1
|
Wani KA, Pukkila-Worley R. Evolutionarily ancient functions of enzymatic TIR proteins in innate immunity. Trends Immunol 2025:S1471-4906(25)00116-4. [PMID: 40393889 DOI: 10.1016/j.it.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
Proteins with a Toll/interleukin-1 receptor/resistance (TIR) domain are among the most ancient immune regulators and include well-known pattern recognition receptors (PRRs). A specialized subset of TIR domain proteins are enzymes that predominantly use nicotinamide adenine dinucleotide (NAD+) to generate second messenger metabolites. These enzymatic TIR proteins have essential roles in bacteria, plant, and animal immunity. The mechanism of activation of these TIR proteins, conserved across Kingdoms, involves oligomerization into higher-ordered structures, which activates their intrinsic enzymatic activity. Here, we review the functions of enzymatic TIR proteins in innate immunity in bacteria, plants, and animals. This work offers insights into the evolutionary origins of immunity itself and defines fundamental principles of immune surveillance across the Tree of Life.
Collapse
Affiliation(s)
- Khursheed A Wani
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| | - Read Pukkila-Worley
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
2
|
Chauhan M, Martinak PE, Hollenberg BM, Goodman AG. Drosophila melanogaster Toll-9 elicits antiviral immunity against Drosophila C virus. J Virol 2025:e0221424. [PMID: 40366172 DOI: 10.1128/jvi.02214-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
The Toll pathway plays a pivotal role in innate immune responses against pathogens. The evolutionarily conserved pattern recognition receptors (PRRs), including Toll-like receptors (TLRs), play a crucial role in recognition of pathogen-associated molecular patterns (PAMPs). The Drosophila genome encodes nine Toll receptors that are orthologous to mammalian TLRs. While mammalian TLRs directly recognize PAMPs, most Drosophila Tolls recognize the proteolytically cleaved ligand Spätzle to activate downstream signaling cascades. In this study, we demonstrated that Toll-9 is crucial for antiviral immunity against Drosophila C virus (DCV), a natural pathogen of Drosophila. A transposable element insertion in the Toll-9 gene renders the flies more susceptible to DCV. The stable expression of Toll-9 in Drosophila S2 cells results in increased Dicer2 induction and reduced AKT phosphorylation, collectively establishing an antiviral state that inhibits DCV replication. Toll-9 localizes to endosomes, where it binds viral double-stranded RNA (dsRNA), highlighting its role in detecting viral replication intermediates. Together, these findings identify Toll-9 as a key player in antiviral immunity against DCV infection, acting through its ability to recognize dsRNA and drive Dicer2 expression, along with other AKT-mediated antiviral responses. IMPORTANCE Insects rely on innate immunity and RNA interference (RNAi) to combat viral infections. Our study underscores the pivotal role of Drosophila Toll-9 in antiviral immunity, aligning with findings in Bombyx mori, where Toll-9 activation upregulates the RNAi component Dicer2. We demonstrate that Drosophila Toll-9 functions as a pattern recognition receptor (PRR) for double-stranded RNA (dsRNA) during Drosophila C virus (DCV) infection, akin to mammalian Toll-like receptors (TLRs). Toll-9 activation during DCV infection leads to the upregulation of Dicer2 and Argonaute2 and dephosphorylation of AKT. This study also reveals that Toll-9 localizes in endosomal compartments where it interacts with dsRNA. These insights enhance our understanding of Drosophila innate immune mechanisms, reflecting the evolutionary conservation of immune responses across diverse species and providing impetus for further research into the conserved roles of TLRs across the animal kingdom.
Collapse
Affiliation(s)
- Manish Chauhan
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Peter E Martinak
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Benjamin M Hollenberg
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
| | - Alan G Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
3
|
Xia J, Peng R, Fei S, Awais MM, Lai W, Huang Y, Wu H, Yu Y, Liang L, Swevers L, Sun J, Feng M. Systematic analysis of innate immune-related genes in the silkworm: Application to antiviral research. INSECT SCIENCE 2025; 32:151-171. [PMID: 38571329 DOI: 10.1111/1744-7917.13363] [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: 09/12/2023] [Revised: 02/21/2024] [Accepted: 03/01/2024] [Indexed: 04/05/2024]
Abstract
The silkworm, a crucial model organism of the Lepidoptera, offers an excellent platform for investigating the molecular mechanisms underlying the innate immune response of insects toward pathogens. Over the years, researchers worldwide have identified numerous immune-related genes in silkworms. However, these identified silkworm immune genes are not well classified and not well known to the scientific community. With the availability of the latest genome data of silkworms and the extensive research on silkworm immunity, it has become imperative to systematically categorize the immune genes of silkworms with different database IDs. In this study, we present a meticulous organization of prevalent immune-related genes in the domestic silkworm, using the SilkDB 3.0 database as a reliable source for updated gene information. Furthermore, utilizing the available data, we classify the collected immune genes into distinct categories: pattern recognition receptors, classical immune pathways, effector genes and others. In-depth data analysis has enabled us to predict some potential antiviral genes. Subsequently, we performed antiviral experiments on selected genes, exploring their impact on Bombyx mori nucleopolyhedrovirus replication. The outcomes of this research furnish novel insights into the immune genes of the silkworm, consequently fostering advancements in the field of silkworm immunity research by establishing a comprehensive classification and functional understanding of immune-related genes in the silkworm. This study contributes to the broader understanding of insect immune responses and opens up new avenues for future investigations in the domain of host-pathogen interactions.
Collapse
Affiliation(s)
- Junming Xia
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ruoxuan Peng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shigang Fei
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mian Muhammad Awais
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wenxuan Lai
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yigui Huang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hailin Wu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yue Yu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lingying Liang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, National Center for Scientific Research Demokritos, Institute of Biosciences and Applications, Athens, Greece
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
4
|
Rhodes VL, Waterhouse RM, Michel K. The molecular toll pathway repertoire in anopheline mosquitoes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105287. [PMID: 39522894 PMCID: PMC11717629 DOI: 10.1016/j.dci.2024.105287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Innate immunity in mosquitoes has received much attention due to its potential impact on vector competence for vector-borne disease pathogens, including malaria parasites. The nuclear factor (NF)-κB-dependent Toll pathway is a major regulator of innate immunity in insects. In mosquitoes, this pathway controls transcription of the majority of the known canonical humoral immune effectors, mediates anti-bacterial, anti-fungal and anti-viral immune responses, and contributes to malaria parasite killing. However, besides initial gene annotation of putative Toll pathway members and genetic analysis of the contribution of few key components to immunity, the molecular make-up and function of the Toll pathway in mosquitoes is largely unexplored. To facilitate functional analyses of the Toll pathway in mosquitoes, we report here manually annotated and refined gene models of Toll-like receptors and all putative components of the intracellular signal transduction cascade across 19 anopheline genomes, and in two culicine genomes. In addition, based on phylogenetic analyses, we identified differing levels of evolutionary constraint across the intracellular Toll pathway members, and identified a recent radiation of TOLL1/5 within the Anopheles gambiae complex. Together, this study provides insight into the evolution of TLRs and the putative members of the intracellular signal transduction cascade within the genus Anopheles.
Collapse
Affiliation(s)
- Victoria L Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA; Biology Department, Missouri Southern University, Joplin, MO 64801, USA
| | | | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
5
|
Aalto AL, Luukkonen V, Meinander A. Ubiquitin signalling in Drosophila innate immune responses. FEBS J 2024; 291:4397-4413. [PMID: 38069549 DOI: 10.1111/febs.17028] [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: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Cells respond to invading pathogens and danger signals from the environment by adapting gene expression to meet the need for protective effector molecules. While this innate immune response is required for the cell and the organism to recover, excess immune activation may lead to loss of homeostasis, thereby promoting chronic inflammation and cancer progression. The molecular basis of innate immune defence is comprised of factors promoting survival and proliferation, such as cytokines, antimicrobial peptides and anti-apoptotic proteins. As the molecular mechanisms regulating innate immune responses are conserved through evolution, the fruit fly Drosophila melanogaster serves as a convenient, affordable and ethical model organism to enhance understanding of immune signalling. Fly immunity against bacterial infection is built up by both cellular and humoral responses, where the latter is regulated by the Imd and Toll pathways activating NF-κB transcription factors Relish, Dorsal and Dif, as well as JNK activation and JAK/STAT signalling. As in mammals, the Drosophila innate immune signalling pathways are characterised by ubiquitination of signalling molecules followed by ubiquitin receptors binding to the ubiquitin chains, as well as by rapid changes in protein levels by ubiquitin-mediated targeted proteasomal and lysosomal degradation. In this review, we summarise the molecular signalling pathways regulating immune responses to pathogen infection in Drosophila, with a focus on ubiquitin-dependent control of innate immunity and inflammatory signalling.
Collapse
Affiliation(s)
- Anna L Aalto
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Veera Luukkonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| |
Collapse
|
6
|
Rhodes VL, Waterhouse RM, Michel K. The Molecular Toll Pathway Repertoire in Anopheline Mosquitoes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612760. [PMID: 39345384 PMCID: PMC11429875 DOI: 10.1101/2024.09.12.612760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Innate immunity in mosquitoes has received much attention due to its potential impact on vector competence for vector-borne disease pathogens, including malaria parasites. The nuclear factor (NF)-κB-dependent Toll pathway is a major regulator of innate immunity in insects. In mosquitoes, this pathway controls transcription of the majority of the known canonical humoral immune effectors, mediates anti-bacterial, anti-fungal and anti-viral immune responses, and contributes to malaria parasite killing. However, besides initial gene annotation of putative Toll pathway members and genetic analysis of the contribution of few key components to immunity, the molecular make-up and function of the Toll pathway in mosquitoes is largely unexplored. To facilitate functional analyses of the Toll pathway in mosquitoes, we report here manually annotated and refined gene models of Toll-like receptors and all putative components of the intracellular signal transduction cascade across 19 anopheline genomes, and in two culicine genomes. In addition, based on phylogenetic analyses, we identified differing levels of evolutionary constraint across the intracellular Toll pathway members, and identified a recent radiation of TOLL1/5 within the An. gambiae complex. Together, this study provides insight into the evolution of TLRs and the putative members of the intracellular signal transduction cascade within the genus Anopheles.
Collapse
Affiliation(s)
- Victoria L. Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
- Biology Department, Missouri Southern University, Joplin, MO 64801, USA
| | | | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
7
|
Chauhan M, Martinak PE, Hollenberg BM, Goodman AG. Drosophila melanogaster Toll-9 elicits antiviral immunity against Drosophila C virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599730. [PMID: 38948804 PMCID: PMC11212974 DOI: 10.1101/2024.06.19.599730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Toll pathway plays a pivotal role in innate immune responses against pathogens. The evolutionary conserved pathogen recognition receptors (PRRs), including Toll like receptors (TLRs), play a crucial role in recognition of pathogen associated molecular patterns (PAMPs). The Drosophila genome encodes nine Toll receptors that are orthologous to mammalian TLRs. While mammalian TLRs directly recognize PAMPs, most Drosophila Tolls recognize the proteolytically cleaved ligand Spätzle to activate downstream signaling cascades. In this study, we demonstrated that Toll-9 is crucial for antiviral immunity against Drosophila C virus (DCV), a natural pathogen of Drosophila . A transposable element insertion in the Toll-9 gene renders the flies more susceptible to DCV. The stable expression of Toll-9 in S2 cells confers resistance against DCV infection by upregulation of the RNAi pathway. Toll-9 promotes the dephosphorylation of AKT, resulting in the induction of antiviral RNAi genes to inhibit DCV replication. Toll-9 localizes to the endosome where it binds dsRNA, suggesting its role to detect viral dsRNA. Toll-9 also induces apoptosis during DCV infection, contributing to its antiviral role. Together, this work identifies the role of Toll-9 in antiviral immunity against DCV infection through its ability to bind dsRNA and induce AKT-mediated RNAi antiviral immunity. IMPORTANCE Insects rely on innate immunity and RNA interference (RNAi) to combat viral infections. Our study underscores the pivotal role of Drosophila Toll-9 in antiviral immunity, aligning with findings in Bombyx mori , where Toll-9 activation upregulates the RNAi component Dicer2 . We demonstrate that Drosophila Toll-9 functions as a pattern recognition receptor (PRR) for double-stranded RNA (dsRNA) during Drosophila C virus (DCV) infection, akin to mammalian TLRs. Toll-9 activation leads to the upregulation of key RNAi components, Dicer2 and Argonaute2 , and dephosphorylation of AKT triggers apoptosis via induction of proapoptotic genes Hid and Reaper . This study also reveals that Toll-9 localizes in endosomal compartments where it interacts with dsRNA. These insights enhance our understanding of Drosophila innate immune mechanisms, reflecting the evolutionary conservation of immune responses across diverse species and providing impetus for further research into the conserved roles of TLRs across the animal kingdom.
Collapse
|
8
|
Bossen J, Kühle JP, Roeder T. The tracheal immune system of insects - A blueprint for understanding epithelial immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 157:103960. [PMID: 37235953 DOI: 10.1016/j.ibmb.2023.103960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
The unique design of respiratory organs in multicellular organisms makes them prone to infection by pathogens. To cope with this vulnerability, highly effective local immune systems evolved that are also operative in the tracheal system of insects. Many pathogens and parasites (including viruses, bacteria, fungi, and metazoan parasites) colonize the trachea or invade the host via this route. Currently, only two modules of the tracheal immune system have been characterized in depth: 1) Immune deficiency pathway-mediated activation of antimicrobial peptide gene expression and 2) local melanization processes that protect the structure from wounding. There is an urgent need to increase our understanding of the architecture of tracheal immune systems, especially regarding those mechanisms that enable the maintenance of immune homeostasis. This need for new studies is particularly exigent for species other than Drosophila.
Collapse
Affiliation(s)
- Judith Bossen
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Jan-Philip Kühle
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany
| | - Thomas Roeder
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
9
|
Abbas MN, Kausar S, Gul I, Li J, Yu H, Dong M, Cui H. The Potential Biological Roles of Circular RNAs in the Immune Systems of Insects to Pathogen Invasion. Genes (Basel) 2023; 14:genes14040895. [PMID: 37107653 PMCID: PMC10137924 DOI: 10.3390/genes14040895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Circular RNAs (circRNAs) are a newly discovered class of endogenously expressed non-coding RNAs (ncRNAs). They are highly stable, covalently closed molecules that frequently exhibit tissue-specific expression in eukaryotes. A small number of circRNAs are abundant and have been remarkably conserved throughout evolution. Numerous circRNAs are known to play important biological roles by acting as microRNAs (miRNAs) or protein inhibitors ('sponges'), by regulating the function of proteins, or by being translated themselves. CircRNAs have distinct cellular functions due to structural and production differences from mRNAs. Recent advances highlight the importance of characterizing circRNAs and their targets in a variety of insect species in order to fully understand how they contribute to the immune responses of these insects. Here, we focus on the recent advances in our understanding of the biogenesis of circRNAs, regulation of their abundance, and biological roles, such as serving as templates for translation and in the regulation of signaling pathways. We also discuss the emerging roles of circRNAs in regulating immune responses to various microbial pathogens. Furthermore, we describe the functions of circRNAs encoded by microbial pathogens that play in their hosts.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Saima Kausar
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Isma Gul
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Jisheng Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Huijuan Yu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Mengyao Dong
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
10
|
Gao J, Guo Y, Chen J, Diao QY, Wang Q, Dai PL, Zhang L, Li WM, Wu YY. Acute oral toxicity, apoptosis, and immune response in nurse bees ( Apis mellifera) induced by flupyradifurone. Front Physiol 2023; 14:1150340. [PMID: 37057182 PMCID: PMC10086230 DOI: 10.3389/fphys.2023.1150340] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
The potential toxicity of flupyradifurone (FPF) to honey bees has been a subject of controversy in recent years. Understanding the effect of pesticides on nurse bees is important because the fitness of nurse bees is critical for in-hive activities, such as larval survival and performing hive maintenance. In order to evaluate the acute oral toxicity of flupyradifurone on nurse bees, flupyradifurone at five different concentrations was selected to feed both larvae and nurse bees. Our results showed that nurse bees were more sensitive to flupyradifurone than larvae (LD50 of the acute oral toxicity of flupyradifurone was 17.72 μg a.i./larva and 3.368 μg a.i./nurse bee). In addition, the apoptotic rates of neurons in mushroom bodies of nurse bees were significantly induced by flupyradifurone at sublethal concentrations (8 mg/L, 20 mg/L, and 50 mg/L) and the median lethal concentration LC50 (125 mg/L). The expression of immune-related genes (Hsp90, Toll-8/Tollo, and defensin) was significantly changed in exposed nurse bees at the field-realistic concentration of flupyradifurone. However, three detoxifying enzyme genes (CYP9Q1, -2, and -3) were not affected by pesticide exposure. Our data suggest that although flupyradifurone had a relatively lower acute oral toxicity than many other common pesticides, exposures to the field-realistic and other sublethal concentrations of flupyradifurone still have cytotoxicity and immune-responsive effects on nurse bees. Therefore, flupyradifurone should be considered for its application in crops.
Collapse
Affiliation(s)
- Jing Gao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yi Guo
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jin Chen
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qing-Yun Diao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiang Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ping-Li Dai
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Li Zhang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wen-Min Li
- College of Life Sciences and Agriculture and Forestry, Qiqihar University, Qiqihar, China
| | - Yan-Yan Wu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
11
|
Bossen J, Prange R, Kühle JP, Künzel S, Niu X, Hammel JU, Krieger L, Knop M, Ehrhardt B, Uliczka K, Krauss-Etschmann S, Roeder T. Adult and Larval Tracheal Systems Exhibit Different Molecular Architectures in Drosophila. Int J Mol Sci 2023; 24:ijms24065628. [PMID: 36982710 PMCID: PMC10052349 DOI: 10.3390/ijms24065628] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Knowing the molecular makeup of an organ system is required for its in-depth understanding. We analyzed the molecular repertoire of the adult tracheal system of the fruit fly Drosophila melanogaster using transcriptome studies to advance our knowledge of the adult insect tracheal system. Comparing this to the larval tracheal system revealed several major differences that likely influence organ function. During the transition from larval to adult tracheal system, a shift in the expression of genes responsible for the formation of cuticular structure occurs. This change in transcript composition manifests in the physical properties of cuticular structures of the adult trachea. Enhanced tonic activation of the immune system is observed in the adult trachea, which encompasses the increased expression of antimicrobial peptides. In addition, modulatory processes are conspicuous, in this case mainly by the increased expression of G protein-coupled receptors in the adult trachea. Finally, all components of a peripheral circadian clock are present in the adult tracheal system, which is not the case in the larval tracheal system. Comparative analysis of driver lines targeting the adult tracheal system revealed that even the canonical tracheal driver line breathless (btl)-Gal4 is not able to target all parts of the adult tracheal system. Here, we have uncovered a specific transcriptome pattern of the adult tracheal system and provide this dataset as a basis for further analyses of the adult insect tracheal system.
Collapse
Affiliation(s)
- Judith Bossen
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
- German Lung Center (DZL), Airway Research Center North (ARCN), 24118 Kiel, Germany
| | - Ruben Prange
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Jan-Philip Kühle
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Sven Künzel
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany
| | - Xiao Niu
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Jörg U. Hammel
- Helmholtz-Zentrum Hereon, Institute of Materials Physics, 21502 Geesthacht, Germany
| | - Laura Krieger
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Mirjam Knop
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Birte Ehrhardt
- Research Center Borstel, Priority Research Area Chronic Lung Diseases, Early Life Origins of CLD, 23485 Borstel, Germany
| | - Karin Uliczka
- Research Center Borstel, Priority Research Area Chronic Lung Diseases, Early Life Origins of CLD, 23485 Borstel, Germany
| | - Susanne Krauss-Etschmann
- German Lung Center (DZL), Airway Research Center North (ARCN), 24118 Kiel, Germany
- Research Center Borstel, Priority Research Area Chronic Lung Diseases, Early Life Origins of CLD, 23485 Borstel, Germany
- Institute for Experimental Medicine, Kiel University, 24118 Kiel, Germany
| | - Thomas Roeder
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
- German Lung Center (DZL), Airway Research Center North (ARCN), 24118 Kiel, Germany
- Correspondence: ; Tel.: +49-431-880-81
| |
Collapse
|
12
|
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
|
13
|
Sakakibara Y, Yamashiro R, Chikamatsu S, Hirota Y, Tsubokawa Y, Nishijima R, Takei K, Sekiya M, Iijima KM. Drosophila Toll-9 is induced by aging and neurodegeneration to modulate stress signaling and its deficiency exacerbates tau-mediated neurodegeneration. iScience 2023; 26:105968. [PMID: 36718365 PMCID: PMC9883205 DOI: 10.1016/j.isci.2023.105968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Drosophila Toll-9 is most closely related to mammalian Toll-like receptors; however, physiological functions of Toll-9 remain elusive. We examined the roles of Toll-9 in fly brains in aging and neurodegeneration. Toll-9 mRNA levels were increased in aged fly heads accompanied by activation of nuclear factor-kappa B (NF-kB) and stress-activated protein kinase (SAPK) signaling, and many of these changes were modulated by Toll-9 in glial cells. The loss of Toll-9 did not affect lifespan or brain integrity, whereas it exacerbated hydrogen peroxide-induced lethality. Toll-9 expression was also induced by nerve injury but did not affect acute stress response or glial engulfment activity, suggesting Toll-9 may modulate subsequent neurodegeneration. In a fly tauopathy model, Toll-9 deficiency enhanced neurodegeneration and disease-related tau phosphorylation with reduced SAPK activity, and blocking SAPK enhanced tau phosphorylation and neurodegeneration. In sum, Toll-9 is induced upon aging and nerve injury and affects neurodegeneration by modulating stress kinase signaling.
Collapse
Affiliation(s)
- Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Risa Yamashiro
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Sachie Chikamatsu
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan,Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yu Hirota
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan,Reseach Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yoko Tsubokawa
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Risa Nishijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan,Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan,Corresponding author
| | - Koichi M. Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan,Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan,Corresponding author
| |
Collapse
|
14
|
Lu MY, Chtarbanova S. The role of micro RNAs (miRNAs) in the regulation of Drosophila melanogaster's innate immunity. Fly (Austin) 2022; 16:382-396. [PMID: 36412256 PMCID: PMC9683055 DOI: 10.1080/19336934.2022.2149204] [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] [Indexed: 11/23/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs ~19-22 nt long which post-transcriptionally regulate gene expression. Their ability to exhibit dynamic expression patterns coupled with their wide variety of targets allows miRNAs to regulate many processes, including the innate immune response of Drosophila melanogaster. Recent studies have identified miRNAs in Drosophila which are differentially expressed during infection with different pathogens as well as miRNAs that may affect immune signalling when differentially expressed. This review provides an overview of miRNAswhich have been identified to play a role in the immune response of Drosophila through targeting of the Toll and IMD signalling pathways and other immune processes. It will also explore the role of miRNAs in fine-tuning the immune response in Drosophila and highlight current gaps in knowledge regarding the role of miRNAs in immunity and areas for further research.
Collapse
Affiliation(s)
- Max Yang Lu
- Department of Biological Sciences, the University of Alabama, Tuscaloosa, AL, USA
| | - Stanislava Chtarbanova
- Department of Biological Sciences, the University of Alabama, Tuscaloosa, AL, USA,Center for Convergent Bioscience & Medicine, University of Alabama, Tuscaloosa, AL, USA,Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, USA,CONTACT Stanislava Chtarbanova Department of Biological Sciences, the University of Alabama, 300, Hackberry Ln, Tuscaloosa, AL-35487, USA
| |
Collapse
|
15
|
Xiong XP, Liang W, Liu W, Xu S, Li JL, Tito A, Situ J, Martinez D, Wu C, Perera RJ, Zhang S, Zhou R. The circular RNA Edis regulates neurodevelopment and innate immunity. PLoS Genet 2022; 18:e1010429. [PMID: 36301822 PMCID: PMC9612488 DOI: 10.1371/journal.pgen.1010429] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/13/2022] [Indexed: 11/07/2022] Open
Abstract
Circular RNAs (circRNAs) are widely expressed in eukaryotes. However, only a subset has been functionally characterized. We identify and validate a collection of circRNAs in Drosophila, and show that depletion of the brain-enriched circRNA Edis (circ_Ect4) causes hyperactivation of antibacterial innate immunity both in cultured cells and in vivo. Notably, Edis depleted flies display heightened resistance to bacterial infection and enhanced pathogen clearance. Conversely, ectopic Edis expression blocks innate immunity signaling. In addition, inactivation of Edis in vivo leads to impaired locomotor activity and shortened lifespan. Remarkably, these phenotypes can be recapitulated with neuron-specific depletion of Edis, accompanied by defective neurodevelopment. Furthermore, inactivation of Relish suppresses the innate immunity hyperactivation phenotype in the fly brain. Moreover, we provide evidence that Edis encodes a functional protein that associates with and compromises the processing and activation of the immune transcription factor Relish. Importantly, restoring Edis expression or ectopic expression of Edis-encoded protein suppresses both innate immunity and neurodevelopment phenotypes elicited by Edis depletion. Thus, our study establishes Edis as a key regulator of neurodevelopment and innate immunity.
Collapse
Affiliation(s)
- Xiao-Peng Xiong
- Tumor Initiation and Maintenance Program; NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Weihong Liang
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Cancer and Blood Disorders Institute. Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
| | - Wei Liu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Cancer and Blood Disorders Institute. Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
| | - Shiyu Xu
- The Brown Foundation Institute of Molecular Medicine, Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Jian-Liang Li
- Tumor Initiation and Maintenance Program; NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- National Institute of Environmental Health Sciences, Durham, North Carolina, United States of America
| | - Antonio Tito
- The Brown Foundation Institute of Molecular Medicine, Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Julia Situ
- Tumor Initiation and Maintenance Program; NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Daniel Martinez
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Chunlai Wu
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Ranjan J. Perera
- Tumor Initiation and Maintenance Program; NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Cancer and Blood Disorders Institute. Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
| | - Sheng Zhang
- The Brown Foundation Institute of Molecular Medicine, Department of Neurobiology and Anatomy, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Programs in Genetics & Epigenetics and Neuroscience, the University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Rui Zhou
- Tumor Initiation and Maintenance Program; NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Cancer and Blood Disorders Institute. Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
- Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, Saint Petersburg, Florida, United States of America
| |
Collapse
|
16
|
Ehrhardt B, El-Merhie N, Kovacevic D, Schramm J, Bossen J, Roeder T, Krauss-Etschmann S. Airway remodeling: The Drosophila model permits a purely epithelial perspective. FRONTIERS IN ALLERGY 2022; 3:876673. [PMID: 36187164 PMCID: PMC9520053 DOI: 10.3389/falgy.2022.876673] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Airway remodeling is an umbrella term for structural changes in the conducting airways that occur in chronic inflammatory lung diseases such as asthma or chronic obstructive pulmonary disease (COPD). The pathobiology of remodeling involves multiple mesenchymal and lymphoid cell types and finally leads to a variety of hardly reversible changes such as hyperplasia of goblet cells, thickening of the reticular basement membrane, deposition of collagen, peribronchial fibrosis, angiogenesis and hyperplasia of bronchial smooth muscle cells. In order to develop solutions for prevention or innovative therapies, these complex processes must be understood in detail which requires their deconstruction into individual building blocks. In the present manuscript we therefore focus on the role of the airway epithelium and introduce Drosophila melanogaster as a model. The simple architecture of the flies’ airways as well as the lack of adaptive immunity allows to focus exclusively on the importance of the epithelium for the remodeling processes. We will review and discuss genetic and environmentally induced changes in epithelial structures and molecular responses and propose an integrated framework of research for the future.
Collapse
Affiliation(s)
- Birte Ehrhardt
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Natalia El-Merhie
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Draginja Kovacevic
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Juliana Schramm
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Judith Bossen
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Correspondence: Susanne Krauss-Etschmann
| |
Collapse
|
17
|
Aalto A, Martínez‐Chacón G, Kietz C, Tsyganova N, Kreutzer J, Kallio P, Broemer M, Meinander A. M1-linked ubiquitination facilitates NF-κB activation and survival during sterile inflammation. FEBS J 2022; 289:5180-5197. [PMID: 35263507 PMCID: PMC9543601 DOI: 10.1111/febs.16425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 01/03/2023]
Abstract
Methionine 1 (M1)-linked ubiquitination plays a key role in the regulation of inflammatory nuclear factor-κB (NF-κB) signalling and is important for clearance of pathogen infection in Drosophila melanogaster. M1-linked ubiquitin (M1-Ub) chains are assembled by the linear ubiquitin E3 ligase (LUBEL) in flies. Here, we have studied the role of LUBEL in sterile inflammation induced by different types of cellular stresses. We have found that the LUBEL catalyses formation of M1-Ub chains in response to hypoxic, oxidative and mechanical stress conditions. LUBEL is shown to be important for flies to survive low oxygen conditions and paraquat-induced oxidative stress. This protective action seems to be driven by stress-induced activation of the NF-κB transcription factor Relish via the immune deficiency (Imd) pathway. In addition to LUBEL, the intracellular mediators of Relish activation, including the transforming growth factor activating kinase 1 (Tak1), Drosophila inhibitor of apoptosis (IAP) Diap2, the IκB kinase γ (IKKγ) Kenny and the initiator caspase Death-related ced-3/Nedd2-like protein (Dredd), but not the membrane receptor peptidoglycan recognition protein (PGRP)-LC, are shown to be required for sterile inflammatory response and survival. Finally, we showed that the stress-induced upregulation of M1-Ub chains in response to hypoxia, oxidative and mechanical stress is also induced in mammalian cells and protects from stress-induced cell death. Taken together, our results suggest that M1-Ub chains are important for NF-κB signalling in inflammation induced by stress conditions often observed in chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Anna Aalto
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
| | | | - Christa Kietz
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
| | - Nadezhda Tsyganova
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
| | - Joose Kreutzer
- Faculty of Medicine and Health TechnologyBioMediTechTampere UniversityFinland
| | - Pasi Kallio
- Faculty of Medicine and Health TechnologyBioMediTechTampere UniversityFinland
| | - Meike Broemer
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Annika Meinander
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
| |
Collapse
|
18
|
Tian S, Monteiro A. A transcriptomic atlas underlying developmental plasticity of seasonal forms of Bicyclus anynana butterflies. Mol Biol Evol 2022; 39:msac126. [PMID: 35679434 PMCID: PMC9218548 DOI: 10.1093/molbev/msac126] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022] Open
Abstract
Organisms residing in regions with alternating seasons often develop different phenotypes, or forms, in each season. These forms are often adaptations to each season and result from an altered developmental response to specific environmental cues such as temperature. While multiple studies have examined form-specific gene expression profiles in a diversity of species, little is known about how environments and developmental transitions, cued by hormone pulses, alter post-transcriptional patterns. In this study, we examine how gene expression, alternative splicing, and miRNA-mediated gene silencing in Bicyclus anynana butterfly hindwing tissue, varies across two rearing temperatures at four developmental timepoints. These timepoints flank two temperature-sensitive periods that coincide with two pulses of the insect hormone 20E. Our results suggest that developmental transitions, coincident with 20E pulses, elicit a greater impact on all these transcriptomic patterns than rearing temperatures per se. More similar transcriptomic patterns are observed pre-20E pulses than those observed post-20E pulses. We also found functionally distinct sets of differentially expressed and differentially spliced genes in the seasonal forms. Furthermore, around 10% of differentially expressed genes are predicted to be direct targets of, and regulated by, differentially expressed miRNAs between the seasonal forms. Many differentially expressed genes, miRNAs, or differentially spliced genes potentially regulate eyespot size plasticity, and we validated the differential splicing pattern of one such gene, daughterless. We present a comprehensive and interactive transcriptomic atlas of the hindwing tissue of both seasonal forms of B. anynana throughout development, a model organism of seasonal plasticity.
Collapse
Affiliation(s)
- Shen Tian
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Host–Pathogen Interactions between Metarhizium spp. and Locusts. J Fungi (Basel) 2022; 8:jof8060602. [PMID: 35736085 PMCID: PMC9224550 DOI: 10.3390/jof8060602] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
The progress in research on the interactions between Metarhizium spp. and locusts has improved our understanding of the interactions between fungal infection and host immunity. A general network of immune responses has been constructed, and the pathways regulating fungal pathogenicity have also been explored in depth. However, there have been no systematic surveys of interaction between Metarhizium spp. and locusts. The pathogenesis of Metarhizium comprises conidial attachment, germination, appressorial formation, and colonization in the body cavity of the host locusts. Meanwhile, the locust resists fungal infection through humoral and cellular immunity. Here, we summarize the crucial pathways that regulate the pathogenesis of Metarhizium and host immune defense. Conidial hydrophobicity is mainly affected by the contents of hydrophobins and chitin. Appressorial formation is regulated by the pathways of MAPKs, cAMP/PKA, and Ca2+/calmodulin. Lipid droplets degradation and secreted enzymes contributed to fungal penetration. The humoral response of locust is coordinated by the Toll pathway and the ecdysone. The regulatory mechanism of hemocyte differentiation and migration is elusive. In addition, behavioral fever and density-dependent population immunity have an impact on the resistance of hosts against fungal infection. This review depicts a prospect to help us understand host–pathogen interactions and provides a foundation for the engineering of entomopathogenic fungi and the discovery of insecticidal targets to control insect pests.
Collapse
|
20
|
Umetsu D. Cell mechanics and cell-cell recognition controls by Toll-like receptors in tissue morphogenesis and homeostasis. Fly (Austin) 2022; 16:233-247. [PMID: 35579305 PMCID: PMC9116419 DOI: 10.1080/19336934.2022.2074783] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Signal transduction by the Toll-like receptors (TLRs) is conserved and essential for innate immunity in metazoans. The founding member of the TLR family, Drosophila Toll-1, was initially identified for its role in dorsoventral axis formation in early embryogenesis. The Drosophila genome encodes nine TLRs that display dynamic expression patterns during development, suggesting their involvement in tissue morphogenesis and homeostasis. Recent progress on the developmental functions of TLRs beyond dorsoventral patterning has revealed not only their diverse functions in various biological processes, but also unprecedented molecular mechanisms in directly regulating cell mechanics and cell-cell recognition independent of the canonical signal transduction pathway involving transcriptional regulation of target genes. In this review, I feature and discuss the non-immune functions of TLRs in the control of epithelial tissue homeostasis, tissue morphogenesis, and cell-cell recognition between cell populations with different cell identities.
Collapse
Affiliation(s)
- Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
21
|
Shields A, Amcheslavsky A, Brown E, Lee TV, Nie Y, Tanji T, Ip YT, Bergmann A. Toll-9 interacts with Toll-1 to mediate a feedback loop during apoptosis-induced proliferation in Drosophila. Cell Rep 2022; 39:110817. [PMID: 35584678 PMCID: PMC9211775 DOI: 10.1016/j.celrep.2022.110817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 11/30/2022] Open
Abstract
Drosophila Toll-1 and all mammalian Toll-like receptors regulate innate immunity. However, the functions of the remaining eight Toll-related proteins in Drosophila are not fully understood. Here, we show that Drosophila Toll-9 is necessary and sufficient for a special form of compensatory proliferation after apoptotic cell loss (undead apoptosis-induced proliferation [AiP]). Mechanistically, for AiP, Toll-9 interacts with Toll-1 to activate the intracellular Toll-1 pathway for nuclear translocation of the NF-κB-like transcription factor Dorsal, which induces expression of the pro-apoptotic genes reaper and hid. This activity contributes to the feedback amplification loop that operates in undead cells. Given that Toll-9 also functions in loser cells during cell competition, we define a general role of Toll-9 in cellular stress situations leading to the expression of pro-apoptotic genes that trigger apoptosis and apoptosis-induced processes such as AiP. This work identifies conceptual similarities between cell competition and AiP.
Collapse
Affiliation(s)
- Alicia Shields
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Alla Amcheslavsky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Elizabeth Brown
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tom V Lee
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yingchao Nie
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Takahiro Tanji
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Y Tony Ip
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Andreas Bergmann
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
22
|
Uccellini MB, Bardina SV, Sánchez-Aparicio MT, White KM, Hou YJ, Lim JK, García-Sastre A. Passenger Mutations Confound Phenotypes of SARM1-Deficient Mice. Cell Rep 2021; 31:107498. [PMID: 32268088 DOI: 10.1016/j.celrep.2020.03.062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/25/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
The Toll/IL-1R-domain-containing adaptor protein SARM1 is expressed primarily in the brain, where it mediates axonal degeneration. Roles for SARM1 in TLR signaling, viral infection, inflammasome activation, and chemokine and Xaf1 expression have also been described. Much of the evidence for SARM1 function relies on SARM1-deficient mice generated in 129 ESCs and backcrossed to B6. The Sarm1 gene lies in a gene-rich region encompassing Xaf1 and chemokine loci, which remain 129 in sequence. We therefore generated additional knockout strains on the B6 background, confirming the role of SARM1 in axonal degeneration and WNV infection, but not in VSV or LACV infection, or in chemokine or Xaf1 expression. Sequence variation in proapoptotic Xaf1 between B6 and 129 results in coding changes and distinct splice variants, which may account for phenotypes previously attributed to SARM1. Reevaluation of phenotypes in these strains will be critical for understanding the function of SARM1.
Collapse
Affiliation(s)
- Melissa B Uccellini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Susana V Bardina
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maria Teresa Sánchez-Aparicio
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ying-Ju Hou
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
23
|
Rahman SR, Terranova T, Tian L, Hines HM. Developmental Transcriptomics Reveals a Gene Network Driving Mimetic Color Variation in a Bumble Bee. Genome Biol Evol 2021; 13:6244266. [PMID: 33881508 PMCID: PMC8220310 DOI: 10.1093/gbe/evab080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2021] [Indexed: 11/24/2022] Open
Abstract
A major goal of evolutionary genetics and evo-devo is to understand how changes in genotype manifest as changes in phenotype. Bumble bees display remarkable color pattern diversity while converging onto numerous regional Müllerian mimicry patterns, thus enabling exploration of the genetic mechanisms underlying convergent phenotypic evolution. In western North America, multiple bumble bee species converge onto local mimicry patterns through parallel shifts of midabdominal segments from red to black. It was previously demonstrated that a Hox gene, Abd-B, is the key regulator of the phenotypic switch in one of these species, Bombus melanopygus, however, the mechanism by which Abd-B regulates color differentiation remains unclear. Using tissue/stage-specific transcriptomic analysis followed by qRT–PCR validation, this study reveals a suite of genes potentially involved downstream of Abd-B during color pattern differentiation. The data support differential genes expression of not only the first switch gene Abd-B, but also an intermediate developmental gene nubbin, and a whole suite of downstream melanin and redox genes that together reinforce the observed eumelanin (black)-pheomelanin (red) ratios. These include potential genes involved in the production of insect pheomelanins, a pigment until recently not thought to occur in insects and thus lacking known regulatory enzymes. The results enhance understanding of pigmentation gene networks involved in bumble bee color pattern development and diversification, while providing insights into how upstream regulators such as Hox genes interact with downstream morphogenic players to facilitate this adaptive phenotypic radiation.
Collapse
Affiliation(s)
- Sarthok Rasique Rahman
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA.,Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
| | - Tatiana Terranova
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Li Tian
- Department of Entomology, China Agricultural University, Beijing, China
| | - Heather M Hines
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA.,Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
24
|
Wagner C, Uliczka K, Bossen J, Niu X, Fink C, Thiedmann M, Knop M, Vock C, Abdelsadik A, Zissler UM, Isermann K, Garn H, Pieper M, Wegmann M, Koczulla AR, Vogelmeier CF, Schmidt-Weber CB, Fehrenbach H, König P, Silverman N, Renz H, Pfefferle P, Heine H, Roeder T. Constitutive immune activity promotes JNK- and FoxO-dependent remodeling of Drosophila airways. Cell Rep 2021; 35:108956. [PMID: 33826881 DOI: 10.1016/j.celrep.2021.108956] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/29/2020] [Accepted: 03/17/2021] [Indexed: 01/07/2023] Open
Abstract
Extensive remodeling of the airways is a major characteristic of chronic inflammatory lung diseases such as asthma or chronic obstructive pulmonary disease (COPD). To elucidate the importance of a deregulated immune response in the airways for remodeling processes, we established a matching Drosophila model. Here, triggering the Imd (immune deficiency) pathway in tracheal cells induced organ-wide remodeling. This structural remodeling comprises disorganization of epithelial structures and comprehensive epithelial thickening. We show that these structural changes do not depend on the Imd pathway's canonical branch terminating on nuclear factor κB (NF-κB) activation. Instead, activation of a different segment of the Imd pathway that branches off downstream of Tak1 and comprises activation of c-Jun N-terminal kinase (JNK) and forkhead transcription factor of the O subgroup (FoxO) signaling is necessary and sufficient to mediate the observed structural changes of the airways. Our findings imply that targeting JNK and FoxO signaling in the airways could be a promising strategy to interfere with disease-associated airway remodeling processes.
Collapse
Affiliation(s)
- Christina Wagner
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; Division of Invertebrate Models, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany
| | - Karin Uliczka
- Division of Invertebrate Models, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Division of Innate Immunity, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany
| | - Judith Bossen
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Xiao Niu
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Christine Fink
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Marcus Thiedmann
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Mirjam Knop
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Christina Vock
- Division of Experimental Pneumology, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany
| | - Ahmed Abdelsadik
- Zoology, Aswan University, Aswan 81528, Egypt; Molecular Biotechnology Program, Faculty of Advanced Basic Sciences, Galala University, 43552 New Galala, Egypt
| | - Ulrich M Zissler
- Center of Allergy and Environment (ZAUM), Technical University Munich and Helmholtz Center Munich, German Research Center for Environmental Health, 80802 Munich, Germany; CPC-M, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kerstin Isermann
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Mario Pieper
- University Lübeck, Anatomical Institute, 23538 Lübeck, Germany
| | - Michael Wegmann
- Division of Asthma Exacerbation & Regulation, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Andreas R Koczulla
- Pulmonary and Critical Care Medicine, Department of Medicine, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Claus F Vogelmeier
- Pulmonary and Critical Care Medicine, Department of Medicine, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University Munich and Helmholtz Center Munich, German Research Center for Environmental Health, 80802 Munich, Germany; CPC-M, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Heinz Fehrenbach
- Division of Experimental Pneumology, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Peter König
- University Lübeck, Anatomical Institute, 23538 Lübeck, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Neil Silverman
- University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Harald Renz
- Molecular Diagnostics, Institute of Laboratory Medicine and Pathobiochemistry, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Petra Pfefferle
- Comprehensive Biobank Marburg, University Medical Center Giessen and Marburg, Medical Faculty, Philipps University Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Holger Heine
- Division of Innate Immunity, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Thomas Roeder
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany.
| |
Collapse
|
25
|
Li Y, Yuan W, Xu Q, Liu H, Dai X. The regulation of immune responses against white spot syndrome virus or Vibrio alginolyticus in toll-like receptors silenced giant freshwater prawn (Macrobrachium rosenbergii). FISH & SHELLFISH IMMUNOLOGY 2020; 107:84-94. [PMID: 33035653 DOI: 10.1016/j.fsi.2020.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Toll-like receptors, which are a class of cell-surface proteins, have been regarded as the most important pattern recognition receptors in the innate immunity and play a vital role in multiple innate immune responses against pathogen invasion. The full-length cDNA of a novel Toll-like receptor (MrToll3) was identified from Macrobrachium rosenbergii in the current research. The nucleotide sequence of MrToll3 is 4481 bp long and contains a 3726-bp open reading frame encoding a putative protein of 1241 amino acids. MrToll3 was constitutively expressed in all the examined tissues, and high expression of MrToll3 was detected in gill, heart, and ganglion. The result of RNA interference assay revealed that silencing of MrToll1 remarkably suppressed the prophenoloxidase (proPO) expression and phenoloxidase (PO) activities while enhancing MrToll2 expression in the prawns. Furthermore, the expression of myeloid differentiation factor 88 (MyD88), anti-lipopolysaccharide factor (ALF) and crustin was remarkably down-regulated in the MrToll1-silenced prawns after white spot syndrome virus (WSSV) or Vibrio alginolyticus challenge. MrToll2-silenced prawns exhibited the significant decline of ALF and crustin expression post the pathogen challenges, and silencing of MrToll3 obviously improved the immune deficiency (IMD) expression during the whole RNA interference assay. Additionally, higher mortality was observed in MrToll1-or MrToll2-silenced prawns after V. alginolyticus challenge, and the MrToll1-silenced prawns also showed the obviously enhanced susceptibility to WSSV. These results suggested that MrToll1, 2, and 3 were involved in the innate immune responses against WSSV and V. alginolyticus in M. rosenbergii.
Collapse
Affiliation(s)
- Yun Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, 201306, China
| | - Wei Yuan
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, 201306, China
| | - Qi Xu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, 201306, China
| | - Hong Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, 201306, China
| | - Xilin Dai
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
26
|
Do AN, Chun Y, Grishina G, Grishin A, Rogers AJ, Raby BA, Weiss ST, Vicencio A, Schadt EE, Bunyavanich S. Network study of nasal transcriptome profiles reveals master regulator genes of asthma. J Allergy Clin Immunol 2020; 147:879-893. [PMID: 32828590 DOI: 10.1016/j.jaci.2020.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Nasal transcriptomics can provide an accessible window into asthma pathobiology. OBJECTIVE Our goal was to move beyond gene signatures of asthma to identify master regulator genes that causally regulate genes associated with asthma phenotypes. METHODS We recruited 156 children with severe persistent asthma and controls for nasal transcriptome profiling and applied network-based and probabilistic causal methods to identify severe asthma genes and their master regulators. We then took the same approach in an independent cohort of 190 adults with mild/moderate asthma and controls to identify mild/moderate asthma genes and their master regulators. Comparative analysis of the master regulator genes followed by validation testing in independent children with severe asthma (n = 21) and mild/moderate asthma (n = 154) was then performed. RESULTS Nasal gene signatures for severe persistent asthma and for mild/moderate persistent asthma were identified; both were found to be enriched in coexpression network modules for ciliary function and inflammatory response. By applying probabilistic causal methods to these gene signatures and validation testing in independent cohorts, we identified (1) a master regulator gene common to asthma across severity and ages (FOXJ1); (2) master regulator genes of severe persistent asthma in children (LRRC23, TMEM231, CAPS, PTPRC, and FYB); and (3) master regulator genes of mild/moderate persistent asthma in children and adults (C1orf38 and FMNL1). The identified master regulators were statistically inferred to causally regulate the expression of downstream genes that modulate ciliary function and inflammatory response to influence asthma. CONCLUSION The identified master regulator genes of asthma provide a novel path forward to further uncovering asthma mechanisms and therapy.
Collapse
Affiliation(s)
- Anh N Do
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yoojin Chun
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Galina Grishina
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexander Grishin
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Angela J Rogers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Benjamin A Raby
- Division of Pulmonary Medicine, Children's Hospital Boston, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass
| | - Alfin Vicencio
- Division of Pulmonary Medicine, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric E Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
27
|
Chowdhury A, Modahl CM, Tan ST, Wong Wei Xiang B, Missé D, Vial T, Kini RM, Pompon JF. JNK pathway restricts DENV2, ZIKV and CHIKV infection by activating complement and apoptosis in mosquito salivary glands. PLoS Pathog 2020; 16:e1008754. [PMID: 32776975 PMCID: PMC7444518 DOI: 10.1371/journal.ppat.1008754] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/20/2020] [Accepted: 06/26/2020] [Indexed: 11/18/2022] Open
Abstract
Arbovirus infection of Aedes aegypti salivary glands (SGs) determines transmission. However, there is a dearth of knowledge on SG immunity. Here, we characterized SG immune response to dengue, Zika and chikungunya viruses using high-throughput transcriptomics. We also describe a transcriptomic response associated to apoptosis, blood-feeding and lipid metabolism. The three viruses differentially regulate components of Toll, Immune deficiency (IMD) and c-Jun N- terminal Kinase (JNK) pathways. However, silencing of the Toll and IMD pathway components showed variable effects on SG infection by each virus. In contrast, regulation of the JNK pathway produced consistent responses in both SGs and midgut. Infection by the three viruses increased with depletion of the activator Kayak and decreased with depletion of the negative regulator Puckered. Virus-induced JNK pathway regulates the complement factor, Thioester containing protein-20 (TEP20), and the apoptosis activator, Dronc, in SGs. Individual and co-silencing of these genes demonstrate their antiviral effects and that both may function together. Co-silencing either TEP20 or Dronc with Puckered annihilates JNK pathway antiviral effect. Upon infection in SGs, TEP20 induces antimicrobial peptides (AMPs), while Dronc is required for apoptosis independently of TEP20. In conclusion, we revealed the broad antiviral function of JNK pathway in SGs and showed that it is mediated by a TEP20 complement and Dronc-induced apoptosis response. These results expand our understanding of the immune arsenal that blocks arbovirus transmission. Arboviral diseases caused by dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses are responsible for large number of death and debilitation around the world. These viruses are transmitted to humans by the mosquito vector, Aedes aegypti. During the bites, infected salivary glands (SGs) release saliva containing viruses, which initiate human infection. As the tissue where transmitted viruses are produced, SG infection is a key determinant of transmission. To bridge the knowledge gap in vector-virus molecular interactions in SGs, we describe the transcriptome after DENV, ZIKV and CHIKV infection using RNA-sequencing and characterized the immune response in this tissue. Our study reveals the broad antiviral function of c-Jun N-terminal kinase (JNK) pathway against DENV, ZIKV and CHIKV in SGs. We further show that it is mediated by the complement system and apoptosis, identifying the mechanism. Our study adds the JNK pathway to the immune arsenal that can be harnessed to engineer refractory vectors.
Collapse
Affiliation(s)
- Avisha Chowdhury
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Cassandra M. Modahl
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Siok Thing Tan
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Dorothée Missé
- MIVEGEC, IRD, CNRS, Univ. Montpellier, Montpellier, France
| | - Thomas Vial
- Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - R. Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, Singapore
- * E-mail: (RMK); (JFP)
| | - Julien Francis Pompon
- Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- MIVEGEC, IRD, CNRS, Univ. Montpellier, Montpellier, France
- * E-mail: (RMK); (JFP)
| |
Collapse
|
28
|
Lang L, Bao M, Jing W, Chen W, Wang L. Clone, identification and functional characterization of a novel toll (Shtoll1) from the freshwater crab Sinopotamon henanense in response to cadmium exposure and Aeromonas hydrophila infection. FISH & SHELLFISH IMMUNOLOGY 2020; 98:401-413. [PMID: 31953198 DOI: 10.1016/j.fsi.2020.01.012] [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: 09/10/2019] [Revised: 12/31/2019] [Accepted: 01/05/2020] [Indexed: 06/10/2023]
Abstract
Toll is essential in innate immune system which is important for defense against bacterial, fungal and viral infections in invertebrates. Our previous study showed that cadmium (Cd) could change the expression pattern of ShToll3 in the epithelium (gills and midgut from the freshwater crab Sinopotamon henanense) infected by Aeromonas hydrophila. To investigate the diverse innate immune roles of crustacean homolog Tolls, in this study, we cloned Shtoll1 from S. henanense. The full-length cDNA of Shtoll1 was 4746 bp, with an ORF of 3033 bp encoding a putative protein of 111 amino acids, a 5'-untranslated region of 255 bp and a 3'-untranslated region of 1713 bp. Phylogenetic analysis showed that ShToll1 was clustered into the group of DmToll1, DmToll 4 and DmToll 5. In addition, the tissue distribution results showed that Shtoll1 was expressed widely in different tissues, with the highest expression in heamocytes. Besides, Shtoll1 expressions were upregulated in heamocytes and hepatopancreas after A. hydrophila infection. At the same time, the increase of Shtoll1 expressions were examined in heamocytes in response to Cd exposure and A. hydrophila infection in combination. Through western blotting and immunohistochemical analysis, the ShToll1 expressions in heamocytes were increased in response to A. hydrophila and Cd independently as well as in combination. Moreover, the mRNA level of three antimicrobial peptides (AMPs) alf5, alf6, and c-lys, which possibly responded to Cd and A. hydrophila stimulation through Shtoll1, were analyzed. Thus, we conclude that Cd expand the susceptibility of ShToll1 to A. hydrophila infection in heamocytes. This suggest that ShToll1 may contribute to the innate immune defense of S. henanense against A. hydrophila and Cd in heamocytes.
Collapse
Affiliation(s)
- Lang Lang
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, China
| | - Minnan Bao
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, China
| | - Weixin Jing
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, China
| | - Wei Chen
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Lan Wang
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, China.
| |
Collapse
|
29
|
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
|
30
|
Sanchez Bosch P, Makhijani K, Herboso L, Gold KS, Baginsky R, Woodcock KJ, Alexander B, Kukar K, Corcoran S, Jacobs T, Ouyang D, Wong C, Ramond EJV, Rhiner C, Moreno E, Lemaitre B, Geissmann F, Brückner K. Adult Drosophila Lack Hematopoiesis but Rely on a Blood Cell Reservoir at the Respiratory Epithelia to Relay Infection Signals to Surrounding Tissues. Dev Cell 2019; 51:787-803.e5. [PMID: 31735669 PMCID: PMC7263735 DOI: 10.1016/j.devcel.2019.10.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 08/08/2019] [Accepted: 10/17/2019] [Indexed: 12/23/2022]
Abstract
The use of adult Drosophila melanogaster as a model for hematopoiesis or organismal immunity has been debated. Addressing this question, we identify an extensive reservoir of blood cells (hemocytes) at the respiratory epithelia (tracheal air sacs) of the thorax and head. Lineage tracing and functional analyses demonstrate that the majority of adult hemocytes are phagocytic macrophages (plasmatocytes) from the embryonic lineage that parallels vertebrate tissue macrophages. Surprisingly, we find no sign of adult hemocyte expansion. Instead, hemocytes play a role in relaying an innate immune response to the blood cell reservoir: through Imd signaling and the Jak/Stat pathway ligand Upd3, hemocytes act as sentinels of bacterial infection, inducing expression of the antimicrobial peptide Drosocin in respiratory epithelia and colocalizing fat body domains. Drosocin expression in turn promotes animal survival after infection. Our work identifies a multi-signal relay of organismal humoral immunity, establishing adult Drosophila as model for inter-organ immunity.
Collapse
Affiliation(s)
- Pablo Sanchez Bosch
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Kalpana Makhijani
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Leire Herboso
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Katrina S Gold
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Rowan Baginsky
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Brandy Alexander
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Katelyn Kukar
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Sean Corcoran
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Thea Jacobs
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Debra Ouyang
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Corinna Wong
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | | | - Frederic Geissmann
- King's College London, London, UK; Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katja Brückner
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
31
|
Loring HS, Thompson PR. Emergence of SARM1 as a Potential Therapeutic Target for Wallerian-type Diseases. Cell Chem Biol 2019; 27:1-13. [PMID: 31761689 DOI: 10.1016/j.chembiol.2019.11.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
Wallerian degeneration is a neuronal death pathway that is triggered in response to injury or disease. Death was thought to occur passively until the discovery of a mouse strain, i.e., Wallerian degeneration slow (WLDS), which was resistant to degeneration. Given that the WLDS mouse encodes a gain-of-function fusion protein, its relevance to human disease was limited. The later discovery that SARM1 (sterile alpha and toll/interleukin receptor [TIR] motif-containing protein 1) promotes Wallerian degeneration suggested the existence of a pathway that might be targeted therapeutically. More recently, SARM1 was found to execute degeneration by hydrolyzing NAD+. Notably, SARM1 knockdown or knockout prevents neuron degeneration in response to a range of insults that lead to peripheral neuropathy, traumatic brain injury, and neurodegenerative disease. Here, we discuss the role of SARM1 in Wallerian degeneration and the opportunities to target this enzyme therapeutically.
Collapse
Affiliation(s)
- Heather S Loring
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, LRB 826, 364 Plantation Street, Worcester, MA 01605, USA; Program in Chemical Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, LRB 826, 364 Plantation Street, Worcester, MA 01605, USA; Program in Chemical Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
32
|
Chowdhury M, Li CF, He Z, Lu Y, Liu XS, Wang YF, Ip YT, Strand MR, Yu XQ. Toll family members bind multiple Spätzle proteins and activate antimicrobial peptide gene expression in Drosophila. J Biol Chem 2019; 294:10172-10181. [PMID: 31088910 DOI: 10.1074/jbc.ra118.006804] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/10/2019] [Indexed: 12/31/2022] Open
Abstract
The Toll signaling pathway in Drosophila melanogaster regulates several immune-related functions, including the expression of antimicrobial peptide (AMP) genes. The canonical Toll receptor (Toll-1) is activated by the cytokine Spätzle (Spz-1), but Drosophila encodes eight other Toll genes and five other Spz genes whose interactions with one another and associated functions are less well-understood. Here, we conducted in vitro assays in the Drosophila S2 cell line with the Toll/interleukin-1 receptor (TIR) homology domains of each Toll family member to determine whether they can activate a known target of Toll-1, the promoter of the antifungal peptide gene drosomycin. All TIR family members activated the drosomycin promoter, with Toll-1 and Toll-7 TIRs producing the highest activation. We found that the Toll-1 and Toll-7 ectodomains bind Spz-1, -2, and -5, and also vesicular stomatitis virus (VSV) virions, and that Spz-1, -2, -5, and VSV all activated the promoters of drosomycin and several other AMP genes in S2 cells expressing full-length Toll-1 or Toll-7. In vivo experiments indicated that Toll-1 and Toll-7 mutants could be systemically infected with two bacterial species (Enterococcus faecalis and Pseudomonas aeruginosa), the opportunistic fungal pathogen Candida albicans, and VSV with different survival times in adult females and males compared with WT fly survival. Our results suggest that all Toll family members can activate several AMP genes. Our results further indicate that Toll-1 and Toll-7 bind multiple Spz proteins and also VSV, but they differentially affect adult survival after systemic infection, potentially because of sex-specific differences in Toll-1 and Toll-7 expression.
Collapse
Affiliation(s)
- Munmun Chowdhury
- From the Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64110
| | - Chun-Feng Li
- From the Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64110.,the State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Zhen He
- From the Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64110.,the School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Yuzhen Lu
- the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xu-Sheng Liu
- the School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Yu-Feng Wang
- the School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Y Tony Ip
- the Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Michael R Strand
- the Department of Entomology, University of Georgia, Athens, Georgia 30602, and
| | - Xiao-Qiang Yu
- From the Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64110, .,the School of Life Sciences, Central China Normal University, Wuhan 430079, China.,the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
33
|
Park S, Jo YH, Park KB, Ko HJ, Kim CE, Bae YM, Kim B, Jun SA, Bang IS, Lee YS, Kim YJ, Han YS. TmToll-7 Plays a Crucial Role in Innate Immune Responses Against Gram-Negative Bacteria by Regulating 5 AMP Genes in Tenebrio molitor. Front Immunol 2019; 10:310. [PMID: 30930888 PMCID: PMC6424196 DOI: 10.3389/fimmu.2019.00310] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/06/2019] [Indexed: 11/22/2022] Open
Abstract
Although it is known that the Drosophila Toll-7 receptor plays a critical role in antiviral autophagy, its function in other insects has not yet been reported. Here, we have identified a Toll-like receptor 7 gene, TmToll-7, in the coleopteran insect T. molitor and examined its potential role in antibacterial and antifungal immunity. We showed that TmToll-7 expression was significantly induced in larvae 6 h after infection with Escherichia coli and Staphylococcus aureus and 9 h after infection with Candida albicans. However, even though TmToll-7 was induced by all three pathogens, we found that TmToll-7 knockdown significantly reduced larval survival to E. coli, but not to S. aureus, and C. albicans infections. To understand the reasons for this difference, we examined the effects of TmToll-7 knockdown on antimicrobial peptide (AMP) gene expression and found a significant reduction of E. coli-induced expression of AMP genes such as TmTenecin-1, TmDefensin-1, TmDefensin-2, TmColeoptericin-1, and TmAttacin-2. Furthermore, TmToll-7 knockdown larvae infected with E. coli showed significantly higher bacterial growth in the hemolymph compared to control larvae treated with Vermilion dsRNA. Taken together, our results suggest that TmToll-7 plays an important role in regulating the immune response of T. molitor to E. coli.
Collapse
Affiliation(s)
- Soyi Park
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Yong Hun Jo
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Ki Beom Park
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Hye Jin Ko
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Chang Eun Kim
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Young Min Bae
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Bobae Kim
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| | - Sung Ah Jun
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - In Seok Bang
- Department of Biological Science, Hoseo University, Asan, South Korea
| | - Yong Seok Lee
- Department of Life Science and Biotechnology, College of Natural Sciences, Soonchunhyang University, Asan, South Korea
| | - Yu Jung Kim
- Department of Chemistry and Biochemistry, College of Natural Sciences, California State University, San Bernardino, CA, United States
| | - Yeon Soo Han
- Division of Plant Biotechnology, College of Agriculture and Life Sciences, Institute of Environmentally-Friendly Agriculture Chonnam National University, Gwangju, South Korea
| |
Collapse
|
34
|
Carty M, Bowie AG. SARM: From immune regulator to cell executioner. Biochem Pharmacol 2019; 161:52-62. [PMID: 30633870 DOI: 10.1016/j.bcp.2019.01.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
SARM is the fifth and most conserved member of the Toll/Il-1 Receptor (TIR) adaptor family. However, unlike the other TIR adaptors, MyD88, Mal, TRIF and TRAM, SARM does not participate in transducing signals downstream of TLRs. By contrast SARM inhibits TLR signalling by interacting with the adaptors TRIF and MyD88. In addition, SARM also has positive roles in innate immunity by activating specific transcriptional programs following immune challenge. SARM has a pivotal role in activating different forms of cell death following cellular stress and viral infection. Many of these functions of mammalian SARM are also reflected in SARM orthologues in lower organisms such as C. elegans and Drosophila. SARM expression is particularly enriched in neurons of the CNS and SARM has a critical role in neuronal death and in axon degeneration. Recent fascinating molecular insights have been revealed as to the molecular mechanism of SARM mediated axon degeneration. SARM has been shown to deplete NAD+ by possessing intrinsic NADase activity in the TIR domain of the protein. This activity can be activated experimentally by forced dimerization of the TIR domain. It is thought that this activity of SARM is normally switched off by the axo-protective activities of NMNAT2 which maintain low levels of the NAD+ precursor NMN. Therefore, there is now great excitement in the field of SARM research as targeting this enzymatic activity of SARM may lead to the development of new therapies for neurodegenerative diseases such as multiple sclerosis and motor neuron disease.
Collapse
Affiliation(s)
- Michael Carty
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
35
|
Wang F, Xia Q. Back to homeostasis: Negative regulation of NF-κB immune signaling in insects. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 87:216-223. [PMID: 29908201 DOI: 10.1016/j.dci.2018.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 06/08/2023]
Abstract
Maintenance of homeostasis requires prompt activation and down-regulation of immune signaling pathways. This review attempts to summarize our current knowledge regarding the negative regulation of two NF-κB signaling pathways in insects, Toll and IMD pathway, which are mostly essential for host defense against bacteria and fungus. Various types of negative regulators and their mechanisms are discussed here with the emphasis on the prominent roles of ubiquitination. The counterbalance between these two pathways, the crosstalk with other physiological pathways, and the difference in their repertoires of negative regulators are also discussed.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China.
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400716, China
| |
Collapse
|
36
|
Shmueli A, Shalit T, Okun E, Shohat-Ophir G. The Toll Pathway in the Central Nervous System of Flies and Mammals. Neuromolecular Med 2018; 20:419-436. [PMID: 30276585 DOI: 10.1007/s12017-018-8515-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Toll receptors, first identified to regulate embryogenesis and immune responses in the adult fly and subsequently defined as the principal sensors of infection in mammals, are increasingly appreciated for their impact on the homeostasis of the central as well as the peripheral nervous systems. Whereas in the context of immunity, the fly Toll and the mammalian TLR pathways have been researched in parallel, the expression pattern and functionality have largely been researched disparately. Herein, we provide data on the expression pattern of the Toll homologues, signaling components, and downstream effectors in ten different cell populations of the adult fly central nervous system (CNS). We have compared the expression of the different Toll pathways in the fly to the expression of TLRs in the mouse brain and discussed the implications with respect to commonalities, differences, and future perspectives.
Collapse
Affiliation(s)
- Anat Shmueli
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Tali Shalit
- The Mantoux Bioinformatics institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Eitan Okun
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.
- The Paul Feder Laboratory on Alzheimer's Disease Research, Ramat-Gan, Israel.
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Building 901, room 315, Ramat-Gan, 5290000, Israel.
| | - Galit Shohat-Ophir
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
37
|
Abstract
Like humans, insects face the threat of viral infection. Despite having repercussions on human health and disease, knowledge gaps exist for how insects cope with viral pathogens. Drosophila melanogaster serves as an ideal insect model due to its genetic tractability. When encountering a pathogen, two major approaches to fight disease are resistance strategies and tolerance strategies. Disease resistance strategies promote the health of the infected host by reducing pathogen load. Multiple disease resistance mechanisms have been identified in Drosophila: RNA interference, Jak/STAT signaling, Toll signaling, IMD signaling, and autophagy. Disease tolerance mechanisms, in contrast, do not reduce pathogen load directly, but rather mitigate the stress and damage incurred by infection. The main benefit of tolerance mechanisms may therefore be to provide the host with time to engage antiviral resistance mechanisms that eliminate the threat. In this review, antiviral resistance mechanisms used by Drosophila will be described and compared to mammalian antiviral mechanisms. Disease tolerance will then be explained in a broader context as this is a burgeoning field of study.
Collapse
Affiliation(s)
- Jonathan Chow
- Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jonathan C Kagan
- Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States.
| |
Collapse
|
38
|
Nazario-Toole AE, Robalino J, Okrah K, Corrada-Bravo H, Mount SM, Wu LP. The Splicing Factor RNA-Binding Fox Protein 1 Mediates the Cellular Immune Response in Drosophila melanogaster. THE JOURNAL OF IMMUNOLOGY 2018; 201:1154-1164. [PMID: 29997126 DOI: 10.4049/jimmunol.1800496] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022]
Abstract
The uptake and destruction of bacteria by phagocytic cells is an essential defense mechanism in metazoans. To identify novel genes involved in the phagocytosis of Staphylococcus aureus, a major human pathogen, we assessed the phagocytic capacity of adult blood cells (hemocytes) of the fruit fly, Drosophila melanogaster, by testing several lines of the Drosophila Genetic Reference Panel. Natural genetic variation in the gene RNA-binding Fox protein 1 (Rbfox1) correlated with low phagocytic capacity in hemocytes, pointing to Rbfox1 as a candidate regulator of phagocytosis. Loss of Rbfox1 resulted in increased expression of the Ig superfamily member Down syndrome adhesion molecule 4 (Dscam4). Silencing of Dscam4 in Rbfox1-depleted blood cells rescued the fly's cellular immune response to S. aureus, indicating that downregulation of Dscam4 by Rbfox1 is critical for S. aureus phagocytosis in Drosophila To our knowledge, this study is the first to demonstrate a link between Rbfox1, Dscam4, and host defense against S. aureus.
Collapse
Affiliation(s)
- Ashley E Nazario-Toole
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742.,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742; and
| | - Javier Robalino
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742
| | - Kwame Okrah
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Hector Corrada-Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Stephen M Mount
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742.,Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20742
| | - Louisa P Wu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742; .,Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742; and
| |
Collapse
|
39
|
Lang L, Zhang Z, Jing W, Hwang JS, Lee SC, Wang L. Identification of a novel toll gene (Shtoll3) from the freshwater crab Sinopotamon henanense and its expression pattern changes in response to cadmium followed by Aeromonas hydrophila infection. FISH & SHELLFISH IMMUNOLOGY 2017; 71:177-190. [PMID: 29017939 DOI: 10.1016/j.fsi.2017.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 10/03/2017] [Accepted: 10/06/2017] [Indexed: 06/07/2023]
Abstract
Toll signaling is essential for expression of immune genes which are important for defense against bacterial, fungal and viral infections in invertebrates. Although several toll genes have been identified in the crustaceans, none of them has been investigated in freshwater crab Sinopotamon henanense. Moreover, the effect of cadmium (Cd) on toll gene expression has never been examined on the freshwater crabs which live in the sediments and are prone to heavy metal bioaccumulation. Our transcriptomic analysis of hepatopancreas tissue reveals that toll3 gene expression has been decreased when treated with Cd. In this study, we cloned one toll gene (hereby designated Shtoll3) from the crab. The full-length cDNA of Shtoll3 was 4488 bp, with an ORF of 3693 bp encoding a putative protein of 1230 amino acids, a 5'-untranslated region of 414 bp and a 3'-untranslated region of 781 bp. Phylogenetic analysis showed that ShToll3 was clustered into the group of DmToll8. The tissue distribution results showed that Shtoll3 was expressed widely in different tissues, with the highest in gills, and the lowest in hemocytes. Shtoll3 expression was down-regulated only in midguts after Aeromonas hydrophila infection. With Cd presence, Shtoll3 expression in response to A. hydrophila were up-regulated in midguts and gills, which was further confirmed by western blotting analysis. Moreover, the mRNA level of two antimicrobial peptides (AMPs) crustin and c-lys, which possibly responded to Cd and A. hydrophila stimulation through Shtoll3, were analysised. Thus, we conclude that Cd changes the susceptibility of Shtoll3 to A. hydrophila infection in gills and midguts. This suggest that Shtoll3 may contribute to the innate immune defense of S. henanense to A. hydrophila and Cd can modify the immune function in epithelium.
Collapse
Affiliation(s)
- Lang Lang
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China
| | - Zuobing Zhang
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China
| | - Weixin Jing
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China
| | - Jiang-Shiou Hwang
- Institute of Marine Biology, National Taiwan Ocean University, Keelung 20224, Taiwan, ROC
| | - Shao-Chin Lee
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China
| | - Lan Wang
- School of Life Science, Shanxi University, 92 Wucheng Road, Taiyuan 030006, China.
| |
Collapse
|
40
|
Lonsdale Z, Lee K, Kiriakidu M, Amarasinghe H, Nathanael D, O’Connor CJ, Mallon EB. Allele specific expression and methylation in the bumblebee, Bombus terrestris. PeerJ 2017; 5:e3798. [PMID: 28929021 PMCID: PMC5600721 DOI: 10.7717/peerj.3798] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/21/2017] [Indexed: 12/29/2022] Open
Abstract
The social hymenoptera are emerging as models for epigenetics. DNA methylation, the addition of a methyl group, is a common epigenetic marker. In mammals and flowering plants methylation affects allele specific expression. There is contradictory evidence for the role of methylation on allele specific expression in social insects. The aim of this paper is to investigate allele specific expression and monoallelic methylation in the bumblebee, Bombus terrestris. We found nineteen genes that were both monoallelically methylated and monoallelically expressed in a single bee. Fourteen of these genes express the hypermethylated allele, while the other five express the hypomethylated allele. We also searched for allele specific expression in twenty-nine published RNA-seq libraries. We found 555 loci with allele-specific expression. We discuss our results with reference to the functional role of methylation in gene expression in insects and in the as yet unquantified role of genetic cis effects in insect allele specific methylation and expression.
Collapse
Affiliation(s)
- Zoë Lonsdale
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Kate Lee
- Bioinformatics and Biostatistics Support Hub (B/BASH), University of Leicester, Leicester, United Kingdom
| | - Maria Kiriakidu
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Harindra Amarasinghe
- Academic Unit of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Despina Nathanael
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | | | - Eamonn B. Mallon
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
41
|
Two White Spot Syndrome Virus MicroRNAs Target the Dorsal Gene To Promote Virus Infection in Marsupenaeus japonicus Shrimp. J Virol 2017; 91:JVI.02261-16. [PMID: 28179524 DOI: 10.1128/jvi.02261-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 01/27/2017] [Indexed: 01/10/2023] Open
Abstract
In eukaryotes, microRNAs (miRNAs) serve as regulators of many biological processes, including virus infection. An miRNA can generally target diverse genes during virus-host interactions. However, the regulation of gene expression by multiple miRNAs has not yet been extensively explored during virus infection. This study found that the Spaztle (Spz)-Toll-Dorsal-antilipopolysaccharide factor (ALF) signaling pathway plays a very important role in antiviral immunity against invasion of white spot syndrome virus (WSSV) in shrimp (Marsupenaeus japonicus). Dorsal, the central gene in the Toll pathway, was targeted by two viral miRNAs (WSSV-miR-N13 and WSSV-miR-N23) during WSSV infection. The regulation of Dorsal expression by viral miRNAs suppressed the Spz-Toll-Dorsal-ALF signaling pathway in shrimp in vivo, leading to virus infection. Our study contributes novel insights into the viral miRNA-mediated Toll signaling pathway during the virus-host interaction.IMPORTANCE An miRNA can target diverse genes during virus-host interactions. However, the regulation of gene expression by multiple miRNAs during virus infection has not yet been extensively explored. The results of this study indicated that the shrimp Dorsal gene, the central gene in the Toll pathway, was targeted by two viral miRNAs during infection with white spot syndrome virus. Regulation of Dorsal expression by viral miRNAs suppressed the Spz-Toll-Dorsal-ALF signaling pathway in shrimp in vivo, leading to virus infection. Our study provides new insight into the viral miRNA-mediated Toll signaling pathway in virus-host interactions.
Collapse
|
42
|
Bergman P, Seyedoleslami Esfahani S, Engström Y. Drosophila as a Model for Human Diseases—Focus on Innate Immunity in Barrier Epithelia. Curr Top Dev Biol 2017; 121:29-81. [DOI: 10.1016/bs.ctdb.2016.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
43
|
McLaughlin CN, Nechipurenko IV, Liu N, Broihier HT. A Toll receptor-FoxO pathway represses Pavarotti/MKLP1 to promote microtubule dynamics in motoneurons. J Cell Biol 2016; 214:459-74. [PMID: 27502486 PMCID: PMC4987293 DOI: 10.1083/jcb.201601014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023] Open
Abstract
McLaughlin et al. uncover a motoneuronal Toll-6–directed pathway that functions via dSARM and FoxO to attenuate microtubule stability through repression of Pavarotti/MKLP1 transcription. Genetic and pharmacological strategies reveal a novel requirement for dynamic synaptic microtubules in structural plasticity, which are established by Toll-6–FoxO signaling. FoxO proteins are evolutionarily conserved regulators of neuronal structure and function, yet the neuron-specific pathways within which they act are poorly understood. To elucidate neuronal FoxO function in Drosophila melanogaster, we first screened for FoxO’s upstream regulators and downstream effectors. On the upstream side, we present genetic and molecular pathway analyses indicating that the Toll-6 receptor, the Toll/interleukin-1 receptor domain adaptor dSARM, and FoxO function in a linear pathway. On the downstream side, we find that Toll-6–FoxO signaling represses the mitotic kinesin Pavarotti/MKLP1 (Pav-KLP), which itself attenuates microtubule (MT) dynamics. We next probed in vivo functions for this novel pathway and found that it is essential for axon transport and structural plasticity in motoneurons. We demonstrate that elevated expression of Pav-KLP underlies transport and plasticity phenotypes in pathway mutants, indicating that Toll-6–FoxO signaling promotes MT dynamics by limiting Pav-KLP expression. In addition to uncovering a novel molecular pathway, our work reveals an unexpected function for dynamic MTs in enabling rapid activity-dependent structural plasticity.
Collapse
Affiliation(s)
- Colleen N McLaughlin
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Inna V Nechipurenko
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Nan Liu
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
44
|
Analysis of the Contribution of Hemocytes and Autophagy to Drosophila Antiviral Immunity. J Virol 2016; 90:5415-5426. [PMID: 27009948 DOI: 10.1128/jvi.00238-16] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Antiviral immunity in the model organism Drosophila melanogaster involves the broadly active intrinsic mechanism of RNA interference (RNAi) and virus-specific inducible responses. Here, using a panel of six viruses, we investigated the role of hemocytes and autophagy in the control of viral infections. Injection of latex beads to saturate phagocytosis, or genetic depletion of hemocytes, resulted in decreased survival and increased viral titers following infection with Cricket paralysis virus (CrPV), Flock House virus (FHV), and vesicular stomatitis virus (VSV) but had no impact on Drosophila C virus (DCV), Sindbis virus (SINV), and Invertebrate iridescent virus 6 (IIV6) infection. In the cases of CrPV and FHV, apoptosis was induced in infected cells, which were phagocytosed by hemocytes. In contrast, VSV did not trigger any significant apoptosis but we confirmed that the autophagy gene Atg7 was required for full virus resistance, suggesting that hemocytes use autophagy to recognize the virus. However, this recognition does not depend on the Toll-7 receptor. Autophagy had no impact on DCV, CrPV, SINV, or IIV6 infection and was required for replication of the sixth virus, FHV. Even in the case of VSV, the increases in titers were modest in Atg7 mutant flies, suggesting that autophagy does not play a major role in antiviral immunity in Drosophila Altogether, our results indicate that, while autophagy plays a minor role, phagocytosis contributes to virus-specific immune responses in insects. IMPORTANCE Phagocytosis and autophagy are two cellular processes that involve lysosomal degradation and participate in Drosophila immunity. Using a panel of RNA and DNA viruses, we have addressed the contribution of phagocytosis and autophagy in the control of viral infections in this model organism. We show that, while autophagy plays a minor role, phagocytosis contributes to virus-specific immune responses in Drosophila This work brings to the front a novel facet of antiviral host defense in insects, which may have relevance in the control of virus transmission by vector insects or in the resistance of beneficial insects to viral pathogens.
Collapse
|
45
|
Gold KS, Brückner K. Macrophages and cellular immunity in Drosophila melanogaster. Semin Immunol 2016; 27:357-68. [PMID: 27117654 DOI: 10.1016/j.smim.2016.03.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/08/2016] [Indexed: 12/16/2022]
Abstract
The invertebrate Drosophila melanogaster has been a powerful model for understanding blood cell development and immunity. Drosophila is a holometabolous insect, which transitions through a series of life stages from embryo, larva and pupa to adulthood. In spite of this, remarkable parallels exist between Drosophila and vertebrate macrophages, both in terms of development and function. More than 90% of Drosophila blood cells (hemocytes) are macrophages (plasmatocytes), making this highly tractable genetic system attractive for studying a variety of questions in macrophage biology. In vertebrates, recent findings revealed that macrophages have two independent origins: self-renewing macrophages, which reside and proliferate in local microenvironments in a variety of tissues, and macrophages of the monocyte lineage, which derive from hematopoietic stem or progenitor cells. Like vertebrates, Drosophila possesses two macrophage lineages with a conserved dual ontogeny. These parallels allow us to take advantage of the Drosophila model when investigating macrophage lineage specification, maintenance and amplification, and the induction of macrophages and their progenitors by local microenvironments and systemic cues. Beyond macrophage development, Drosophila further serves as a paradigm for understanding the mechanisms underlying macrophage function and cellular immunity in infection, tissue homeostasis and cancer, throughout development and adult life.
Collapse
Affiliation(s)
| | - Katja Brückner
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; Department of Cell and Tissue Biology; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
46
|
Wang K, del Castillo C, Corre E, Pales Espinosa E, Allam B. Clam focal and systemic immune responses to QPX infection revealed by RNA-seq technology. BMC Genomics 2016; 17:146. [PMID: 26921237 PMCID: PMC4769524 DOI: 10.1186/s12864-016-2493-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/17/2016] [Indexed: 12/31/2022] Open
Abstract
Background The hard clam Mercenaria mercenaria is an important seafood species widely exploited along the eastern coasts of the United States and play a crucial role in coastal ecology and economy. Severe hard clam mortalities have been associated with the protistan parasite QPX (Quahog Parasite Unknown). QPX infection establishes in pallial organs with the lesions typically characterized as nodules, which represent inflammatory masses formed by hemocyte infiltration and encapsulation of parasites. QPX infection is known to induce host changes on both the whole-organism level and at specific lesion areas, which imply systemic and focal defense responses, respectively. However, little is known about the molecular mechanisms underlying these alterations. Results RNA-seq was performed using Illumina Hiseq 2000 (641 Million 100 bp reads) to characterize M. mercenaria focal and systemic immune responses to QPX. Transcripts were assembled and the expression levels were compared between nodule and healthy tissues from infected clams, and between these and tissues from healthy clams. De novo assembly reconstructed a consensus transcriptome of 62,980 sequences that was functionally-annotated. A total of 3,131 transcripts were identified as differentially expressed in different tissues. Results allowed the identification of host immune factors implicated in the systemic and focal responses against QPX and unraveled the pathways involved in parasite neutralization. Among transcripts significantly modulated upon host-pathogen interactions, those involved in non-self recognition, signal transduction and defense response were over-represented. Alterations in pathways regulating hemocyte focal adhesion, migration and apoptosis were also demonstrated. Conclusions Our study is the first attempt to thoroughly characterize M. mercenaria transcriptome and identify molecular features associated with QPX infection. It is also one of the first studies contrasting focal and systemic responses to infections in invertebrates using high-throughput sequencing. Results identified the molecular signatures of clam systemic and focal defense responses, to collectively mediate immune processes such as hemocyte recruitment and local inflammation. These investigations improve our understanding of bivalve immunity and provide molecular targets for probing the biological bases of clam resistance towards QPX. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2493-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kailai Wang
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY, 11794-5000, USA.
| | - Carmelo del Castillo
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY, 11794-5000, USA.
| | - Erwan Corre
- Analyses and Bioinformatics for Marine Science, Station Biologique de Roscoff, 29688, Roscoff Cedex, France.
| | - Emmanuelle Pales Espinosa
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY, 11794-5000, USA.
| | - Bassem Allam
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY, 11794-5000, USA.
| |
Collapse
|
47
|
Gerdts J, Summers DW, Milbrandt J, DiAntonio A. Axon Self-Destruction: New Links among SARM1, MAPKs, and NAD+ Metabolism. Neuron 2016; 89:449-60. [PMID: 26844829 PMCID: PMC4742785 DOI: 10.1016/j.neuron.2015.12.023] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Wallerian axon degeneration is a form of programmed subcellular death that promotes axon breakdown in disease and injury. Active degeneration requires SARM1 and MAP kinases, including DLK, while the NAD+ synthetic enzyme NMNAT2 prevents degeneration. New studies reveal that these pathways cooperate in a locally mediated axon destruction program, with NAD+ metabolism playing a central role. Here, we review the biology of Wallerian-type axon degeneration and discuss the most recent findings, with special emphasis on critical signaling events and their potential as therapeutic targets for axonopathy.
Collapse
Affiliation(s)
- Josiah Gerdts
- Department of Genetics, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA
| | - Daniel W Summers
- Department of Genetics, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, 660 Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
48
|
Zhou SM, Yuan XM, Liu S, Li M, Tao Z, Wang GL. Three novel Toll genes (PtToll1-3) identified from a marine crab, Portunus trituberculatus: Different tissue expression and response to pathogens. FISH & SHELLFISH IMMUNOLOGY 2015; 46:737-744. [PMID: 26238349 DOI: 10.1016/j.fsi.2015.07.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 06/04/2023]
Abstract
The Toll signaling pathway is one of the most important regulators of the immune response in both vertebrates and invertebrates. Herein, three novel Toll (PtToll1-3) cDNA sequences were cloned from the swimming crab, Portunus trituberculatus. PtToll1 has 1003 amino acid residues and consists of an extracellular domain containing 15 leucine-rich repeats (LRRs) and a cytoplasmic Toll/interleukin-1 receptor (TIR) domain of 139 residues. PtToll2 encodes 1196 peptides, with an extracellular domain containing 28 LRRs and a cytoplasmic TIR domain. PtToll3 is 1229 residues long and contains 26 LRRs and a cytoplasmic TIR domain. Based on sequence and phylogenetic analyses, PtToll1 distinctly clustered with almost all crustacean Tolls, except Litopenaeus vannamei Toll3. However, PtToll2 and PtToll3 were separated from most reported crustacean Tolls, which mostly clustered with Drosophila melanogaster (Dm) Toll8, L. vannamei Toll3, and DmToll6. Reverse transcription PCR and real-time quantitative PCR analyses showed that PtToll1 and PtToll3 were constitutively expressed in all tissues tested, but PtToll2 mRNA was only highly enriched in gills. Upon challenges with Vibrio alginolyticus, Candida lusitaniae, or white spot syndrome virus (WSSV), the three Tolls exhibited different responses: the PtToll1 transcript was up-regulated in response to C. lusitaniae or V. alginolyticus challenge, but did not respond to WSSV challenge; both PtToll2 and PtToll3 mRNAs were down-regulated 12 h after C. lusitaniae or V. alginolyticus infection. However, WSSV elicited the expression of PtToll2 at 6 h post-infection, but suppressed transcription of PtToll3 at 24 h post-infection. The study provides valuable data for understanding the role of Toll pathways in the host defense against microbial pathogens, which will facilitate future studies on host-pathogen interactions in crabs.
Collapse
Affiliation(s)
- Su-Ming Zhou
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Xue-Mei Yuan
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Shun Liu
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Meng Li
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Zhen Tao
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China
| | - Guo-Liang Wang
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, School of Marine Science, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
49
|
Dennison NJ, BenMarzouk-Hidalgo OJ, Dimopoulos G. MicroRNA-regulation of Anopheles gambiae immunity to Plasmodium falciparum infection and midgut microbiota. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 49:170-8. [PMID: 25445902 PMCID: PMC4447300 DOI: 10.1016/j.dci.2014.10.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/24/2014] [Accepted: 10/25/2014] [Indexed: 05/03/2023]
Abstract
Invasion of the malaria vector Anopheles gambiae midgut by Plasmodium parasites triggers transcriptional changes of immune genes that mediate the antiparasitic defense. This response is largely regulated by the Toll and Immune deficiency (IMD) pathways. To determine whether A. gambiae microRNAs (miRNAs) are involved in regulating the anti-Plasmodium defense, we showed that suppression of miRNA biogenesis results in increased resistance to Plasmodium falciparum infection. In silico analysis of A. gambiae immune effector genes identified multiple transcripts with miRNA binding sites. A comparative miRNA microarray abundance analysis of P. falciparum infected and naïve mosquito midgut tissues showed elevated abundance of miRNAs aga-miR-989 and aga-miR-305 in infected midguts. Antagomir inhibition of aga-miR-305 increased resistance to P. falciparum infection and suppressed the midgut microbiota. Conversely, treatment of mosquitoes with an artificial aga-miR-305 mimic increased susceptibility to P. falciparum infection and resulted in expansion of midgut microbiota, suggesting that aga-miR-305 acts as a P. falciparum and gut microbiota agonist by negatively regulating the mosquito immune response. In silico prediction of aga-miR-305 target genes identified several anti-Plasmodium effectors. Our study shows that A. gambiae aga-miR-305 regulates the anti-Plasmodium response and midgut microbiota, likely through post-transcriptional modification of immune effector genes.
Collapse
Affiliation(s)
- Nathan J Dennison
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Omar J BenMarzouk-Hidalgo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
50
|
Insulin- and warts-dependent regulation of tracheal plasticity modulates systemic larval growth during hypoxia in Drosophila melanogaster. PLoS One 2014; 9:e115297. [PMID: 25541690 PMCID: PMC4277339 DOI: 10.1371/journal.pone.0115297] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 11/22/2014] [Indexed: 01/07/2023] Open
Abstract
Adaptation to dynamic environmental cues during organismal development requires coordination of tissue growth with available resources. More specifically, the effects of oxygen availability on body size have been well-documented, but the mechanisms through which hypoxia restricts systemic growth have not been fully elucidated. Here, we characterize the larval growth and metabolic defects in Drosophila that result from hypoxia. Hypoxic conditions reduced fat body opacity and increased lipid droplet accumulation in this tissue, without eliciting lipid aggregation in hepatocyte-like cells called oenocytes. Additionally, hypoxia increased the retention of Dilp2 in the insulin-producing cells of the larval brain, associated with a reduction of insulin signaling in peripheral tissues. Overexpression of the wildtype form of the insulin receptor ubiquitously and in the larval trachea rendered larvae resistant to hypoxia-induced growth restriction. Furthermore, Warts downregulation in the trachea was similar to increased insulin receptor signaling during oxygen deprivation, which both rescued hypoxia-induced growth restriction, inhibition of tracheal molting, and developmental delay. Insulin signaling and loss of Warts function increased tracheal growth and augmented tracheal plasticity under hypoxic conditions, enhancing oxygen delivery during periods of oxygen deprivation. Our findings demonstrate a mechanism that coordinates oxygen availability with systemic growth in which hypoxia-induced reduction of insulin receptor signaling decreases plasticity of the larval trachea that is required for the maintenance of systemic growth during times of limiting oxygen availability.
Collapse
|