, Varet H, Saleh MC, Lambrechts L. Tudor-SN Promotes Early Replication of Dengue Virus in the Aedes aegypti Midgut. iScience 2020; 23:100870. [PMID: 32059176 PMCID: PMC7054812 DOI: 10.1016/j.isci.2020.100870] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/24/2019] [Accepted: 01/24/2020] [Indexed: 01/15/2023] Open
Abstract
Diseases caused by mosquito-borne viruses have been on the rise for the last decades, and novel methods aiming to use laboratory-engineered mosquitoes that are incapable of carrying viruses have been developed to reduce pathogen transmission. This has stimulated efforts to identify optimal target genes that are naturally involved in mosquito antiviral defenses or required for viral replication. Here, we investigated the role of a member of the Tudor protein family, Tudor-SN, upon dengue virus infection in the mosquito Aedes aegypti. Tudor-SN knockdown reduced dengue virus replication in the midgut of Ae. aegypti females. In immunofluorescence assays, Tudor-SN localized to the nucleolus in both Ae. aegypti and Aedes albopictus cells. A reporter assay and small RNA profiling demonstrated that Tudor-SN was not required for RNA interference function in vivo. Collectively, these results defined a novel proviral role for Tudor-SN upon early dengue virus infection of the Ae. aegypti midgut.
Collapse
Affiliation(s)
- Sarah Hélène Merkling
- Institut Pasteur, Insect-Virus Interactions Unit, UMR2000, CNRS, 75015 Paris, France
| | - Vincent Raquin
- Institut Pasteur, Insect-Virus Interactions Unit, UMR2000, CNRS, 75015 Paris, France
| | - Stéphanie Dabo
- Institut Pasteur, Insect-Virus Interactions Unit, UMR2000, CNRS, 75015 Paris, France
| | | | - Hervé Blanc
- Institut Pasteur, Viruses and RNA Interference Unit, UMR3569, CNRS, 75015 Paris, France
| | | | - Lionel Frangeul
- Institut Pasteur, Viruses and RNA Interference Unit, UMR3569, CNRS, 75015 Paris, France
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756, CNRS, Paris, France; Plate-forme Technologique Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, Paris, France
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, UMR3569, CNRS, 75015 Paris, France.
| | - Louis Lambrechts
- Institut Pasteur, Insect-Virus Interactions Unit, UMR2000, CNRS, 75015 Paris, France.
| |
Collapse
|
30
|
Belmonte RL, Corbally MK, Duneau DF, Regan JC. Sexual Dimorphisms in Innate Immunity and Responses to Infection in Drosophila melanogaster. Front Immunol 2020; 10:3075. [PMID: 32076419 PMCID: PMC7006818 DOI: 10.3389/fimmu.2019.03075] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022] Open
Abstract
The sexes show profound differences in responses to infection and the development of autoimmunity. Dimorphisms in immune responses are ubiquitous across taxa, from arthropods to vertebrates. Drosophila melanogaster shows strong sex dimorphisms in immune system responses at baseline, upon pathogenic challenge, and over aging. We have performed an exhaustive survey of peer-reviewed literature on Drosophila immunity, and present a database of publications indicating the sex(es) analyzed in each study. While we found a growing interest in the community in adult immunity and in reporting both sexes, the main body of work in this field uses only one sex, or does not stratify by sex. We synthesize evidence for sexually dimorphic responses to bacterial, viral, and fungal infections. Dimorphisms may be mediated by distinct immune compartments, and we review work on sex differences in behavioral, epithelial, cellular, and systemic (fat body-mediated) immunity. Emerging work on sexually dimorphic aging of immune tissues, immune senescence, and inflammation are examined. We consider evolutionary drivers for sex differences in immune investment, highlight the features of Drosophila biology that make it particularly amenable to studies of immune dimorphisms, and discuss areas for future exploration.
Collapse
Affiliation(s)
- Rebecca L. Belmonte
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Mary-Kate Corbally
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - David F. Duneau
- Laboratoire Evolution & Diversite Biologique, UMR5174 EDB, CNRS, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Jennifer C. Regan
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
31
|
Abstract
The inexorable emergence of mosquito-borne arboviruses and the failure of traditional vector control methods to prevent their transmission have triggered the development of alternative entomological interventions to render mosquito populations incapable of carrying arboviruses. Here, we use a theoretical framework to argue that decreasing mosquito tolerance to arbovirus infection could be a more evolutionarily sustainable disease control strategy than increasing mosquito resistance. Increasing resistance is predicted to select for mutant arboviruses escaping resistance, whereas reducing tolerance should lead to the death of infected vectors and thus select for mosquito-attenuated arbovirus variants that are less transmissible.
Collapse
Affiliation(s)
- Louis Lambrechts
- Institut Pasteur, Insect-Virus Interactions Unit, Department of Virology, UMR2000, CNRS, 75015 Paris, France.
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, UMR3569, CNRS, 75015 Paris, France.
| |
Collapse
|
32
|
Mačinković I, Theofel I, Hundertmark T, Kovač K, Awe S, Lenz J, Forné I, Lamp B, Nist A, Imhof A, Stiewe T, Renkawitz-Pohl R, Rathke C, Brehm A. Distinct CoREST complexes act in a cell-type-specific manner. Nucleic Acids Res 2019; 47:11649-11666. [PMID: 31701127 PMCID: PMC7145674 DOI: 10.1093/nar/gkz1050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 01/10/2023] Open
Abstract
CoREST has been identified as a subunit of several protein complexes that generate transcriptionally repressive chromatin structures during development. However, a comprehensive analysis of the CoREST interactome has not been carried out. We use proteomic approaches to define the interactomes of two dCoREST isoforms, dCoREST-L and dCoREST-M, in Drosophila. We identify three distinct histone deacetylase complexes built around a common dCoREST/dRPD3 core: A dLSD1/dCoREST complex, the LINT complex and a dG9a/dCoREST complex. The latter two complexes can incorporate both dCoREST isoforms. By contrast, the dLSD1/dCoREST complex exclusively assembles with the dCoREST-L isoform. Genome-wide studies show that the three dCoREST complexes associate with chromatin predominantly at promoters. Transcriptome analyses in S2 cells and testes reveal that different cell lineages utilize distinct dCoREST complexes to maintain cell-type-specific gene expression programmes: In macrophage-like S2 cells, LINT represses germ line-related genes whereas other dCoREST complexes are largely dispensable. By contrast, in testes, the dLSD1/dCoREST complex prevents transcription of germ line-inappropriate genes and is essential for spermatogenesis and fertility, whereas depletion of other dCoREST complexes has no effect. Our study uncovers three distinct dCoREST complexes that function in a lineage-restricted fashion to repress specific sets of genes thereby maintaining cell-type-specific gene expression programmes.
Collapse
Affiliation(s)
- Igor Mačinković
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Ina Theofel
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Tim Hundertmark
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Kristina Kovač
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Stephan Awe
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Jonathan Lenz
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Ignasi Forné
- Protein Analysis Unit, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Großhadernerstrasse 9, 82152 Martinsried, Germany
| | - Boris Lamp
- Genomics Core Facility, Institute of Molecular Oncology, Philipps-University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Institute of Molecular Oncology, Philipps-University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Axel Imhof
- Protein Analysis Unit, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Großhadernerstrasse 9, 82152 Martinsried, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Institute of Molecular Oncology, Philipps-University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Renate Renkawitz-Pohl
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Christina Rathke
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Alexander Brehm
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| |
Collapse
|
33
|
Harman JL, Dobnikar L, Chappell J, Stokell BG, Dalby A, Foote K, Finigan A, Freire-Pritchett P, Taylor AL, Worssam MD, Madsen RR, Loche E, Uryga A, Bennett MR, Jørgensen HF. Epigenetic Regulation of Vascular Smooth Muscle Cells by Histone H3 Lysine 9 Dimethylation Attenuates Target Gene-Induction by Inflammatory Signaling. Arterioscler Thromb Vasc Biol 2019; 39:2289-2302. [PMID: 31434493 PMCID: PMC6818986 DOI: 10.1161/atvbaha.119.312765] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Vascular inflammation underlies cardiovascular disease. Vascular smooth muscle cells (VSMCs) upregulate selective genes, including MMPs (matrix metalloproteinases) and proinflammatory cytokines upon local inflammation, which directly contribute to vascular disease and adverse clinical outcome. Identification of factors controlling VSMC responses to inflammation is therefore of considerable therapeutic importance. Here, we determine the role of Histone H3 lysine 9 di-methylation (H3K9me2), a repressive epigenetic mark that is reduced in atherosclerotic lesions, in regulating the VSMC inflammatory response. Approach and Results: We used VSMC-lineage tracing to reveal reduced H3K9me2 levels in VSMCs of arteries after injury and in atherosclerotic lesions compared with control vessels. Intriguingly, chromatin immunoprecipitation showed H3K9me2 enrichment at a subset of inflammation-responsive gene promoters, including MMP3, MMP9, MMP12, and IL6, in mouse and human VSMCs. Inhibition of G9A/GLP (G9A-like protein), the primary enzymes responsible for H3K9me2, significantly potentiated inflammation-induced gene induction in vitro and in vivo without altering NFκB (nuclear factor kappa-light-chain-enhancer of activated B cell) and MAPK (mitogen-activated protein kinase) signaling. Rather, reduced G9A/GLP activity enhanced inflammation-induced binding of transcription factors NFκB-p65 and cJUN to H3K9me2 target gene promoters MMP3 and IL6. Taken together, these results suggest that promoter-associated H3K9me2 directly attenuates the induction of target genes in response to inflammation in human VSMCs. CONCLUSIONS This study implicates H3K9me2 in regulating the proinflammatory VSMC phenotype. Our findings suggest that reduced H3K9me2 in disease enhance binding of NFκB and AP-1 (activator protein-1) transcription factors at specific inflammation-responsive genes to augment proinflammatory stimuli in VSMC. Therefore, H3K9me2-regulation could be targeted clinically to limit expression of MMPs and IL6, which are induced in vascular disease.
Collapse
Affiliation(s)
- Jennifer L. Harman
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Lina Dobnikar
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
- Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom (L.D., P.F.-P.)
| | - Joel Chappell
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Benjamin G. Stokell
- Statistical Laboratory, Centre for Mathematical Sciences, University of Cambridge, United Kingdom (B.G.S.)
| | - Amanda Dalby
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Kirsty Foote
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Alison Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | | | - Annabel L. Taylor
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Matthew D. Worssam
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Ralitsa R. Madsen
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Elena Loche
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Anna Uryga
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Martin R. Bennett
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Helle F. Jørgensen
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| |
Collapse
|
34
|
Gordon O, Reis e Sousa C. Cytoskeletal Exposure in the Regulation of Immunity and Initiation of Tissue Repair. Bioessays 2019; 41:e1900021. [PMID: 31157930 DOI: 10.1002/bies.201900021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/25/2019] [Indexed: 01/08/2023]
Abstract
This article reviews and discusses emerging evidence suggesting an evolutionarily-conserved connection between injury-associated exposure of cytoskeletal proteins and the induction of tolerance to infection, repair of tissue damage and restoration of homeostasis. While differences exist between vertebrates and invertebrates with respect to the receptor(s), cell types, and effector mechanisms involved, the response to exposed cytoskeletal proteins appears to be protective and to rely on a conserved signaling cassette involving Src family kinases, the nonreceptor tyrosine kinase Syk, and tyrosine phosphatases. A case is made for research programs that integrate different model organisms in order to increase the understanding of this putative response to tissue damage.
Collapse
Affiliation(s)
- Oliver Gordon
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| |
Collapse
|
35
|
Coll-Tané M, Krebbers A, Castells-Nobau A, Zweier C, Schenck A. Intellectual disability and autism spectrum disorders 'on the fly': insights from Drosophila. Dis Model Mech 2019; 12:dmm039180. [PMID: 31088981 PMCID: PMC6550041 DOI: 10.1242/dmm.039180] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intellectual disability (ID) and autism spectrum disorders (ASD) are frequently co-occurring neurodevelopmental disorders and affect 2-3% of the population. Rapid advances in exome and genome sequencing have increased the number of known implicated genes by threefold, to more than a thousand. The main challenges in the field are now to understand the various pathomechanisms associated with this bewildering number of genetic disorders, to identify new genes and to establish causality of variants in still-undiagnosed cases, and to work towards causal treatment options that so far are available only for a few metabolic conditions. To meet these challenges, the research community needs highly efficient model systems. With an increasing number of relevant assays and rapidly developing novel methodologies, the fruit fly Drosophila melanogaster is ideally positioned to change gear in ID and ASD research. The aim of this Review is to summarize some of the exciting work that already has drawn attention to Drosophila as a model for these disorders. We highlight well-established ID- and ASD-relevant fly phenotypes at the (sub)cellular, brain and behavioral levels, and discuss strategies of how this extraordinarily efficient and versatile model can contribute to 'next generation' medical genomics and to a better understanding of these disorders.
Collapse
Affiliation(s)
- Mireia Coll-Tané
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Alina Krebbers
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Anna Castells-Nobau
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Christiane Zweier
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
36
|
Wang L, Cappelle K, Santos D, Vanden Broeck J, Smagghe G, Swevers L. Short-term persistence precedes pathogenic infection: Infection kinetics of cricket paralysis virus in silkworm-derived Bm5 cells. JOURNAL OF INSECT PHYSIOLOGY 2019; 115:1-11. [PMID: 30905610 DOI: 10.1016/j.jinsphys.2019.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/16/2019] [Accepted: 03/16/2019] [Indexed: 06/09/2023]
Abstract
Next generation sequencing has revealed the widespread occurrence of persistent virus infections in insects but little is known regarding to what extent persistent infections can affect cellular physiology and how they might contribute to the development of disease. In contrast to the pathogenic infections occurring in Drosophila S2 cells, it was observed that Cricket Paralysis virus (CrPV; Dicistroviridae) causes persistent infections in 9 lepidopteran and 2 coleopteran cell lines. The status of the persistent infection was subsequently investigated in more detail using silkworm-derived Bm5 cells, where the infection eventually becomes pathogenic after 3-4 weeks. The short-term persistence period in Bm5 cells is characterized by low levels of viral replication and virion production as well as by the production of viral siRNAs. However, during this period cellular physiology also becomes altered since the cells become susceptible to infection by the nodavirus Flock House virus (FHV). Pathogenicity and widespread mortality at 4 weeks is preceded by a large increase in virion production and the transcriptional activation of immune-related genes encoding RNAi factors and transcription factors in the Toll, Imd and Jak-STAT pathways. During the infection of Bm5 cells, the infective properties of CrPV are not altered, indicating changes in the physiology of the host cells during the transition from short-term persistence to pathogenicity. The in vitro system of Bm5 cells persistently infected with CrPV can therefore be presented as an easily accessible model to study the nature of persistent virus infections and the processes that trigger the transition to pathogenicity, for instance through the application of different "omics" approaches (transcriptomics, proteomics, metabolomics). The different factors that can cause the transition from persistence to pathogenicity in the Bm5-CrPV infection model are discussed.
Collapse
Affiliation(s)
- Luoluo Wang
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
| | - Kaat Cappelle
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
| | - Dulce Santos
- Molecular Developmental Physiology and Signal Transduction, KU Leuven, Leuven, Belgium.
| | - Jozef Vanden Broeck
- Molecular Developmental Physiology and Signal Transduction, KU Leuven, Leuven, Belgium.
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", Athens, Greece.
| |
Collapse
|
37
|
Doering KRS, Taubert S. Epigenetic regulator G9a provides glucose as a sweet key to stress resistance. PLoS Biol 2019; 17:e3000236. [PMID: 31002662 PMCID: PMC6493764 DOI: 10.1371/journal.pbio.3000236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/01/2019] [Indexed: 11/18/2022] Open
Abstract
The ability to adapt to acute and chronic stress is important for organisms to thrive in evolutionary niches and for cells to survive in adverse conditions. The regulatory networks that control stress responses are evolutionarily conserved, and many factors that selectively activate stress responses have been identified. Less well understood are mechanisms that guard against unnecessary induction of cytoprotective factors and that connect stress responses with cellular metabolism to control energy expenditure during stress. The work of Riahi and colleagues represents important progress in this regard because it identifies the histone methyltransferase G9a as a modulator of oxidative stress responses. G9a dampens the expression of antioxidant genes, thus preventing inappropriate energy consumption. Moreover, G9a promotes the well-paced catabolism of storage glycogen and fat during stress. The importance of energy availability during stress is further evidenced by exogenous glucose rescuing the vulnerability of the G9a mutant to oxidative stress. Prior work in multiple model systems has implicated G9a in several other adaptive responses. Therefore, its role in pacing energy consumption and in restraining excessive stress response gene expression under stress may extend to other adaptive responses across species. Stress responses are important for survival and evolutionary adaptation. This Primer explores a study showing that the fruit fly histone methyltransferase G9a (EHMT1/2) couples energy availability to finely tuned regulation of the stress response.
Collapse
Affiliation(s)
- Kelsie R. S. Doering
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Stefan Taubert
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
38
|
The histone methyltransferase G9a regulates tolerance to oxidative stress-induced energy consumption. PLoS Biol 2019; 17:e2006146. [PMID: 30860988 PMCID: PMC6413895 DOI: 10.1371/journal.pbio.2006146] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 02/06/2019] [Indexed: 12/24/2022] Open
Abstract
Stress responses are crucial processes that require activation of genetic programs that protect from the stressor. Stress responses are also energy consuming and can thus be deleterious to the organism. The mechanisms coordinating energy consumption during stress response in multicellular organisms are not well understood. Here, we show that loss of the epigenetic regulator G9a in Drosophila causes a shift in the transcriptional and metabolic responses to oxidative stress (OS) that leads to decreased survival time upon feeding the xenobiotic paraquat. During OS exposure, G9a mutants show overactivation of stress response genes, rapid depletion of glycogen, and inability to access lipid energy stores. The OS survival deficiency of G9a mutants can be rescued by a high-sugar diet. Control flies also show improved OS survival when fed a high-sugar diet, suggesting that energy availability is generally a limiting factor for OS tolerance. Directly limiting access to glycogen stores by knocking down glycogen phosphorylase recapitulates the OS-induced survival defects of G9a mutants. We propose that G9a mutants are sensitive to stress because they experience a net reduction in available energy due to (1) rapid glycogen use, (2) an inability to access lipid energy stores, and (3) an overinduced transcriptional response to stress that further exacerbates energy demands. This suggests that G9a acts as a critical regulatory hub between the transcriptional and metabolic responses to OS. Our findings, together with recent studies that established a role for G9a in hypoxia resistance in cancer cell lines, suggest that G9a is of wide importance in controlling the cellular and organismal response to multiple types of stress. Stress responses require proper activation of genetic programs to protect the organism from the stressor. However, the mechanisms controlling energy consumption during stress responses are not well understood. Here, we investigate the role of epigenetic modifier G9a in regulating metabolism and gene transcription during oxidative stress responses in Drosophila. Flies lacking G9a show a shift in the metabolic and transcriptional responses to oxidative stress, leading to decreased stress tolerance despite intact oxidative stress defense mechanisms. During oxidative stress exposure, G9a mutants show overactivation of stress response and many other genes, rapid depletion of glycogen energy stores, and an inability to access lipid energy stores. The increased susceptibility of G9a mutant flies to oxidative stress can be rescued simply by providing extra sugar. This suggests that G9a mutants are sensitive to stress because of reduced access to immediately available energy. Wild-type flies also become more tolerant to oxidative stress when they are fed extra sugar, whereas blocking energy access by genetically reducing a key metabolic enzyme leads to oxidative stress sensitivity. Though the genetic response to oxidative stress has long been appreciated, our study emphasizes the importance of energy metabolism for stress tolerance and identifies the histone methyltransferase G9a as an important player regulating both.
Collapse
|
39
|
Burgan SC, Gervasi SS, Johnson LR, Martin LB. How Individual Variation in Host Tolerance Affects Competence to Transmit Parasites. Physiol Biochem Zool 2019; 92:49-57. [PMID: 30481116 DOI: 10.1086/701169] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Tolerance, or the maintenance of host health or fitness at a given parasite burden, has often been studied in evolutionary and medical contexts, particularly with respect to effects on the evolution of parasite virulence and individual patient outcomes. These bodies of work have provided insight about tolerance for evolutionary phenomena (e.g., virulence) and individual health (e.g., recovering from an infection). However, due to the specific motivations of that work, few studies have considered the ecological ramifications of variation in tolerance, namely, how variation in forms of tolerance could mediate parasite movement through populations and even community-level disease dynamics. Tolerance is most commonly regarded as the relationship between host fitness and parasite burden. However, few if any studies have actually quantified host fitness, instead utilizing proxies of fitness as the response variables to be regressed against parasite burden. Here, we address how attention to the effects of parasite burden on traits that are relevant to host competence (i.e., the ability to amplify parasites to levels transmissible to other hosts/vectors) will enhance our understanding of disease dynamics in nature. We also provide several forms of guidance for how to overcome the challenges of quantifying tolerance in wild organisms.
Collapse
|
40
|
Merkling SH, Riahi H, Overheul GJ, Schenck A, van Rij RP. Peroxisome-associated Sgroppino links fat metabolism with survival after RNA virus infection in Drosophila. Sci Rep 2019; 9:2065. [PMID: 30765784 PMCID: PMC6375949 DOI: 10.1038/s41598-019-38559-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 12/31/2018] [Indexed: 12/31/2022] Open
Abstract
The fruit fly Drosophila melanogaster is a valuable model organism for the discovery and characterization of innate immune pathways, but host responses to virus infection remain incompletely understood. Here, we describe a novel player in host defense, Sgroppino (Sgp). Genetic depletion of Sgroppino causes hypersensitivity of adult flies to infections with the RNA viruses Drosophila C virus, cricket paralysis virus, and Flock House virus. Canonical antiviral immune pathways are functional in Sgroppino mutants, suggesting that Sgroppino exerts its activity via an as yet uncharacterized process. We demonstrate that Sgroppino localizes to peroxisomes, organelles involved in lipid metabolism. In accordance, Sgroppino-deficient flies show a defect in lipid metabolism, reflected by higher triglyceride levels, higher body mass, and thicker abdominal fat tissue. In addition, knock-down of Pex3, an essential peroxisome biogenesis factor, increases sensitivity to virus infection. Together, our results establish a genetic link between the peroxisomal protein Sgroppino, fat metabolism, and resistance to virus infection.
Collapse
Affiliation(s)
- Sarah H Merkling
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Insect-Virus Interactions Group, Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - Human Riahi
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
41
|
Ruiz JL, Yerbanga RS, Lefèvre T, Ouedraogo JB, Corces VG, Gómez-Díaz E. Chromatin changes in Anopheles gambiae induced by Plasmodium falciparum infection. Epigenetics Chromatin 2019; 12:5. [PMID: 30616642 PMCID: PMC6322293 DOI: 10.1186/s13072-018-0250-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/19/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Infection by the human malaria parasite leads to important changes in mosquito phenotypic traits related to vector competence. However, we still lack a clear understanding of the underlying mechanisms and, in particular, of the epigenetic basis for these changes. We have examined genome-wide distribution maps of H3K27ac, H3K9ac, H3K9me3 and H3K4me3 by ChIP-seq and the transcriptome by RNA-seq, of midguts from Anopheles gambiae mosquitoes blood-fed uninfected and infected with natural isolates of the human malaria parasite Plasmodium falciparum in Burkina Faso. RESULTS We report 15,916 regions containing differential histone modification enrichment between infected and uninfected, of which 8339 locate at promoters and/or intersect with genes. The functional annotation of these regions allowed us to identify infection-responsive genes showing differential enrichment in various histone modifications, such as CLIP proteases, antimicrobial peptides-encoding genes, and genes related to melanization responses and the complement system. Further, the motif analysis of regions differentially enriched in various histone modifications predicts binding sites that might be involved in the cis-regulation of these regions, such as Deaf1, Pangolin and Dorsal transcription factors (TFs). Some of these TFs are known to regulate immunity gene expression in Drosophila and are involved in the Notch and JAK/STAT signaling pathways. CONCLUSIONS The analysis of malaria infection-induced chromatin changes in mosquitoes is important not only to identify regulatory elements and genes underlying mosquito responses to P. falciparum infection, but also for possible applications to the genetic manipulation of mosquitoes and to other mosquito-borne systems.
Collapse
Affiliation(s)
- José L. Ruiz
- Estación Biológica de Doñana (EBD), Consejo Superior de Investigaciones Científicas, 41092 Seville, Spain
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas, 18016 Granada, Spain
| | - Rakiswendé S. Yerbanga
- Institut de Recherche en Sciences de la Santé (IRSS), 01 BP 171, Bobo Dioulasso, Burkina Faso
| | - Thierry Lefèvre
- Institut de Recherche en Sciences de la Santé (IRSS), 01 BP 171, Bobo Dioulasso, Burkina Faso
- MIVEGEC, IRD, CNRS, University of Montpellier, Montpellier, France
| | - Jean B. Ouedraogo
- Institut de Recherche en Sciences de la Santé (IRSS), 01 BP 171, Bobo Dioulasso, Burkina Faso
| | - Victor G. Corces
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322 USA
| | - Elena Gómez-Díaz
- Estación Biológica de Doñana (EBD), Consejo Superior de Investigaciones Científicas, 41092 Seville, Spain
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas, 18016 Granada, Spain
| |
Collapse
|
42
|
Mondotte JA, Gausson V, Frangeul L, Blanc H, Lambrechts L, Saleh MC. Immune priming and clearance of orally acquired RNA viruses in Drosophila. Nat Microbiol 2018; 3:1394-1403. [PMID: 30374170 DOI: 10.1038/s41564-018-0265-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/06/2018] [Indexed: 12/17/2022]
Abstract
Immune responses in insects are differentially triggered depending on the infection route used by the pathogen. In most studies involving Drosophila melanogaster and viruses, infection is done by injection, while oral infection, which is probably the most common route of viral entry in nature, remains unexplored. Here, we orally infected adults and larvae from wild-type and RNA interference (RNAi) mutant flies with different RNA viruses. We found that, in contrast with what is observed following virus injection, oral infections initiated at larval or adult stages are cleared in adult flies. Virus elimination occurred despite a larger infectious dose than for injected flies and evidence of viral replication. RNAi mutant flies suffered greater mortality relative to wild-type flies following oral infection, but they also eliminated the virus, implying that RNAi is not essential for viral clearance and that other immune mechanisms act during oral infections. We further showed that information of infection by RNA viruses acquired orally leaves a trace under a DNA form, which confers protection against future reinfection by the same virus. Together, this work presents evidence of clearance and immune priming for RNA viruses in insects and challenges the current view of antiviral immunity in insects.
Collapse
Affiliation(s)
- Juan A Mondotte
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Valérie Gausson
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Lionel Frangeul
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Hervé Blanc
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Louis Lambrechts
- Institut Pasteur, Insect-Virus Interactions Group, Department of Genomes and Genetics, CNRS Unité Mixte de Recherche 2000, Paris, France
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France.
| |
Collapse
|
43
|
Lissner MM, Schneider DS. The physiological basis of disease tolerance in insects. CURRENT OPINION IN INSECT SCIENCE 2018; 29:133-136. [PMID: 30551820 DOI: 10.1016/j.cois.2018.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 09/10/2018] [Indexed: 06/09/2023]
Abstract
Immunology textbooks teach us about the ways hosts can recognize and kill microbes but leave out something important: the mechanisms used to survive infections. Survival depends on more than simply detecting and eliminating microbes; it requires that we prevent and repair the damage caused by pathogens and the immune response. Recent work in insects is helping to build our understanding of this aspect of pathology, called disease tolerance. Here we discuss papers that explore disease tolerance using theoretical, population genetics, and mechanistic approaches.
Collapse
Affiliation(s)
- Michelle M Lissner
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, United States
| | - David S Schneider
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, United States.
| |
Collapse
|
44
|
Goto A, Okado K, Martins N, Cai H, Barbier V, Lamiable O, Troxler L, Santiago E, Kuhn L, Paik D, Silverman N, Holleufer A, Hartmann R, Liu J, Peng T, Hoffmann JA, Meignin C, Daeffler L, Imler JL. The Kinase IKKβ Regulates a STING- and NF-κB-Dependent Antiviral Response Pathway in Drosophila. Immunity 2018; 49:225-234.e4. [PMID: 30119996 DOI: 10.1016/j.immuni.2018.07.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/08/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022]
Abstract
Antiviral immunity in Drosophila involves RNA interference and poorly characterized inducible responses. Here, we showed that two components of the IMD pathway, the kinase dIKKβ and the transcription factor Relish, were required to control infection by two picorna-like viruses. We identified a set of genes induced by viral infection and regulated by dIKKβ and Relish, which included an ortholog of STING. We showed that dSTING participated in the control of infection by picorna-like viruses, acting upstream of dIKKβ to regulate expression of Nazo, an antiviral factor. Our data reveal an antiviral function for STING in an animal model devoid of interferons and suggest an evolutionarily ancient role for this molecule in antiviral immunity.
Collapse
Affiliation(s)
- Akira Goto
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| | - Kiyoshi Okado
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Nelson Martins
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Hua Cai
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Vincent Barbier
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Olivier Lamiable
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Laurent Troxler
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Estelle Santiago
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Lauriane Kuhn
- Université de Strasbourg, CNRS, Plateforme Protéomique Strasbourg-Esplanade, 67000 Strasbourg, France
| | - Donggi Paik
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Neal Silverman
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Andreas Holleufer
- Center for Structural Biology, Aarhus University, 8000 Aarhus C, Denmark
| | - Rune Hartmann
- Center for Structural Biology, Aarhus University, 8000 Aarhus C, Denmark
| | - Jiyong Liu
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Tao Peng
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Jules A Hoffmann
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France
| | - Carine Meignin
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Laurent Daeffler
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
45
|
Palmer WH, Medd NC, Beard PM, Obbard DJ. Isolation of a natural DNA virus of Drosophila melanogaster, and characterisation of host resistance and immune responses. PLoS Pathog 2018; 14:e1007050. [PMID: 29864164 PMCID: PMC6002114 DOI: 10.1371/journal.ppat.1007050] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 06/14/2018] [Accepted: 04/20/2018] [Indexed: 12/15/2022] Open
Abstract
Drosophila melanogaster has played a key role in our understanding of invertebrate immunity. However, both functional and evolutionary studies of host-virus interaction in Drosophila have been limited by a dearth of native virus isolates. In particular, despite a long history of virus research, DNA viruses of D. melanogaster have only recently been described, and none have been available for experimental study. Here we report the isolation and comprehensive characterisation of Kallithea virus, a large double-stranded DNA virus, and the first DNA virus to have been reported from wild populations of D. melanogaster. We find that Kallithea virus infection is costly for adult flies, reaching high titres in both sexes and disproportionately reducing survival in males, and movement and late fecundity in females. Using the Drosophila Genetic Reference Panel, we quantify host genetic variance for virus-induced mortality and viral titre and identify candidate host genes that may underlie this variation, including Cdc42-interacting protein 4. Using full transcriptome sequencing of infected males and females, we examine the transcriptional response of flies to Kallithea virus infection and describe differential regulation of virus-responsive genes. This work establishes Kallithea virus as a new tractable model to study the natural interaction between D. melanogaster and DNA viruses, and we hope it will serve as a basis for future studies of immune responses to DNA viruses in insects.
Collapse
Affiliation(s)
- William H Palmer
- Institute of Evolutionary Biology University of Edinburgh, Charlotte Auerbach Road, Edinburgh, United Kingdom
| | - Nathan C Medd
- Institute of Evolutionary Biology University of Edinburgh, Charlotte Auerbach Road, Edinburgh, United Kingdom
| | - Philippa M Beard
- The Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - Darren J Obbard
- Institute of Evolutionary Biology University of Edinburgh, Charlotte Auerbach Road, Edinburgh, United Kingdom
- Centre for Infection, Evolution and Immunity, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
46
|
Swevers L, Liu J, Smagghe G. Defense Mechanisms against Viral Infection in Drosophila: RNAi and Non-RNAi. Viruses 2018; 10:E230. [PMID: 29723993 PMCID: PMC5977223 DOI: 10.3390/v10050230] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/20/2018] [Accepted: 04/27/2018] [Indexed: 12/20/2022] Open
Abstract
RNAi is considered a major antiviral defense mechanism in insects, but its relative importance as compared to other antiviral pathways has not been evaluated comprehensively. Here, it is attempted to give an overview of the antiviral defense mechanisms in Drosophila that involve both RNAi and non-RNAi. While RNAi is considered important in most viral infections, many other pathways can exist that confer antiviral resistance. It is noted that very few direct recognition mechanisms of virus infections have been identified in Drosophila and that the activation of immune pathways may be accomplished indirectly through cell damage incurred by viral replication. In several cases, protection against viral infection can be obtained in RNAi mutants by non-RNAi mechanisms, confirming the variability of the RNAi defense mechanism according to the type of infection and the physiological status of the host. This analysis is aimed at more systematically investigating the relative contribution of RNAi in the antiviral response and more specifically, to ask whether RNAi efficiency is affected when other defense mechanisms predominate. While Drosophila can function as a useful model, this issue may be more critical for economically important insects that are either controlled (agricultural pests and vectors of diseases) or protected from parasite infection (beneficial insects as bees) by RNAi products.
Collapse
Affiliation(s)
- Luc Swevers
- Institute of Biosciences & Applications, NCSR "Demokritos", 15341 Athens, Greece.
| | - Jisheng Liu
- School of Life Sciences, Guangzhou University, 510006 Guangzhou, China.
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
47
|
Invertebrate Iridoviruses: A Glance over the Last Decade. Viruses 2018; 10:v10040161. [PMID: 29601483 PMCID: PMC5923455 DOI: 10.3390/v10040161] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 02/06/2023] Open
Abstract
Members of the family Iridoviridae (iridovirids) are large dsDNA viruses that infect both invertebrate and vertebrate ectotherms and whose symptoms range in severity from minor reductions in host fitness to systemic disease and large-scale mortality. Several characteristics have been useful for classifying iridoviruses; however, novel strains are continuously being discovered and, in many cases, reliable classification has been challenging. Further impeding classification, invertebrate iridoviruses (IIVs) can occasionally infect vertebrates; thus, host range is often not a useful criterion for classification. In this review, we discuss the current classification of iridovirids, focusing on genomic and structural features that distinguish vertebrate and invertebrate iridovirids and viral factors linked to host interactions in IIV6 (Invertebrate iridescent virus 6). In addition, we show for the first time how complete genome sequences of viral isolates can be leveraged to improve classification of new iridovirid isolates and resolve ambiguous relations. Improved classification of the iridoviruses may facilitate the identification of genus-specific virulence factors linked with diverse host phenotypes and host interactions.
Collapse
|
48
|
Abstract
The power and ease of Drosophila genetics and the medical relevance of mosquito-transmitted viruses have made dipterans important model organisms in antiviral immunology. Studies of virus-host interactions at the molecular and population levels have illuminated determinants of resistance to virus infection. Here, we review the sources and nature of variation in antiviral immunity and virus susceptibility in model dipteran insects, specifically the fruit fly Drosophila melanogaster and vector mosquitoes of the genera Aedes and Culex. We first discuss antiviral immune mechanisms and describe the virus-specificity of these responses. In the following sections, we review genetic and microbiota-dependent variation in antiviral immunity. In the final sections, we explore less well-studied sources of variation, including abiotic factors, sexual dimorphism, infection history, and endogenous viral elements. We borrow from work on other pathogen types and non-dipteran species when it parallels or complements studies in dipterans. Understanding natural variation in virus-host interactions may lead to the identification of novel restriction factors and immune mechanisms and shed light on the molecular determinants of vector competence.
Collapse
Affiliation(s)
- William H Palmer
- Institute of Evolutionary Biology and Centre for Infection, Evolution and Immunity, University of Edinburgh, Edinburgh EH9 3FL UK.
| | - Finny S Varghese
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| |
Collapse
|
49
|
Mondotte JA, Saleh MC. Antiviral Immune Response and the Route of Infection in Drosophila melanogaster. Adv Virus Res 2017; 100:247-278. [PMID: 29551139 DOI: 10.1016/bs.aivir.2017.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The use of Drosophila as a model organism has made an important contribution to our understanding of the function and regulation of innate immunity in insects. Indeed, insects can discriminate between different types of pathogens and mount specific and effective responses. Strikingly, the same pathogen can trigger a different immune response in the same organism, depending solely on the route of infection by which the pathogen is delivered. In this review, we recapitulate what is known about antiviral responses in Drosophila, and how they are triggered depending on the route and the mode used for the virus to infect its host.
Collapse
Affiliation(s)
- Juan A Mondotte
- Institut Pasteur, Viruses and RNA Interference Unit, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, CNRS Unité Mixte de Recherche 3569, Paris, France.
| |
Collapse
|
50
|
Koemans TS, Kleefstra T, Chubak MC, Stone MH, Reijnders MRF, de Munnik S, Willemsen MH, Fenckova M, Stumpel CTRM, Bok LA, Sifuentes Saenz M, Byerly KA, Baughn LB, Stegmann APA, Pfundt R, Zhou H, van Bokhoven H, Schenck A, Kramer JM. Functional convergence of histone methyltransferases EHMT1 and KMT2C involved in intellectual disability and autism spectrum disorder. PLoS Genet 2017; 13:e1006864. [PMID: 29069077 PMCID: PMC5656305 DOI: 10.1371/journal.pgen.1006864] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/10/2017] [Indexed: 11/18/2022] Open
Abstract
Kleefstra syndrome, caused by haploinsufficiency of euchromatin histone methyltransferase 1 (EHMT1), is characterized by intellectual disability (ID), autism spectrum disorder (ASD), characteristic facial dysmorphisms, and other variable clinical features. In addition to EHMT1 mutations, de novo variants were reported in four additional genes (MBD5, SMARCB1, NR1I3, and KMT2C), in single individuals with clinical characteristics overlapping Kleefstra syndrome. Here, we present a novel cohort of five patients with de novo loss of function mutations affecting the histone methyltransferase KMT2C. Our clinical data delineates the KMT2C phenotypic spectrum and reinforces the phenotypic overlap with Kleefstra syndrome and other related ID disorders. To elucidate the common molecular basis of the neuropathology associated with mutations in KMT2C and EHMT1, we characterized the role of the Drosophila KMT2C ortholog, trithorax related (trr), in the nervous system. Similar to the Drosophila EHMT1 ortholog, G9a, trr is required in the mushroom body for short term memory. Trr ChIP-seq identified 3371 binding sites, mainly in the promoter of genes involved in neuronal processes. Transcriptional profiling of pan-neuronal trr knockdown and G9a null mutant fly heads identified 613 and 1123 misregulated genes, respectively. These gene sets show a significant overlap and are associated with nearly identical gene ontology enrichments. The majority of the observed biological convergence is derived from predicted indirect target genes. However, trr and G9a also have common direct targets, including the Drosophila ortholog of Arc (Arc1), a key regulator of synaptic plasticity. Our data highlight the clinical and molecular convergence between the KMT2 and EHMT protein families, which may contribute to a molecular network underlying a larger group of ID/ASD-related disorders. Neurodevelopmental disorders (NDDs) like intellectual disability (ID) and autism spectrum disorder (ASD) present an enormous challenge to affected individuals, their families, and society. Understanding the mechanisms underlying NDDs may lead to the development of targeted therapeutics, but this is complicated by the great clinical and genetic heterogeneity seen in patients. Mutations in hundreds of genes have been implicated in NDDs, giving rise to diverse clinical presentations. However, evidence suggests that many of these genes lie in common biological pathways, and mutations in genes that are involved in similar biological functions give rise to more similar clinical phenotypes. Here, we define a novel ID disorder with comorbid ASD (ID/ASD) caused by mutations in KMT2C. This disorder is defined by clinical features that overlap with a group of other disorders, including Kleefstra syndrome, which is caused by EHMT1 mutations. In the fruit fly, we show that the KMT2 and EHMT protein families regulate a highly converging set of biological processes. Both EHMT1 and KMT2C encode histone methyltransferases, which regulate gene transcription by modifying chromatin structure. Further understanding of the common gene regulatory networks associated with this group of ID- and ASD-related disorders may lead to the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Tom S. Koemans
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Tjitske Kleefstra
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Melissa C. Chubak
- Department of Biology, Faculty of Science, Western University, London, Ontario, Canada
| | - Max H. Stone
- Department of Biology, Faculty of Science, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children’s Health Research Institute, London, Ontario, Canada
| | - Margot R. F. Reijnders
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Sonja de Munnik
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Marjolein H. Willemsen
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Michaela Fenckova
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Connie T. R. M. Stumpel
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Levinus A. Bok
- Department of Pediatrics, Máxima Medical Centre, Veldhoven, The Netherlands
| | | | - Kyna A. Byerly
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Linda B. Baughn
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Alexander P. A. Stegmann
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Rolph Pfundt
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Molecular Developmental Biology, Radboud University, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
| | - Annette Schenck
- Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Nijmegen, The Netherlands
- * E-mail: (AS); (JMK)
| | - Jamie M. Kramer
- Department of Biology, Faculty of Science, Western University, London, Ontario, Canada
- Division of Genetics and Development, Children’s Health Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- * E-mail: (AS); (JMK)
| |
Collapse
|