1
|
Shi C, Jin Z, Yu Y, Tang Z, Zhang Y, Qu C, Lin TH. Identification and characterization of a TLR4 homologue in Eriocheir sinensis based on structure analysis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 157:105192. [PMID: 38714270 DOI: 10.1016/j.dci.2024.105192] [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: 02/01/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/09/2024]
Abstract
Toll-like receptor 4 (TLR4) plays an essential role in the activation of innate immunity by recognizing diverse pathogenic components of bacteria. Six Tolls were found in Eriocheir sinensis but have not yet been identified as mammalian TLR4 homolog. For this purpose, we predicted three-dimensional (3D) structures of EsTolls (EsToll1-6) with AlphaFold2. 3D structure of LRRs and TIR most had high accuracy (pLDDT >70). By structure analysis, 3D structures of EsToll6 had a high overlap with HsTLR4. Moreover, we also predicted potential 11 hydrogen bonds and 3 salt bridges in the 3D structure of EsToll6-EsML1 complex. 18 hydrogen bonds and 7 salt bridges were predicted in EsToll6-EsML2 complex. Co-immunoprecipitation assay showed that EsToll6 could interact with EsML1 and EsML2, respectively. Importantly, TAK242 (a mammalian TLR4-specific inhibitor) could inhibit the generation of ROS stimulated by lipopolysaccharides (LPS) in EsToll6-EsML2-overexpression Hela cells. Collectively, these results implied that EsToll6 was a mammalian TLR4 homolog and provided a new insight for researching mammalian homologs in invertebrates.
Collapse
Affiliation(s)
- Chenchen Shi
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhixin Jin
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Yanping Yu
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China; Department of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, 361023, China
| | - Zhuyun Tang
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yuguo Zhang
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China; Department of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, 361023, China
| | - Chen Qu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| | - Ta-Hui Lin
- Fujian Provincial Key Laboratory of Functional and Clinical Translational Medicine, Xiamen Medical College, Xiamen, Fujian, 361023, China; State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, 361102, China; Department of Basic Medical Science, Xiamen Medical College, Xiamen, Fujian, 361023, China.
| |
Collapse
|
2
|
Critchlow JT, Prakash A, Zhong KY, Tate AT. Mapping the functional form of the trade-off between infection resistance and reproductive fitness under dysregulated immune signaling. PLoS Pathog 2024; 20:e1012049. [PMID: 38408106 PMCID: PMC10919860 DOI: 10.1371/journal.ppat.1012049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/07/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
Immune responses benefit organismal fitness by clearing parasites but also exact costs associated with immunopathology and energetic investment. Hosts manage these costs by tightly regulating the induction of immune signaling to curtail excessive responses and restore homeostasis. Despite the theoretical importance of turning off the immune response to mitigate these costs, experimentally connecting variation in the negative regulation of immune responses to organismal fitness remains a frontier in evolutionary immunology. In this study, we used a dose-response approach to manipulate the RNAi-mediated knockdown efficiency of cactus (IκBα), a central regulator of Toll pathway signal transduction in flour beetles (Tribolium castaneum). By titrating cactus activity across four distinct levels, we derived the shape of the relationship between immune response investment and traits associated with host fitness, including infection susceptibility, lifespan, fecundity, body mass, and gut homeostasis. Cactus knock-down increased the overall magnitude of inducible immune responses and delayed their resolution in a dsRNA dose-dependent manner, promoting survival and resistance following bacterial infection. However, these benefits were counterbalanced by dsRNA dose-dependent costs to lifespan, fecundity, body mass, and gut integrity. Our results allowed us to move beyond the qualitative identification of a trade-off between immune investment and fitness to actually derive its functional form. This approach paves the way to quantitatively compare the evolution and impact of distinct regulatory elements on life-history trade-offs and fitness, filling a crucial gap in our conceptual and theoretical models of immune signaling network evolution and the maintenance of natural variation in immune systems.
Collapse
Affiliation(s)
- Justin T Critchlow
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Arun Prakash
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Katherine Y Zhong
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ann T Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Evolutionary Studies Institute, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
3
|
Chou J, Ramroop JR, Saravia-Butler AM, Wey B, Lera MP, Torres ML, Heavner ME, Iyer J, Mhatre SD, Bhattacharya S, Govind S. Drosophila parasitoids go to space: Unexpected effects of spaceflight on hosts and their parasitoids. iScience 2024; 27:108759. [PMID: 38261932 PMCID: PMC10797188 DOI: 10.1016/j.isci.2023.108759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/15/2023] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
While fruit flies (Drosophila melanogaster) and humans exhibit immune system dysfunction in space, studies examining their immune systems' interactions with natural parasites in space are lacking. Drosophila parasitoid wasps modify blood cell function to suppress host immunity. In this study, naive and parasitized ground and space flies from a tumor-free control and a blood tumor-bearing mutant strain were examined. Inflammation-related genes were activated in space in both fly strains. Whereas control flies did not develop tumors, tumor burden increased in the space-returned tumor-bearing mutants. Surprisingly, control flies were more sensitive to spaceflight than mutant flies; many of their essential genes were downregulated. Parasitoids appeared more resilient than fly hosts, and spaceflight did not significantly impact wasp survival or the expression of their virulence genes. Previously undocumented mutant wasps with novel wing color and wing shape were isolated post-flight and will be invaluable for host-parasite studies on Earth.
Collapse
Affiliation(s)
- Jennifer Chou
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Johnny R. Ramroop
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Amanda M. Saravia-Butler
- KBR NASA Ames Research Center, Moffett Field, CA 94035, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Brian Wey
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
- PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Matthew P. Lera
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Medaya L. Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Bionetics, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Mary Ellen Heavner
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Janani Iyer
- KBR NASA Ames Research Center, Moffett Field, CA 94035, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Universities Space Research Association, Mountain View, CA 94043, USA
| | - Siddhita D. Mhatre
- KBR NASA Ames Research Center, Moffett Field, CA 94035, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | - Shubha Govind
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
- PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
4
|
Qin B, Yu S, Chen Q, Jin LH. Atg2 Regulates Cellular and Humoral Immunity in Drosophila. INSECTS 2023; 14:706. [PMID: 37623416 PMCID: PMC10455222 DOI: 10.3390/insects14080706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
Autophagy is a process that promotes the lysosomal degradation of cytoplasmic proteins and is highly conserved in eukaryotic organisms. Autophagy maintains homeostasis in organisms and regulates multiple developmental processes, and autophagy disruption is related to human diseases. However, the functional roles of autophagy in mediating innate immune responses are largely unknown. In this study, we sought to understand how Atg2, an autophagy-related gene, functions in the innate immunity of Drosophila melanogaster. The results showed that a large number of melanotic nodules were produced upon inhibition of Atg2. In addition, inhibiting Atg2 suppressed the phagocytosis of latex beads, Staphylococcus aureus and Escherichia coli; the proportion of Nimrod C1 (one of the phagocytosis receptors)-positive hemocytes also decreased. Moreover, inhibiting Atg2 altered actin cytoskeleton patterns, showing longer filopodia but with decreased numbers of filopodia. The expression of AMP-encoding genes was altered by inhibiting Atg2. Drosomycin was upregulated, and the transcript levels of Attacin-A, Diptericin and Metchnikowin were decreased. Finally, the above alterations caused by the inhibition of Atg2 prevented flies from resisting invading pathogens, showing that flies with low expression of Atg2 were highly susceptible to Staphylococcus aureus and Erwinia carotovora carotovora 15 infections. In conclusion, Atg2 regulated both cellular and humoral innate immunity in Drosophila. We have identified Atg2 as a crucial regulator in mediating the homeostasis of immunity, which further established the interactions between autophagy and innate immunity.
Collapse
Affiliation(s)
| | | | | | - Li Hua Jin
- College of Life Science, Northeast Forestry University, Harbin 150040, China; (B.Q.); (S.Y.); (Q.C.)
| |
Collapse
|
5
|
Critchlow JT, Prakash A, Zhong KY, Tate AT. Mapping the functional form of the trade-off between infection resistance and reproductive fitness under dysregulated immune signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552815. [PMID: 37645726 PMCID: PMC10461925 DOI: 10.1101/2023.08.10.552815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Immune responses benefit organismal fitness by clearing parasites but also exact costs associated with immunopathology and energetic investment. Hosts manage these costs by tightly regulating the induction of immune signaling to curtail excessive responses and restore homeostasis. Despite the theoretical importance of turning off the immune response to mitigate these costs, experimentally connecting variation in the negative regulation of immune responses to organismal fitness remains a frontier in evolutionary immunology. In this study, we used a dose-response approach to manipulate the RNAi-mediated knockdown efficiency of cactus (IκBα), a central regulator of Toll pathway signal transduction in flour beetles (Tribolium castaneum). By titrating cactus activity along a continuous gradient, we derived the shape of the relationship between immune response investment and traits associated with host fitness, including infection susceptibility, lifespan, fecundity, body mass, and gut homeostasis. Cactus knock-down increased the overall magintude of inducible immune responses and delayed their resolution in a dsRNA dose-dependent manner, promoting survival and resistance following bacterial infection. However, these benefits were counterbalanced by dsRNA dose-dependent costs to lifespan, fecundity, body mass, and gut integrity. Our results allowed us to move beyond the qualitative identification of a trade-off between immune investment and fitness to actually derive its functional form. This approach paves the way to quantitatively compare the evolution and impact of distinct regulatory elements on life-history trade-offs and fitness, filling a crucial gap in our conceptual and theoretical models of immune signaling network evolution and the maintenance of natural variation in immune systems.
Collapse
Affiliation(s)
- Justin T. Critchlow
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Arun Prakash
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Katherine Y. Zhong
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ann T. Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Evolutionary Studies Institute, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
6
|
Arguelles J, Lee J, Cardenas LV, Govind S, Singh S. In Silico Analysis of a Drosophila Parasitoid Venom Peptide Reveals Prevalence of the Cation-Polar-Cation Clip Motif in Knottin Proteins. Pathogens 2023; 12:pathogens12010143. [PMID: 36678491 PMCID: PMC9865768 DOI: 10.3390/pathogens12010143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
As generalist parasitoid wasps, Leptopilina heterotoma are highly successful on many species of fruit flies of the genus Drosophila. The parasitoids produce specialized multi-strategy extracellular vesicle (EV)-like structures in their venom. Proteomic analysis identified several immunity-associated proteins, including the knottin peptide, LhKNOT, containing the structurally conserved inhibitor cysteine knot (ICK) fold, which is present in proteins from diverse taxa. Our structural and docking analysis of LhKNOT's 36-residue core knottin fold revealed that in addition to the knottin motif itself, it also possesses a Cation-Polar-Cation (CPC) clip. The CPC clip motif is thought to facilitate antimicrobial activity in heparin-binding proteins. Surprisingly, a majority of ICKs tested also possess the CPC clip motif, including 75 bona fide plant and arthropod knottin proteins that share high sequence and/or structural similarity with LhKNOT. Like LhKNOT and these other 75 knottin proteins, even the Drosophila Drosomycin antifungal peptide, a canonical target gene of the fly's Toll-NF-kappa B immune pathway, contains this CPC clip motif. Together, our results suggest a possible defensive function for the parasitoid LhKNOT. The prevalence of the CPC clip motif, intrinsic to the cysteine knot within the knottin proteins examined here, suggests that the resultant 3D topology is important for their biochemical functions. The CPC clip is likely a highly conserved structural motif found in many diverse proteins with reported heparin binding capacity, including amyloid proteins. Knottins are targets for therapeutic drug development, and insights into their structure-function relationships will advance novel drug design.
Collapse
Affiliation(s)
- Joseph Arguelles
- Department of Biology, Brooklyn College, Brooklyn, NY 11210, USA
| | - Jenny Lee
- Department of Biology, Brooklyn College, Brooklyn, NY 11210, USA
| | - Lady V. Cardenas
- Department of Biology, The City College of New York, New York, NY 10031, USA
| | - Shubha Govind
- Department of Biology, The City College of New York, New York, NY 10031, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
- PhD Program in Biology, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Shaneen Singh
- Department of Biology, Brooklyn College, Brooklyn, NY 11210, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
- PhD Program in Biology, The Graduate Center of the City University of New York, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
7
|
Cardoso-Jaime V, Tikhe CV, Dong S, Dimopoulos G. The Role of Mosquito Hemocytes in Viral Infections. Viruses 2022; 14:v14102088. [PMID: 36298644 PMCID: PMC9608948 DOI: 10.3390/v14102088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Insect hemocytes are the only immune cells that can mount a humoral and cellular immune response. Despite the critical involvement of hemocytes in immune responses against bacteria, fungi, and parasites in mosquitoes, our understanding of their antiviral potential is still limited. It has been shown that hemocytes express humoral factors such as TEP1, PPO, and certain antimicrobial peptides that are known to restrict viral infections. Insect hemocytes also harbor the major immune pathways, such as JAK/STAT, TOLL, IMD, and RNAi, which are critical for the control of viral infection. Recent research has indicated a role for hemocytes in the regulation of viral infection through RNA interference and autophagy; however, the specific mechanism by which this regulation occurs remains uncharacterized. Conversely, some studies have suggested that hemocytes act as agonists of arboviral infection because they lack basal lamina and circulate throughout the whole mosquito, likely facilitating viral dissemination to other tissues such as salivary glands. In addition, hemocytes produce arbovirus agonist factors such as lectins, which enhance viral infection. Here, we summarize our current understanding of hemocytes’ involvement in viral infections.
Collapse
|
8
|
Yan Y, Sigle LT, Rinker DC, Estévez-Lao TY, Capra JA, Hillyer JF. The immune deficiency and c-Jun N-terminal kinase pathways drive the functional integration of the immune and circulatory systems of mosquitoes. Open Biol 2022; 12:220111. [PMID: 36069078 PMCID: PMC9449813 DOI: 10.1098/rsob.220111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune and circulatory systems of animals are functionally integrated. In mammals, the spleen and lymph nodes filter and destroy microbes circulating in the blood and lymph, respectively. In insects, immune cells that surround the heart valves (ostia), called periostial haemocytes, destroy pathogens in the areas of the body that experience the swiftest haemolymph (blood) flow. An infection recruits additional periostial haemocytes, amplifying heart-associated immune responses. Although the structural mechanics of periostial haemocyte aggregation have been defined, the genetic factors that regulate this process remain less understood. Here, we conducted RNA sequencing in the African malaria mosquito, Anopheles gambiae, and discovered that an infection upregulates multiple components of the immune deficiency (IMD) and c-Jun N-terminal kinase (JNK) pathways in the heart with periostial haemocytes. This upregulation is greater in the heart with periostial haemocytes than in the circulating haemocytes or the entire abdomen. RNA interference-based knockdown then showed that the IMD and JNK pathways drive periostial haemocyte aggregation and alter phagocytosis and melanization on the heart, thereby demonstrating that these pathways regulate the functional integration between the immune and circulatory systems. Understanding how insects fight infection lays the foundation for novel strategies that could protect beneficial insects and harm detrimental ones.
Collapse
Affiliation(s)
- Yan Yan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Leah T. Sigle
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - David C. Rinker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | - John A. Capra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA,Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
9
|
Rashidi M, Killiny N. In Silico Characterization and Gene Expression Analysis of Toll Signaling Pathway-Related Genes in Diaphorina citri. INSECTS 2022; 13:783. [PMID: 36135484 PMCID: PMC9500897 DOI: 10.3390/insects13090783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
The Asian citrus psyllid, Diaphorina citri is the main vector of citrus greening disease, also known as Huanglongbing (HLB). Currently, mitigating HLB depends on the control of D. citri using insecticides. To design innovative control strategies, we should investigate various biological aspects of D. citri at the molecular level. Herein we explored the Toll signaling system-related proteins in D. citri using in silico analyzes. Additionally, the transcripts of the identified genes were determined in all life stages from eggs to adults. Our findings reveal that D. citri genome possesses Toll signaling pathway-related genes similar to the insect model, Drosophila melanogaster, with slight differences. These genes include cact, TI, Myd88, Dif/DI, pll, tub, and spz encoding Cactus, Toll, Myeloid differentiation factor 88, Dorsal related immunity factor/Dorsal, Pelle, Tube, and Spaetzle, respectively. Unlike D. melanogaster, in D. citri Dorsal, immunity factor and Dorsal are the same protein. In addition, in D. citri, Pelle protein possesses a kinase domain, which is absent in Pelle of D. melanogaster. Gene expression analysis showed the transcript for cact, TI, Myd88, pll, tub, and spz are maximum in adults, suggesting the immunity increases with maturity. Instead, Dif/DI transcripts were maximal in eggs and adults and minimal in nymphal stages, indicating its role in embryonic development. The overall findings will help in designing pioneering control strategies of D. citri based on repressing its immunity by RNAi or CRISPR and combining that with biological control.
Collapse
|
10
|
Yu S, Luo F, Xu Y, Zhang Y, Jin LH. Drosophila Innate Immunity Involves Multiple Signaling Pathways and Coordinated Communication Between Different Tissues. Front Immunol 2022; 13:905370. [PMID: 35911716 PMCID: PMC9336466 DOI: 10.3389/fimmu.2022.905370] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune response provides the first line of defense against invading pathogens, and immune disorders cause a variety of diseases. The fruit fly Drosophila melanogaster employs multiple innate immune reactions to resist infection. First, epithelial tissues function as physical barriers to prevent pathogen invasion. In addition, macrophage-like plasmatocytes eliminate intruders through phagocytosis, and lamellocytes encapsulate large particles, such as wasp eggs, that cannot be phagocytosed. Regarding humoral immune responses, the fat body, equivalent to the mammalian liver, secretes antimicrobial peptides into hemolymph, killing bacteria and fungi. Drosophila has been shown to be a powerful in vivo model for studying the mechanism of innate immunity and host-pathogen interactions because Drosophila and higher organisms share conserved signaling pathways and factors. Moreover, the ease with which Drosophila genetic and physiological characteristics can be manipulated prevents interference by adaptive immunity. In this review, we discuss the signaling pathways activated in Drosophila innate immunity, namely, the Toll, Imd, JNK, JAK/STAT pathways, and other factors, as well as relevant regulatory networks. We also review the mechanisms by which different tissues, including hemocytes, the fat body, the lymph gland, muscles, the gut and the brain coordinate innate immune responses. Furthermore, the latest studies in this field are outlined in this review. In summary, understanding the mechanism underlying innate immunity orchestration in Drosophila will help us better study human innate immunity-related diseases.
Collapse
|
11
|
Hegde S, Sreejan A, Gadgil CJ, Ratnaparkhi GS. SUMOylation of Dorsal attenuates Toll/NF-κB signaling. Genetics 2022; 221:iyac081. [PMID: 35567478 PMCID: PMC9252280 DOI: 10.1093/genetics/iyac081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022] Open
Abstract
In Drosophila, Toll/NF-κB signaling plays key roles in both animal development and in host defense. The activation, intensity, and kinetics of Toll signaling are regulated by posttranslational modifications such as phosphorylation, SUMOylation, or ubiquitination that target multiple proteins in the Toll/NF-κB cascade. Here, we have generated a CRISPR-Cas9 edited Dorsal (DL) variant that is SUMO conjugation resistant. Intriguingly, embryos laid by dlSCR mothers overcome dl haploinsufficiency and complete the developmental program. This ability appears to be a result of higher transcriptional activation by DLSCR. In contrast, SUMOylation dampens DL transcriptional activation, ultimately conferring robustness to the dorso-ventral program. In the larval immune response, dlSCR animals show an increase in crystal cell numbers, stronger activation of humoral defense genes, and high cactus levels. A mathematical model that evaluates the contribution of the small fraction of SUMOylated DL (1-5%) suggests that it acts to block transcriptional activation, which is driven primarily by DL that is not SUMO conjugated. Our findings define SUMO conjugation as an important regulator of the Toll signaling cascade, in both development and host defense. Our results broadly suggest that SUMO attenuates DL at the level of transcriptional activation. Furthermore, we hypothesize that SUMO conjugation of DL may be part of a Ubc9-dependent mechanism that restrains Toll/NF-κB signaling.
Collapse
Affiliation(s)
- Sushmitha Hegde
- Biology, Indian Institute of Science Education & Research, Pune 411008, India
| | - Ashley Sreejan
- Chemical Engineering and Process Development Division, CSIR—National Chemical Laboratory, Pune 411008, India
| | - Chetan J Gadgil
- Chemical Engineering and Process Development Division, CSIR—National Chemical Laboratory, Pune 411008, India
- CSIR—Institute of Genomics and Integrative Biology, New Delhi 110020, India
| | | |
Collapse
|
12
|
Cheung YP, Park S, Pagtalunan J, Maringer K. The antiviral role of NF-κB-mediated immune responses and their antagonism by viruses in insects. J Gen Virol 2022; 103. [PMID: 35510990 DOI: 10.1099/jgv.0.001741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The antiviral role of innate immune responses mediated by the NF-κB family of transcription factors is well established in vertebrates but was for a long time less clear in insects. Insects encode two canonical NF-κB pathways, the Toll and Imd ('immunodeficiency') pathways, which are best characterised for their role in antibacterial and antifungal defence. An increasing body of evidence has also implicated NF-κB-mediated innate immunity in antiviral responses against some, but not all, viruses. Specific pattern recognition receptors (PRRs) and molecular events leading to NF-κB activation by viral pathogen-associated molecular patterns (PAMPs) have been elucidated for a number of viruses and insect species. Particularly interesting are recent findings indicating that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway detects viral RNA to activate NF-κB-regulated gene expression. We summarise the literature on virus-NF-κB pathway interactions across the class Insecta, with a focus on the dipterans Drosophila melanogaster and Aedes aegypti. We discuss potential reasons for differences observed between different virus-host combinations, and highlight similarities and differences between cGAS-STING signalling in insects versus vertebrates. Finally, we summarise the increasing number of known molecular mechanisms by which viruses antagonise NF-κB responses, which suggest that NF-κB-mediated immunity exerts strong evolutionary pressures on viruses. These developments in our understanding of insect antiviral immunity have relevance to the large number of insect species that impact on humans through their transmission of human, livestock and plant diseases, exploitation as biotechnology platforms, and role as parasites, pollinators, livestock and pests.
Collapse
Affiliation(s)
- Yin P Cheung
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Sohyun Park
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Justine Pagtalunan
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Kevin Maringer
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
| |
Collapse
|
13
|
Abubaker D, Baassiri A, Ghannam M, Al Outa A, Ghais A, Rahal E, Nasr R, Shirinian M. Expression of chronic myeloid leukemia oncogenes BCR-ABL P210 and BCR-ABL T315I affect cellular and humoral innate immunity in Drosophila melanogaster. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000551. [PMID: 35622506 PMCID: PMC9008464 DOI: 10.17912/micropub.biology.000551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 11/06/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm that results from a chromosomal translocation between chromosome 9 and chromosome 22. The resulting fusion gene ( BCR-ABL ) encodes a constitutively active BCR-ABL tyrosine kinase. Some mutations of this oncogene, especially the Threonine 315 to Isoleucine substitution of the ABL kinase is resistant to first and second-generation tyrosine kinase inhibitors (TKIs) conventionally used in CML therapy. We have previously validated a CML fruit fly model for drug screening using the adult fly compound eye. Here we expressed wild-type BCR-ABL P210 and mutated BCR-ABL T315I in Drosophila melanogaster hematopoietic system to understand the phenotypic consequences of this expression and its impact on innate immune pathways. Flies expressing both wild-type BCR-ABL P210 and mutant BCR-ABL T315I showed increased number of circulating hemocytes, disruption in sessile patterning of resident hemocytes, dysregulation in the humoral Toll, ImD, and JAK/STAT pathways at the mRNA level in both the 3 rd instar larva and adult stages. Of note, BCR-ABL T315I flies presented more severe phenotypes and a higher deviation in humoral dysregulation than BCR -ABL P210 flies pointing towards more complex oncogenic effect of this mutant which requires further investigation.
Collapse
Affiliation(s)
- Dana Abubaker
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amro Baassiri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon
| | - Mirna Ghannam
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amani Al Outa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon
| | - Ali Ghais
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Elias Rahal
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Lebanon
| | - Margret Shirinian
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
14
|
Abstract
Inflammatory response in Drosophila to sterile (axenic) injury in embryos and adults has received some attention in recent years, and most concentrate on the events at the injury site. Here we focus on the effect sterile injury has on the hematopoietic organ, the lymph gland, and the circulating blood cells in the larva, the developmental stage at which major events of hematopoiesis are evident. In mammals, injury activates Toll-like receptor/NF-κB signaling in macrophages, which then express and secrete secondary, proinflammatory cytokines. In Drosophila larvae, distal puncture injury of the body wall epidermis causes a rapid activation of Toll and Jun kinase (JNK) signaling throughout the hematopoietic system and the differentiation of a unique blood cell type, the lamellocyte. Furthermore, we find that Toll and JNK signaling are coupled in their activation. Secondary to this Toll/JNK response, a cytokine, Upd3, is induced as a Toll pathway transcriptional target, which then promotes JAK/STAT signaling within the blood cells. Toll and JAK/STAT signaling are required for the emergence of the injury-induced lamellocytes. This is akin to the derivation of specialized macrophages in mammalian systems. Upstream, at the injury site, a Duox- and peroxide-dependent signal causes the activation of the proteases Grass and SPE, needed for the activation of the Toll-ligand Spz, but microbial sensors or the proteases most closely associated with them during septic injury are not involved in the axenic inflammatory response.
Collapse
|
15
|
Nanda KP, Firdaus H. Dietary cadmium (Cd) reduces hemocyte level by induction of apoptosis in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 2021; 250:109188. [PMID: 34517133 DOI: 10.1016/j.cbpc.2021.109188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022]
Abstract
Drosophila melanogaster larvae ensure continuous proliferation and differentiation of hemocytes to maintain a fixed range of different blood cell types during its various stages of development. Variation in this number is often an indicator of animal well-being, its genotype or an effect of environmental perturbation, including exposure to heavy metals. The present study investigates the effect of Cd on larval hemocytes. Embryos were allowed to grow in metal media till third instar larvae and finally circulating hemocyte were collected. The number of major hemocytes, plasmatocytes and crystal cells was determined to be lowered in Cd exposed animals. Our results also showed modulation of antioxidant biology of Cd exposed hemocytes by changing the major antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT) activity, and decreasing reduced glutathione (GSH) levels in hemocytes suspended in the hemolymph. Acridine orange (AO) staining further revealed induction of apoptosis in hemocytes of metal treated larvae. Our results suggest a negative impact of Cd exposure on the hemocytes of the Drosophila larvae culminating in their lowered count by induction of apoptosis.
Collapse
Affiliation(s)
- Kumari Pragati Nanda
- Department of Life Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India
| | - Hena Firdaus
- Department of Life Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835205, Jharkhand, India.
| |
Collapse
|
16
|
Gong S, Zhang Y, Tian A, Deng W. Tumor models in various Drosophila tissues. WIREs Mech Dis 2021; 13:e1525. [PMID: 34730289 PMCID: PMC8566734 DOI: 10.1002/wsbm.1525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 01/07/2023]
Abstract
The development of cancer is a complex multistage process. Over the past few decades, the model organism Drosophila melanogaster has been crucial in identifying cancer-related genes and pathways and elucidating mechanisms underlying growth regulation in development. Investigations using Drosophila has yielded new insights into the molecular mechanisms involved in tumor initiation and progression. In this review, we describe various tumor models that have been developed in recent years using different Drosophila tissues, such as the imaginal tissue, the neural tissue, the gut, the ovary, and hematopoietic cells. We discuss underlying genetic alterations, cancer-like characteristics, as well as similarities and key differences among these models. We also discuss how disruptions in stem cell division and differentiation result in tumor formation in diverse tissues, and highlight new concepts developed using the fly model to understand context-dependent tumorigenesis. We further discuss the progress made in Drosophila to explore tumor-host interactions that involve the innate immune response to tumor growth and the cachexia wasting phenotype. This article is categorized under: Cancer > Genetics/Genomics/Epigenetics Cancer > Stem Cells and Development Cancer > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Shangyu Gong
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yichi Zhang
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Aiguo Tian
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Wu‐Min Deng
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
17
|
Eleftherianos I, Heryanto C, Bassal T, Zhang W, Tettamanti G, Mohamed A. Haemocyte-mediated immunity in insects: Cells, processes and associated components in the fight against pathogens and parasites. Immunology 2021; 164:401-432. [PMID: 34233014 PMCID: PMC8517599 DOI: 10.1111/imm.13390] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
The host defence of insects includes a combination of cellular and humoral responses. The cellular arm of the insect innate immune system includes mechanisms that are directly mediated by haemocytes (e.g., phagocytosis, nodulation and encapsulation). In addition, melanization accompanying coagulation, clot formation and wound healing, nodulation and encapsulation processes leads to the formation of cytotoxic redox-cycling melanin precursors and reactive oxygen and nitrogen species. However, demarcation between cellular and humoral immune reactions as two distinct categories is not straightforward. This is because many humoral factors affect haemocyte functions and haemocytes themselves are an important source of many humoral molecules. There is also a considerable overlap between cellular and humoral immune functions that span from recognition of foreign intruders to clot formation. Here, we review these immune reactions starting with the cellular mechanisms that limit haemolymph loss and participate in wound healing and clot formation and advancing to cellular functions that are critical in restricting pathogen movement and replication. This information is important because it highlights that insect cellular immunity is controlled by a multilayered system, different components of which are activated by different pathogens or during the different stages of the infection.
Collapse
Affiliation(s)
- Ioannis Eleftherianos
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Christa Heryanto
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Taha Bassal
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| | - Wei Zhang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural BioengineeringKey Laboratory of Green Pesticide and Agricultural BioengineeringMinistry of EducationGuizhou UniversityGuiyangChina
| | - Gianluca Tettamanti
- Department of Biotechnology and Life SciencesUniversity of InsubriaVareseItaly
- BAT Center‐Interuniversity Center for Studies on Bioinspired Agro‐Environmental TechnologyUniversity of Napoli Federico IINapoliItaly
| | - Amr Mohamed
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| |
Collapse
|
18
|
Liu Y, Chang J, Yang C, Zhang T, Chen X, Shi R, Liang Y, Xia Q, Ma S. Genome-wide CRISPR-Cas9 screening in Bombyx mori reveals the toxicological mechanisms of environmental pollutants, fluoride and cadmium. JOURNAL OF HAZARDOUS MATERIALS 2021; 410:124666. [PMID: 33279320 DOI: 10.1016/j.jhazmat.2020.124666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/27/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
Fluoride and cadmium, two typical environmental pollutants, have been extensively existed in the ecosystem and severely injured various organisms including humans. To explore the toxicological properties and the toxicological mechanism of fluoride and cadmium in silkworm, we perform a CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) -based functional genomic screen, which can directly measure the genetic requirement of genes in response to the pollutants. Our screen identifies 751 NaF-resistance genes, 753 NaF-sensitive genes, 757 CdCl2-resistance genes, and 725 CdCl2-sensitive genes. The top-ranked resistant genes are experimentally verified and the results show that their loss conferred resistance to fluoride or cadmium. Functional analysis of the resistant- and sensitive-genes demonstrates enrichment of multiple signaling pathways, among which the MAPK signaling pathway and DNA damage and repair are both required for fluoride- or cadmium-induced cell death, whereas the Toll and Imd signaling pathway and Autophagy are fluoride- or cadmium-specific. Moreover, we confirm that these pathways are truly involved in the toxicological mechanism in both cultured cells and individual tissues. Our results supply potential targets for rescuing the biohazards of fluoride and cadmium in silkworm, and reveal the feasible toxicological mechanism, which highlights the role of functional genomic screens in elucidating the toxicity mechanisms of environmental pollutants.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Jiasong Chang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Chengfei Yang
- Department of Urology, The Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Tong Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Xiaoxu Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Run Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Yan Liang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China.
| | - Sanyuan Ma
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; Biological Science Research Center, Southwest University, Chongqing 400716, China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China; Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China.
| |
Collapse
|
19
|
Lü J, Liu ZQ, Guo W, Guo MJ, Chen SM, Yang CX, Zhang YJ, Pan HP. Oral delivery of dsHvlwr is a feasible method for managing the pest Henosepilachna vigintioctopunctata (Coleoptera: Coccinellidae). INSECT SCIENCE 2021; 28:509-520. [PMID: 32240577 DOI: 10.1111/1744-7917.12784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/18/2020] [Accepted: 03/16/2020] [Indexed: 06/11/2023]
Abstract
RNA interference (RNAi) techniques have emerged as powerful tools that facilitate development of novel management strategies for insect pests, such as Henosepilachna vigintioctopunctata (Coleoptera: Coccinellidae), which is a major pest of solanaceous plants in Asia. In this study, the potential of oral delivery of in vitro-synthesized and bacterially expressed double-stranded H. vigintioctopunctata lesswright (lwr) gene (dsHvlwr) to manage of H. vigintioctopunctata was investigated. Our results showed that the gene Hvlwr had a 480-bp open reading frame and encoded a 160-amino acid protein. Hvlwr expression levels were greater in the fat body than other tissue types. Hvlwr silencing led to greater H. vigintioctopunctata mortality rates and appeared to be time- and partially dose-dependent, likely as a result of the number of hemocytes increasing with dsRNA concentration, but decreasing with time. Bacterially expressed dsHvlwr that was applied to leaf discs caused 88%, 66%, and 36% mortality in 1st instars, 3rd instars, and adults after 10, 10, and 14 d, respectively; when applied to living plants, there was greater mortality in 1st and 3rd instars, but there was no effect on adults. Furthermore, dsHvlwr led to improved plant protection against H. vigintioctopunctata. Our study shows an effective dietary RNAi response in H. vigintioctopunctata and that Hvlwr is a promising RNAi target gene for control of this pest species.
Collapse
Affiliation(s)
- Jing Lü
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Zhuo-Qi Liu
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Wei Guo
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Mu-Juan Guo
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Shi-Min Chen
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, South China Agricultural University, Guangzhou, China
| | - Chun-Xiao Yang
- Lingnan Guangdong Laboratory of Modern Agriculture, Guangzhou, China
| | - You-Jun Zhang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hui-Peng Pan
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, South China Agricultural University, Guangzhou, China
| |
Collapse
|
20
|
Mase A, Augsburger J, Brückner K. Macrophages and Their Organ Locations Shape Each Other in Development and Homeostasis - A Drosophila Perspective. Front Cell Dev Biol 2021; 9:630272. [PMID: 33777939 PMCID: PMC7991785 DOI: 10.3389/fcell.2021.630272] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Across the animal kingdom, macrophages are known for their functions in innate immunity, but they also play key roles in development and homeostasis. Recent insights from single cell profiling and other approaches in the invertebrate model organism Drosophila melanogaster reveal substantial diversity among Drosophila macrophages (plasmatocytes). Together with vertebrate studies that show genuine expression signatures of macrophages based on their organ microenvironments, it is expected that Drosophila macrophage functional diversity is shaped by their anatomical locations and systemic conditions. In vivo evidence for diverse macrophage functions has already been well established by Drosophila genetics: Drosophila macrophages play key roles in various aspects of development and organogenesis, including embryogenesis and development of the nervous, digestive, and reproductive systems. Macrophages further maintain homeostasis in various organ systems and promote regeneration following organ damage and injury. The interdependence and interplay of tissues and their local macrophage populations in Drosophila have implications for understanding principles of organ development and homeostasis in a wide range of species.
Collapse
Affiliation(s)
- Anjeli Mase
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Jordan Augsburger
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Katja Brückner
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
21
|
Gautam DK, Chimata AV, Gutti RK, Paddibhatla I. Comparative hematopoiesis and signal transduction in model organisms. J Cell Physiol 2021; 236:5592-5619. [PMID: 33492678 DOI: 10.1002/jcp.30287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022]
Abstract
Hematopoiesis is a continuous phenomenon involving the formation of hematopoietic stem cells (HSCs) giving rise to diverse functional blood cells. This developmental process of hematopoiesis is evolutionarily conserved, yet comparably different in various model organisms. Vertebrate HSCs give rise to all types of mature cells of both the myeloid and the lymphoid lineages sequentially colonizing in different anatomical tissues. Signal transduction in HSCs facilitates their potency and specifies branching of lineages. Understanding the hematopoietic signaling pathways is crucial to gain insights into their deregulation in several blood-related disorders. The focus of the review is on hematopoiesis corresponding to different model organisms and pivotal role of indispensable hematopoietic pathways. We summarize and discuss the fundamentals of blood formation in both invertebrate and vertebrates, examining the requirement of key signaling nexus in hematopoiesis. Knowledge obtained from such comparative studies associated with developmental dynamics of hematopoiesis is beneficial to explore the therapeutic options for hematopoietic diseases.
Collapse
Affiliation(s)
- Dushyant Kumar Gautam
- Department of Biochemistry, School of Life Sciences (SLS), University of Hyderabad, Hyderabad, Telangana, India
| | | | - Ravi Kumar Gutti
- Department of Biochemistry, School of Life Sciences (SLS), University of Hyderabad, Hyderabad, Telangana, India
| | - Indira Paddibhatla
- Department of Biochemistry, School of Life Sciences (SLS), University of Hyderabad, Hyderabad, Telangana, India
| |
Collapse
|
22
|
Zheng X, Li S, Si Y, Hu J, Xia Y. Locust can detect β-1, 3-glucan of the fungal pathogen before penetration and defend infection via the Toll signaling pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 106:103636. [PMID: 32014469 DOI: 10.1016/j.dci.2020.103636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 06/10/2023]
Abstract
The timing and mechanism by which a host insect initiates an immune response are critical to successful defense against infection. Pathogen recognition, a prerequisite for host defense, has long been recognized to take place during the insect epidermis invasion by fungus. Here we report that insect can sense the fungal pathogen before host cuticle is penetrated by fungus. We discovered the upstream pattern recognition receptor (PRR) genes of the Toll pathway were upregulated in both the integument and fat body early during fungal germination on the epicuticle of Locusta migratoria manilensis. The Toll signaling pathway was strongly activated in the fat body at the penetration stage. RNAi of Myd88 increased the susceptibility of locusts to fungal infection, but that of Cactus showed the opposite effect. In addition, β-1, 3-glucan (laminarin), the main component of the cell wall of the pathogenic fungus Metarhizium acridum, was capable of activating the Toll signaling pathway (Spaetzle and Cactus) when it was applied on the host cuticle. These results demonstrate that host epidemis can effectively defend fungal infection by detecting β-1, 3-glucan on the fungal cell wall and activate the Toll signaling pathway even before fungal penetration.
Collapse
Affiliation(s)
- Xiaoli Zheng
- Genetic Engineering Research Center, School of Life Science, Chongqing University, Chongqing, 400030, PR China; College of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, PR China; Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, 400030, PR China; Key Laboratory of Gene Function and Regulation Technologies under Chongqing Municipal Education Commission, Chongqing, 400030, PR China
| | - Sai Li
- Genetic Engineering Research Center, School of Life Science, Chongqing University, Chongqing, 400030, PR China; Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, 400030, PR China; Key Laboratory of Gene Function and Regulation Technologies under Chongqing Municipal Education Commission, Chongqing, 400030, PR China
| | - Yang Si
- Genetic Engineering Research Center, School of Life Science, Chongqing University, Chongqing, 400030, PR China; Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, 400030, PR China; Key Laboratory of Gene Function and Regulation Technologies under Chongqing Municipal Education Commission, Chongqing, 400030, PR China
| | - Jun Hu
- Genetic Engineering Research Center, School of Life Science, Chongqing University, Chongqing, 400030, PR China; Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, 400030, PR China; Key Laboratory of Gene Function and Regulation Technologies under Chongqing Municipal Education Commission, Chongqing, 400030, PR China
| | - Yuxian Xia
- Genetic Engineering Research Center, School of Life Science, Chongqing University, Chongqing, 400030, PR China; Chongqing Engineering Research Center for Fungal Insecticide, Chongqing, 400030, PR China; Key Laboratory of Gene Function and Regulation Technologies under Chongqing Municipal Education Commission, Chongqing, 400030, PR China.
| |
Collapse
|
23
|
Paddibhatla I, Gautam DK, Mishra RK. SETDB1 modulates the differentiation of both the crystal cells and the lamellocytes in Drosophila. Dev Biol 2019; 456:74-85. [DOI: 10.1016/j.ydbio.2019.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 01/10/2023]
|
24
|
Harsh S, Heryanto C, Eleftherianos I. Intestinal lipid droplets as novel mediators of host-pathogen interaction in Drosophila. Biol Open 2019; 8:bio.039040. [PMID: 31278163 PMCID: PMC6679391 DOI: 10.1242/bio.039040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Lipid droplets (LDs) are lipid-carrying multifunctional organelles, which might also interact with pathogens and influence the host immune response. However, the exact nature of these interactions remains currently unexplored. Here we show that systemic infection of Drosophila adult flies with non-pathogenic Escherichia coli, the extracellular bacterial pathogen Photorhabdus luminescens or the facultative intracellular pathogen Photorhabdus asymbiotica results in intestinal steatosis marked by lipid accumulation in the midgut. Accumulation of LDs in the midgut also correlates with increased whole-body lipid levels characterized by increased expression of genes regulating lipogenesis. The lipid-enriched midgut further displays reduced expression of the enteroendocrine-secreted hormone, Tachykinin. The observed lipid accumulation requires the Gram-negative cell wall pattern recognition molecule, PGRP-LC, but not PGRP-LE, for the humoral immune response. Altogether, our findings indicate that Drosophila LDs are inducible organelles, which can serve as markers for inflammation and, depending on the nature of the challenge, they can dictate the outcome of the infection. Summary: Lipid droplets are inducible organelles, act as inflammatory markers and, depending on the nature of challenge, can dictate the outcome of the infection.
Collapse
Affiliation(s)
- Sneh Harsh
- Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington DC 20052, USA
| | - Christa Heryanto
- Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington DC 20052, USA
| | - Ioannis Eleftherianos
- Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington DC 20052, USA
| |
Collapse
|
25
|
Lethal Interaction of Nuclear and Mitochondrial Genotypes in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2019; 9:2225-2234. [PMID: 31076384 PMCID: PMC6643882 DOI: 10.1534/g3.119.400315] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Drosophilamelanogaster, like most animal species, displays considerable genetic variation in both nuclear and mitochondrial DNA (mtDNA). Here we tested whether any of four natural mtDNA variants was able to modify the effect of the phenotypically mild, nuclear tko25t mutation, affecting mitochondrial protein synthesis. When combined with tko25t, the mtDNA from wild strain KSA2 produced pupal lethality, accompanied by the presence of melanotic nodules in L3 larvae. KSA2 mtDNA, which carries a substitution at a conserved residue of cytochrome b that is predicted to be involved in subunit interactions within respiratory complex III, conferred drastically decreased respiratory capacity and complex III activity in the tko25t but not a wild-type nuclear background. The complex III inhibitor antimycin A was able to phenocopy effects of the tko25t mutation in the KSA2 mtDNA background. This is the first report of a lethal, nuclear-mitochondrial interaction within a metazoan species, representing a paradigm for understanding genetic interactions between nuclear and mitochondrial genotype relevant to human health and disease.
Collapse
|
26
|
Araki M, Kurihara M, Kinoshita S, Awane R, Sato T, Ohkawa Y, Inoue YH. Anti-tumour effects of antimicrobial peptides, components of the innate immune system, against haematopoietic tumours in Drosophila mxc mutants. Dis Model Mech 2019; 12:dmm037721. [PMID: 31160313 PMCID: PMC6602314 DOI: 10.1242/dmm.037721] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
The innate immune response is the first line of defence against microbial infections. In Drosophila, two major pathways of the innate immune system (the Toll- and Imd-mediated pathways) induce the synthesis of antimicrobial peptides (AMPs) within the fat body. Recently, it has been reported that certain cationic AMPs exhibit selective cytotoxicity against human cancer cells; however, little is known about their anti-tumour effects. Drosophila mxcmbn1 mutants exhibit malignant hyperplasia in a larval haematopoietic organ called the lymph gland (LG). Here, using RNA-seq analysis, we found many immunoresponsive genes, including those encoding AMPs, to be upregulated in these mutants. Downregulation of these pathways by either a Toll or imd mutation enhanced the tumour phenotype of the mxc mutants. Conversely, ectopic expression of each of five different AMPs in the fat body significantly suppressed the LG hyperplasia phenotype in the mutants. Thus, we propose that the Drosophila innate immune system can suppress the progression of haematopoietic tumours by inducing AMP gene expression. Overexpression of any one of the five AMPs studied resulted in enhanced apoptosis in mutant LGs, whereas no apoptotic signals were detected in controls. We observed that two AMPs, Drosomycin and Defensin, were taken up by circulating haemocyte-like cells, which were associated with the LG regions and showed reduced cell-to-cell adhesion in the mutants. By contrast, the AMP Diptericin was directly localised at the tumour site without intermediating haemocytes. These results suggest that AMPs have a specific cytotoxic effect that enhances apoptosis exclusively in the tumour cells.
Collapse
Affiliation(s)
- Mayo Araki
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Massanori Kurihara
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Suzuko Kinoshita
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Rie Awane
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Tetsuya Sato
- Medical Institute of Bioregulation, Kyushu University, Kyushu 812-0054, Japan
| | - Yasuyuki Ohkawa
- Medical Institute of Bioregulation, Kyushu University, Kyushu 812-0054, Japan
| | - Yoshihiro H Inoue
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| |
Collapse
|
27
|
Banerjee U, Girard JR, Goins LM, Spratford CM. Drosophila as a Genetic Model for Hematopoiesis. Genetics 2019; 211:367-417. [PMID: 30733377 PMCID: PMC6366919 DOI: 10.1534/genetics.118.300223] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
In this FlyBook chapter, we present a survey of the current literature on the development of the hematopoietic system in Drosophila The Drosophila blood system consists entirely of cells that function in innate immunity, tissue integrity, wound healing, and various forms of stress response, and are therefore functionally similar to myeloid cells in mammals. The primary cell types are specialized for phagocytic, melanization, and encapsulation functions. As in mammalian systems, multiple sites of hematopoiesis are evident in Drosophila and the mechanisms involved in this process employ many of the same molecular strategies that exemplify blood development in humans. Drosophila blood progenitors respond to internal and external stress by coopting developmental pathways that involve both local and systemic signals. An important goal of these Drosophila studies is to develop the tools and mechanisms critical to further our understanding of human hematopoiesis during homeostasis and dysfunction.
Collapse
Affiliation(s)
- Utpal Banerjee
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
- Molecular Biology Institute, University of California, Los Angeles, California 90095
- Department of Biological Chemistry, University of California, Los Angeles, California 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| | - Juliet R Girard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Lauren M Goins
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Carrie M Spratford
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| |
Collapse
|
28
|
Green N, Walker J, Bontrager A, Zych M, Geisbrecht ER. A tissue communication network coordinating innate immune response during muscle stress. J Cell Sci 2018; 131:jcs.217943. [PMID: 30478194 DOI: 10.1242/jcs.217943] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/15/2018] [Indexed: 12/23/2022] Open
Abstract
Complex tissue communication networks function throughout an organism's lifespan to maintain tissue homeostasis. Using the genetic model Drosophila melanogaster, we have defined a network of immune responses that are activated following the induction of muscle stresses, including hypercontraction, detachment and oxidative stress. Of these stressors, loss of the genes that cause muscle detachment produced the strongest levels of JAK-STAT activation. In one of these mutants, fondue (fon), we also observe hemocyte recruitment and the accumulation of melanin at muscle attachment sites (MASs), indicating a broad involvement of innate immune responses upon muscle detachment. Loss of fon results in pathogen-independent Toll signaling in the fat body and increased expression of the Toll-dependent antimicrobial peptide Drosomycin. Interestingly, genetic interactions between fon and various Toll pathway components enhance muscle detachment. Finally, we show that JAK-STAT and Toll signaling are capable of reciprocal activation in larval tissues. We propose a model of tissue communication for the integration of immune responses at the local and systemic level in response to altered muscle physiology.
Collapse
Affiliation(s)
- Nicole Green
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Justin Walker
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Alexandria Bontrager
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Molly Zych
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
29
|
Li H, Yin B, Wang S, Fu Q, Xiao B, Lǚ K, He J, Li C. RNAi screening identifies a new Toll from shrimp Litopenaeus vannamei that restricts WSSV infection through activating Dorsal to induce antimicrobial peptides. PLoS Pathog 2018; 14:e1007109. [PMID: 30256850 PMCID: PMC6175524 DOI: 10.1371/journal.ppat.1007109] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/08/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022] Open
Abstract
The function of Toll pathway defense against bacterial infection has been well established in shrimp, however how this pathway responds to viral infection is still largely unknown. In this study, we report the Toll4-Dorsal-AMPs cascade restricts the white spot syndrome virus (WSSV) infection of shrimp. A total of nine Tolls from Litopenaeus vannamei namely Toll1-9 are identified, and RNAi screening in vivo reveals the Toll4 is important for shrimp to oppose WSSV infection. Knockdown of Toll4 results in elevated viral loads and renders shrimp more susceptible to WSSV. Furthermore, Toll4 could be a one of upstream pattern recognition receptor (PRR) to detect WSSV, and thereby leading to nuclear translocation and phosphorylation of Dorsal, the known NF-κB transcription factor of the canonical Toll pathway. More importantly, silencing of Toll4 and Dorsal contributes to impaired expression of a specific set of antimicrobial peptides (AMPs) such as anti-LPS-factor (ALF) and lysozyme (LYZ) family, which exert potent anti-WSSV activity. Two AMPs of ALF1 and LYZ1 as representatives are demonstrated to have the ability to interact with several WSSV structural proteins to inhibit viral infection. Taken together, we therefore identify that the Toll4-Dorsal pathway mediates strong resistance to WSSV infection by inducing some specific AMPs. The TLR pathway mediated antiviral immune response is well identified in mammals, yet, Toll pathway governing this protection in invertebrates remains unknown. In the present study, we uncover that a shrimp Toll4 from a total of nine Tolls in L. vannamei confers resistance to WSSV thought inducing the NF-κB transcription factor Dorsal to inspire the production of some antimicrobial peptides (AMPs) with antiviral activity. The anti-LPS-factor (ALF) and lysozyme (LYZ) family are identified as the Toll4-Dorsal pathway targeted genes with the ability to interact with viral structural proteins in response to WSSV infection. These results suggest that the Toll receptor induces the expression of AMPs with antiviral activity could be a general antiviral mechanism in invertebrates and Toll pathway established antiviral defense could be conserved during evolution.
Collapse
Affiliation(s)
- Haoyang Li
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Bin Yin
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Sheng Wang
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Qihui Fu
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Bang Xiao
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Kai Lǚ
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
| | - Jianguo He
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
- * E-mail: (JH); (CL)
| | - Chaozheng Li
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, P. R. China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, P. R. China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), Sun Yat-sen University, Guangzhou, P. R. China
- * E-mail: (JH); (CL)
| |
Collapse
|
30
|
Baldeosingh R, Gao H, Wu X, Fossett N. Hedgehog signaling from the Posterior Signaling Center maintains U-shaped expression and a prohemocyte population in Drosophila. Dev Biol 2018; 441:132-145. [PMID: 29966604 PMCID: PMC6064674 DOI: 10.1016/j.ydbio.2018.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022]
Abstract
Hematopoietic progenitor choice between multipotency and differentiation is tightly regulated by intrinsic factors and extrinsic signals from the surrounding microenvironment. The Drosophila melanogaster hematopoietic lymph gland has emerged as a powerful tool to investigate mechanisms that regulate hematopoietic progenitor choice in vivo. The lymph gland contains progenitor cells, which share key characteristics with mammalian hematopoietic progenitors such as quiescence, multipotency and niche-dependence. The lymph gland is zonally arranged, with progenitors located in medullary zone, differentiating cells in the cortical zone, and the stem cell niche or Posterior Signaling Center (PSC) residing at the base of the medullary zone (MZ). This arrangement facilitates investigations into how signaling from the microenvironment controls progenitor choice. The Drosophila Friend of GATA transcriptional regulator, U-shaped, is a conserved hematopoietic regulator. To identify additional novel intrinsic and extrinsic regulators that interface with U-shaped to control hematopoiesis, we conducted an in vivo screen for factors that genetically interact with u-shaped. Smoothened, a downstream effector of Hedgehog signaling, was one of the factors identified in the screen. Here we report our studies that characterized the relationship between Smoothened and U-shaped. We showed that the PSC and Hedgehog signaling are required for U-shaped expression and that U-shaped is an important intrinsic progenitor regulator. These observations identify a potential link between the progenitor regulatory machinery and extrinsic signals from the PSC. Furthermore, we showed that both Hedgehog signaling and the PSC are required to maintain a subpopulation of progenitors. This led to a delineation of PSC-dependent versus PSC-independent progenitors and provided further evidence that the MZ progenitor population is heterogeneous. Overall, we have identified a connection between a conserved hematopoietic master regulator and a putative stem cell niche, which adds to our understanding of how signals from the microenvironment regulate progenitor multipotency.
Collapse
Affiliation(s)
- Rajkumar Baldeosingh
- Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hongjuan Gao
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaorong Wu
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nancy Fossett
- Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
31
|
Pei J, Kinch LN, Grishin NV. FlyXCDB—A Resource for Drosophila Cell Surface and Secreted Proteins and Their Extracellular Domains. J Mol Biol 2018; 430:3353-3411. [DOI: 10.1016/j.jmb.2018.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/31/2018] [Accepted: 06/02/2018] [Indexed: 02/06/2023]
|
32
|
Rhodes VL, Thomas MB, Michel K. The interplay between dose and immune system activation determines fungal infection outcome in the African malaria mosquito, Anopheles gambiae. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 85:125-133. [PMID: 29649553 PMCID: PMC5935592 DOI: 10.1016/j.dci.2018.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 05/27/2023]
Abstract
The Toll pathway is a central regulator of antifungal immunity in insects. In mosquitoes, the Toll pathway affects infections with the fungal entomopathogen, Beauveria bassiana, which is considered a potential mosquito biopesticide. We report here the use of B. bassiana strain I93-825 in Anopheles gambiae to analyze the impact of Toll pathway modulation on mosquito survival. Exposure to a narrow dose range of conidia by direct contact decreased mosquito longevity and median survival. In addition, fungal exposure dose correlated positively and linearly with hazard ratio. Increased Toll signaling by knockdown of its inhibitor, cactus, decreased survivorship of uninfected females, increased mosquito survival after low dose B. bassiana exposure, but had little effect following exposure to higher doses. This observed trade-off could have implications for development of B. bassiana as a prospective vector control tool. On the one hand, selection for small increases in mosquito immune signaling across a narrow dose range could impair efficacy of B. bassiana. On the other hand, costs of immunity and the capacity for higher doses of fungus to overwhelm immune responses could limit evolution of resistance.
Collapse
Affiliation(s)
- Victoria L Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Matthew B Thomas
- Department of Entomology and Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
33
|
Yu S, Luo F, Jin LH. The Drosophila lymph gland is an ideal model for studying hematopoiesis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:60-69. [PMID: 29191551 DOI: 10.1016/j.dci.2017.11.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/30/2017] [Accepted: 11/26/2017] [Indexed: 06/07/2023]
Abstract
Hematopoiesis in Drosophila melanogaster occurs throughout the entire life cycle, from the embryo to adulthood. The healthy lymph gland, as a hematopoietic organ during the larval stage, can give rise to two mature types of hemocytes, plasmatocytes and crystal cells, which persist into the pupal and adult stages. Homeostasis of the lymph gland is tightly controlled by a series of conserved factors and signaling pathways, which also play key roles in mammalian hematopoiesis. Thus, revealing the hematopoietic mechanisms in Drosophila will advance our understanding of hematopoietic stem cells and their niche as well as leukemia in mammals. In addition, the lymph gland employs a battery of strategies to produce lamellocytes, another type of mature hemocyte, to fight against parasitic wasp eggs, making the lymph gland an important immunological organ. In this review, the developmental process of the lymph gland and the regulatory networks of hematopoiesis are summarized. Moreover, we outline the current knowledge and novel insight into homeostasis of the lymph gland.
Collapse
Affiliation(s)
- Shichao Yu
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Fangzhou Luo
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China.
| |
Collapse
|
34
|
Abstract
The power and ease of Drosophila genetics and the medical relevance of mosquito-transmitted viruses have made dipterans important model organisms in antiviral immunology. Studies of virus-host interactions at the molecular and population levels have illuminated determinants of resistance to virus infection. Here, we review the sources and nature of variation in antiviral immunity and virus susceptibility in model dipteran insects, specifically the fruit fly Drosophila melanogaster and vector mosquitoes of the genera Aedes and Culex. We first discuss antiviral immune mechanisms and describe the virus-specificity of these responses. In the following sections, we review genetic and microbiota-dependent variation in antiviral immunity. In the final sections, we explore less well-studied sources of variation, including abiotic factors, sexual dimorphism, infection history, and endogenous viral elements. We borrow from work on other pathogen types and non-dipteran species when it parallels or complements studies in dipterans. Understanding natural variation in virus-host interactions may lead to the identification of novel restriction factors and immune mechanisms and shed light on the molecular determinants of vector competence.
Collapse
Affiliation(s)
- William H Palmer
- Institute of Evolutionary Biology and Centre for Infection, Evolution and Immunity, University of Edinburgh, Edinburgh EH9 3FL UK.
| | - Finny S Varghese
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| |
Collapse
|
35
|
From Drosophila Blood Cells to Human Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1076:195-214. [PMID: 29951821 DOI: 10.1007/978-981-13-0529-0_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The hematopoietic system plays a critical role in establishing the proper response against invading pathogens or in removing cancerous cells. Furthermore, deregulations of the hematopoietic differentiation program are at the origin of numerous diseases including leukemia. Importantly, many aspects of blood cell development have been conserved from human to Drosophila. Hence, Drosophila has emerged as a potent genetic model to study blood cell development and leukemia in vivo. In this chapter, we give a brief overview of the Drosophila hematopoietic system, and we provide a protocol for the dissection and the immunostaining of the larval lymph gland, the most studied hematopoietic organ in Drosophila. We then focus on the various paradigms that have been used in fly to investigate how conserved genes implicated in leukemogenesis control blood cell development. Specific examples of Drosophila models for leukemia are presented, with particular attention to the most translational ones. Finally, we discuss some limitations and potential improvements of Drosophila models for studying blood cell cancer.
Collapse
|
36
|
Yang H, Hultmark D. Drosophila muscles regulate the immune response against wasp infection via carbohydrate metabolism. Sci Rep 2017; 7:15713. [PMID: 29146985 PMCID: PMC5691183 DOI: 10.1038/s41598-017-15940-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 11/06/2017] [Indexed: 11/09/2022] Open
Abstract
We recently found that JAK/STAT signaling in skeletal muscles is important for the immune response of Drosophila larvae against wasp infection, but it was not clear how muscles could affect the immune response. Here we show that insulin signaling is required in muscles, but not in fat body or hemocytes, during larval development for an efficient encapsulation response and for the formation of lamellocytes. This effect requires TOR signaling. We show that muscle tissue affects the immune response by acting as a master regulator of carbohydrate metabolism in the infected animal, via JAK/STAT and insulin signaling in the muscles, and that there is indirect positive feedback between JAK/STAT and insulin signaling in the muscles. Specifically, stimulation of JAK/STAT signaling in the muscles can rescue the deficient immune response when insulin signaling is suppressed. Our results shed new light on the interaction between metabolism, immunity, and tissue communication.
Collapse
Affiliation(s)
- Hairu Yang
- Department of Molecular Biology, Umeå University, S-901 87, Umeå, Sweden.,Immunology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, 10065, USA
| | - Dan Hultmark
- Department of Molecular Biology, Umeå University, S-901 87, Umeå, Sweden. .,Institute of Biomedical Technology, University of Tampere, FI-33520, Tampere, Finland.
| |
Collapse
|
37
|
Louradour I, Sharma A, Morin-Poulard I, Letourneau M, Vincent A, Crozatier M, Vanzo N. Reactive oxygen species-dependent Toll/NF-κB activation in the Drosophila hematopoietic niche confers resistance to wasp parasitism. eLife 2017; 6:25496. [PMID: 29091025 PMCID: PMC5681226 DOI: 10.7554/elife.25496] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem/progenitor cells in the adult mammalian bone marrow ensure blood cell renewal. Their cellular microenvironment, called 'niche', regulates hematopoiesis both under homeostatic and immune stress conditions. In the Drosophila hematopoietic organ, the lymph gland, the posterior signaling center (PSC) acts as a niche to regulate the hematopoietic response to immune stress such as wasp parasitism. This response relies on the differentiation of lamellocytes, a cryptic cell type, dedicated to pathogen encapsulation and killing. Here, we establish that Toll/NF-κB pathway activation in the PSC in response to wasp parasitism non-cell autonomously induces the lymph gland immune response. Our data further establish a regulatory network where co-activation of Toll/NF-κB and EGFR signaling by ROS levels in the PSC/niche controls lymph gland hematopoiesis under parasitism. Whether a similar regulatory network operates in mammals to control emergency hematopoiesis is an open question.
Collapse
Affiliation(s)
- Isabelle Louradour
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Anurag Sharma
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Ismael Morin-Poulard
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Manon Letourneau
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Alain Vincent
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Vanzo
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
38
|
Proinflammatory Signals as Fuel for the Fire of Hematopoietic Stem Cell Emergence. Trends Cell Biol 2017; 28:58-66. [PMID: 28882414 DOI: 10.1016/j.tcb.2017.08.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cells (HSCs) have the extraordinary ability to both self-renew and generate all mature blood cell lineages. The ability to produce or expand patient-derived HSCs in vitro would greatly improve the outcome for patients with blood disorders that are currently treated with allogeneic HSC transplantation. Many laboratories have been working to identify the signals required for HSC emergence in their native environments to apply this knowledge in vitro. Recently, several signals traditionally known to underlie classical inflammation have emerged as essential regulators of HSC development. In this review we synthesize the findings that have established inflammatory cues as key regulators of HSC development.
Collapse
|
39
|
Khadilkar RJ, Vogl W, Goodwin K, Tanentzapf G. Modulation of occluding junctions alters the hematopoietic niche to trigger immune activation. eLife 2017; 6:28081. [PMID: 28841136 PMCID: PMC5597334 DOI: 10.7554/elife.28081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/24/2017] [Indexed: 12/04/2022] Open
Abstract
Stem cells are regulated by signals from their microenvironment, or niche. During Drosophila hematopoiesis, a niche regulates prohemocytes to control hemocyte production. Immune challenges activate cell-signalling to initiate the cellular and innate immune response. Specifically, certain immune challenges stimulate the niche to produce signals that induce prohemocyte differentiation. However, the mechanisms that promote prohemocyte differentiation subsequent to immune challenges are poorly understood. Here we show that bacterial infection induces the cellular immune response by modulating occluding-junctions at the hematopoietic niche. Occluding-junctions form a permeability barrier that regulates the accessibility of prohemocytes to niche derived signals. The immune response triggered by infection causes barrier breakdown, altering the prohemocyte microenvironment to induce immune cell production. Moreover, genetically induced barrier ablation provides protection against infection by activating the immune response. Our results reveal a novel role for occluding-junctions in regulating niche-hematopoietic progenitor signalling and link this mechanism to immune cell production following infection.
Collapse
Affiliation(s)
- Rohan J Khadilkar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Katharine Goodwin
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
40
|
A Genetic Screen Reveals an Unexpected Role for Yorkie Signaling in JAK/STAT-Dependent Hematopoietic Malignancies in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2017; 7:2427-2438. [PMID: 28620086 PMCID: PMC5555452 DOI: 10.1534/g3.117.044172] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A gain-of-function mutation in the tyrosine kinase JAK2 (JAK2V617F) causes human myeloproliferative neoplasms (MPNs). These patients present with high numbers of myeloid lineage cells and have numerous complications. Since current MPN therapies are not curative, there is a need to find new regulators and targets of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling that may represent additional clinical interventions . Drosophila melanogaster offers a low complexity model to study MPNs as JAK/STAT signaling is simplified with only one JAK [Hopscotch (Hop)] and one STAT (Stat92E). hopTumorous-lethal(Tum-l) is a gain-of-function mutation that causes dramatic expansion of myeloid cells, which then form lethal melanotic tumors. Through an F1 deficiency (Df) screen, we identified 11 suppressors and 35 enhancers of melanotic tumors in hopTum-l animals. Dfs that uncover the Hippo (Hpo) pathway genes expanded (ex) and warts (wts) strongly enhanced the hopTum-l tumor burden, as did mutations in ex, wts, and other Hpo pathway genes. Target genes of the Hpo pathway effector Yorkie (Yki) were significantly upregulated in hopTum-l blood cells, indicating that Yki signaling was increased. Ectopic hematopoietic activation of Yki in otherwise wild-type animals increased hemocyte proliferation but did not induce melanotic tumors. However, hematopoietic depletion of Yki significantly reduced the hopTum-l tumor burden, demonstrating that Yki is required for melanotic tumors in this background. These results support a model in which elevated Yki signaling increases the number of hemocytes, which become melanotic tumors as a result of elevated JAK/STAT signaling.
Collapse
|
41
|
Terriente-Félix A, Pérez L, Bray SJ, Nebreda AR, Milán M. A Drosophila model of myeloproliferative neoplasm reveals a feed-forward loop in the JAK pathway mediated by p38 MAPK signalling. Dis Model Mech 2017; 10:399-407. [PMID: 28237966 PMCID: PMC5399568 DOI: 10.1242/dmm.028118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/01/2017] [Indexed: 12/25/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) of the Philadelphia-negative class comprise polycythaemia vera, essential thrombocythaemia and primary myelofibrosis (PMF). They are associated with aberrant numbers of myeloid lineage cells in the blood, and in the case of overt PMF, with development of myelofibrosis in the bone marrow and failure to produce normal blood cells. These diseases are usually caused by gain-of-function mutations in the kinase JAK2. Here, we use Drosophila to investigate the consequences of activation of the JAK2 orthologue in haematopoiesis. We have identified maturing haemocytes in the lymph gland, the major haematopoietic organ in the fly, as the cell population susceptible to induce hypertrophy upon targeted overexpression of JAK. We show that JAK activates a feed-forward loop, including the cytokine-like ligand Upd3 and its receptor, Domeless, which are required to induce lymph gland hypertrophy. Moreover, we present evidence that p38 MAPK signalling plays a key role in this process by inducing expression of the ligand Upd3. Interestingly, we also show that forced activation of the p38 MAPK pathway in maturing haemocytes suffices to generate hypertrophic organs and the appearance of melanotic tumours. Our results illustrate a novel pro-tumourigenic crosstalk between the p38 MAPK pathway and JAK signalling in a Drosophila model of MPNs. Based on the shared molecular mechanisms underlying MPNs in flies and humans, the interplay between Drosophila JAK and p38 signalling pathways unravelled in this work might have translational relevance for human MPNs. Summary: Pro-tumourigenic crosstalk occurs between the p38 MAPK pathway and JAK signalling in a Drosophila model of myeloproliferative neoplasm.
Collapse
Affiliation(s)
- Ana Terriente-Félix
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Lidia Pérez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Sarah J Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain .,ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain
| | - Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain .,ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain
| |
Collapse
|
42
|
Bingsohn L, Knorr E, Billion A, Narva KE, Vilcinskas A. Knockdown of genes in the Toll pathway reveals new lethal RNA interference targets for insect pest control. INSECT MOLECULAR BIOLOGY 2017; 26:92-102. [PMID: 27862545 DOI: 10.1111/imb.12273] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
RNA interference (RNAi) is a promising alternative strategy for ecologically friendly pest management. However, the identification of RNAi candidate genes is challenging owing to the absence of laboratory strains and the seasonality of most pest species. Tribolium castaneum is a well-established model, with a strong and robust RNAi response, which can be used as a high-throughput screening platform to identify potential RNAi target genes. Recently, the cactus gene was identified as a sensitive RNAi target for pest control. To explore whether the spectrum of promising RNAi targets can be expanded beyond those found by random large-scale screening, to encompass others identified using targeted knowledge-based approaches, we constructed a Cactus interaction network. We tested nine genes in this network and found that the delivery of double-stranded RNA corresponding to fusilli and cactin showed lethal effects. The silencing of cactin resulted in 100% lethality at every developmental stage from the larva to the adult. The knockdown of pelle, Dorsal-related immunity factor and short gastrulation reduced or even prevented egg hatching in the next generation. The combination of such targets with lethal and parental RNAi effects can now be tested against different pest species in field studies.
Collapse
Affiliation(s)
- L Bingsohn
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - E Knorr
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - A Billion
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
| | - K E Narva
- Dow AgroSciences, Indianapolis, IN, USA
| | - A Vilcinskas
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology, Giessen, Germany
- Institute for Insect Biotechnology, Giessen, Germany
| |
Collapse
|
43
|
El Chamy L, Matt N, Reichhart JM. Advances in Myeloid-Like Cell Origins and Functions in the Model Organism Drosophila melanogaster. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0038-2016. [PMID: 28102122 PMCID: PMC11687447 DOI: 10.1128/microbiolspec.mchd-0038-2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Indexed: 12/22/2022] Open
Abstract
Drosophila has long served as a valuable model for deciphering many biological processes, including immune responses. Indeed, the genetic tractability of this organism is particularly suited for large-scale analyses. Studies performed during the last 3 decades have proven that the signaling pathways that regulate the innate immune response are conserved between Drosophila and mammals. This review summarizes the recent advances on Drosophila hematopoiesis and immune cellular responses, with a particular emphasis on phagocytosis.
Collapse
Affiliation(s)
- Laure El Chamy
- Laboratoire de Génétique de la drosophile et virulence microbienne, UR. EGFEM, Faculté des Sciences, Université Saint-Joseph de Beyrouth, B.P. 17-5208 Mar Mikhaël Beyrouth 1104 2020, Liban
| | - Nicolas Matt
- Université de Strasbourg, UPR 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg Cedex 67084, France
| | - Jean-Marc Reichhart
- Université de Strasbourg, UPR 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg Cedex 67084, France
| |
Collapse
|
44
|
Zhang G, Hao Y, Jin LH. Overexpression of jumu induces melanotic nodules by activating Toll signaling in Drosophila. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 77:31-38. [PMID: 27507244 DOI: 10.1016/j.ibmb.2016.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/03/2016] [Accepted: 08/03/2016] [Indexed: 06/06/2023]
Abstract
Melanotic nodules are commonly assumed to be caused by an abnormal immune response. Several hematopoietic mutants and signaling pathways, including the Toll, JAK/STAT, Ras and JNK pathways, can cause melanotic nodules to develop when specifically activated in hemocytes. Here, we used the UAS-Gal4 system to overexpress jumeaux (jumu) in the fly immune response system. Jumeaux (Jumu) is a new member of the winged-helix/forkhead (WH/FKH) gene family of transcription factors, which plays an important role in the growth and morphogenesis of Drosophila and participates in the proliferation and differentiation of hemocytes. Overexpressing jumu in both hemocytes and the fat body generated many melanotic nodules in larvae and adult flies. The nodules observed in the fat body were surrounded by large numbers of blood cells through a process that appeared similar to foreign body encapsulation. This phenomenon is caused by Toll pathway activation and leads to blood cells deposited in the fat body. In addition, we also report the dissociation of fat cells and the abnormal proliferation and differentiation of blood cells. These results suggest a Jumu-mediated crosstalk between hematopoiesis and the fat body, especially during the Toll-dependent formation of melanotic nodules.
Collapse
Affiliation(s)
- Gaoqun Zhang
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Yangguang Hao
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China.
| |
Collapse
|
45
|
Papanicolaou A, Schetelig MF, Arensburger P, Atkinson PW, Benoit JB, Bourtzis K, Castañera P, Cavanaugh JP, Chao H, Childers C, Curril I, Dinh H, Doddapaneni H, Dolan A, Dugan S, Friedrich M, Gasperi G, Geib S, Georgakilas G, Gibbs RA, Giers SD, Gomulski LM, González-Guzmán M, Guillem-Amat A, Han Y, Hatzigeorgiou AG, Hernández-Crespo P, Hughes DST, Jones JW, Karagkouni D, Koskinioti P, Lee SL, Malacrida AR, Manni M, Mathiopoulos K, Meccariello A, Munoz-Torres M, Murali SC, Murphy TD, Muzny DM, Oberhofer G, Ortego F, Paraskevopoulou MD, Poelchau M, Qu J, Reczko M, Robertson HM, Rosendale AJ, Rosselot AE, Saccone G, Salvemini M, Savini G, Schreiner P, Scolari F, Siciliano P, Sim SB, Tsiamis G, Ureña E, Vlachos IS, Werren JH, Wimmer EA, Worley KC, Zacharopoulou A, Richards S, Handler AM. The whole genome sequence of the Mediterranean fruit fly, Ceratitis capitata (Wiedemann), reveals insights into the biology and adaptive evolution of a highly invasive pest species. Genome Biol 2016; 17:192. [PMID: 27659211 PMCID: PMC5034548 DOI: 10.1186/s13059-016-1049-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/26/2016] [Indexed: 01/01/2023] Open
Abstract
Background The Mediterranean fruit fly (medfly), Ceratitis capitata, is a major destructive insect pest due to its broad host range, which includes hundreds of fruits and vegetables. It exhibits a unique ability to invade and adapt to ecological niches throughout tropical and subtropical regions of the world, though medfly infestations have been prevented and controlled by the sterile insect technique (SIT) as part of integrated pest management programs (IPMs). The genetic analysis and manipulation of medfly has been subject to intensive study in an effort to improve SIT efficacy and other aspects of IPM control. Results The 479 Mb medfly genome is sequenced from adult flies from lines inbred for 20 generations. A high-quality assembly is achieved having a contig N50 of 45.7 kb and scaffold N50 of 4.06 Mb. In-depth curation of more than 1800 messenger RNAs shows specific gene expansions that can be related to invasiveness and host adaptation, including gene families for chemoreception, toxin and insecticide metabolism, cuticle proteins, opsins, and aquaporins. We identify genes relevant to IPM control, including those required to improve SIT. Conclusions The medfly genome sequence provides critical insights into the biology of one of the most serious and widespread agricultural pests. This knowledge should significantly advance the means of controlling the size and invasive potential of medfly populations. Its close relationship to Drosophila, and other insect species important to agriculture and human health, will further comparative functional and structural studies of insect genomes that should broaden our understanding of gene family evolution. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1049-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexie Papanicolaou
- Hawkesbury Institute for the Environment, Western Sydney University, Sydney, Australia
| | - Marc F Schetelig
- Justus-Liebig-University Giessen, Institute for Insect Biotechnology, 35394, Giessen, Germany
| | - Peter Arensburger
- Department of Biological Sciences, Cal Poly Pomona, Pomona, CA, 91768, USA
| | - Peter W Atkinson
- Department of Entomology and Center for Disease Vector Research, University of California Riverside, Riverside, CA, 92521, USA.,Interdepartmental Graduate Program in Genetics, Genomics & Bioinformatics, University of California Riverside, Riverside, CA, 92521, USA
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Kostas Bourtzis
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, Seibersdorf, Vienna, Austria.,Department of Environmental and Natural Resources Management, University of Patras, Agrinio, Greece
| | - Pedro Castañera
- Department of Environmental Biology, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - John P Cavanaugh
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Hsu Chao
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Ingrid Curril
- Georg-August-Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, 37077, Göttingen, Germany
| | - Huyen Dinh
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - HarshaVardhan Doddapaneni
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Amanda Dolan
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Shannon Dugan
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Giuliano Gasperi
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Scott Geib
- USDA-ARS, Pacific Basin Agricultural Research Center, Hilo, HI, 96720, USA
| | - Georgios Georgakilas
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, 382 21 Volos, Greece and Hellenic Pasteur Institute, 11521, Athens, Greece
| | - Richard A Gibbs
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sarah D Giers
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ludvik M Gomulski
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Miguel González-Guzmán
- Department of Environmental Biology, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Ana Guillem-Amat
- Department of Environmental Biology, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Yi Han
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Artemis G Hatzigeorgiou
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, 382 21 Volos, Greece and Hellenic Pasteur Institute, 11521, Athens, Greece
| | - Pedro Hernández-Crespo
- Department of Environmental Biology, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Daniel S T Hughes
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeffery W Jones
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Dimitra Karagkouni
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, 382 21 Volos, Greece and Hellenic Pasteur Institute, 11521, Athens, Greece
| | - Panagiota Koskinioti
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Sandra L Lee
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna R Malacrida
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Mosè Manni
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Kostas Mathiopoulos
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Angela Meccariello
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | | | - Shwetha C Murali
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Terence D Murphy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Georg Oberhofer
- Georg-August-Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, 37077, Göttingen, Germany
| | - Félix Ortego
- Department of Environmental Biology, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Maria D Paraskevopoulou
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, 382 21 Volos, Greece and Hellenic Pasteur Institute, 11521, Athens, Greece
| | - Monica Poelchau
- National Agricultural Library, USDA, Beltsville, MD, 20705, USA
| | - Jiaxin Qu
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Martin Reczko
- Institute of Molecular Biology and Genetics, Biomedical Sciences Research Centre "Alexander Fleming", Vari, Greece
| | - Hugh M Robertson
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Andrew J Rosendale
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Andrew E Rosselot
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Giuseppe Saccone
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Marco Salvemini
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Grazia Savini
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Patrick Schreiner
- Interdepartmental Graduate Program in Genetics, Genomics & Bioinformatics, University of California Riverside, Riverside, CA, 92521, USA
| | - Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Paolo Siciliano
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Sheina B Sim
- USDA-ARS, Pacific Basin Agricultural Research Center, Hilo, HI, 96720, USA
| | - George Tsiamis
- Department of Environmental and Natural Resources Management, University of Patras, Agrinio, Greece
| | - Enric Ureña
- Department of Environmental Biology, Centro de Investigaciones Biológicas, CSIC, 28040, Madrid, Spain
| | - Ioannis S Vlachos
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, 382 21 Volos, Greece and Hellenic Pasteur Institute, 11521, Athens, Greece
| | - John H Werren
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Ernst A Wimmer
- Georg-August-Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, 37077, Göttingen, Germany
| | - Kim C Worley
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Stephen Richards
- Human Genome Sequencing Center, Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alfred M Handler
- USDA-ARS, Center for Medical, Agricultural, and Veterinary Entomology, 1700 S.W. 23rd Drive, Gainesville, FL, 32608, USA.
| |
Collapse
|
46
|
Smith RC, Barillas-Mury C. Plasmodium Oocysts: Overlooked Targets of Mosquito Immunity. Trends Parasitol 2016; 32:979-990. [PMID: 27639778 DOI: 10.1016/j.pt.2016.08.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/26/2016] [Accepted: 08/30/2016] [Indexed: 12/18/2022]
Abstract
Although the ability of mosquitoes to limit Plasmodium infection is well documented, many questions remain as to how malaria parasites are recognized and killed by the mosquito host. Recent evidence suggests that anti-Plasmodium immunity is multimodal, with different immune mechanisms regulating ookinete and oocyst survival. However, most experiments determine the number of mature oocysts, without considering that different immune mechanisms may target different developmental stages of the parasite. Complement-like proteins have emerged as important determinants of early immunity targeting the ookinete stage, yet the mechanisms by which the mosquito late-phase immune response limits oocyst survival are less understood. Here, we describe the known components of the mosquito immune system that limit oocyst development, and provide insight into their possible mechanisms of action.
Collapse
Affiliation(s)
- Ryan C Smith
- Department of Entomology, Iowa State University, Ames, IA, USA.
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
47
|
Letourneau M, Lapraz F, Sharma A, Vanzo N, Waltzer L, Crozatier M. Drosophila hematopoiesis under normal conditions and in response to immune stress. FEBS Lett 2016; 590:4034-4051. [PMID: 27455465 DOI: 10.1002/1873-3468.12327] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/07/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
The emergence of hematopoietic progenitors and their differentiation into various highly specialized blood cell types constitute a finely tuned process. Unveiling the genetic cascades that control blood cell progenitor fate and understanding how they are modulated in response to environmental changes are two major challenges in the field of hematopoiesis. In the last 20 years, many studies have established important functional analogies between blood cell development in vertebrates and in the fruit fly, Drosophila melanogaster. Thereby, Drosophila has emerged as a powerful genetic model for studying mechanisms that control hematopoiesis during normal development or in pathological situations. Moreover, recent advances in Drosophila have highlighted how intricate cell communication networks and microenvironmental cues regulate blood cell homeostasis. They have also revealed the striking plasticity of Drosophila mature blood cells and the presence of different sites of hematopoiesis in the larva. This review provides an overview of Drosophila hematopoiesis during development and summarizes our current knowledge on the molecular processes controlling larval hematopoiesis, both under normal conditions and in response to an immune challenge, such as wasp parasitism.
Collapse
Affiliation(s)
- Manon Letourneau
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Francois Lapraz
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Anurag Sharma
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France.,Department of Biomedical Sciences, NU Centre for Science Education & Research, Nitte University, Mangalore-18, India
| | - Nathalie Vanzo
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Lucas Waltzer
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| |
Collapse
|
48
|
Schmid MR, Anderl I, Vo HTM, Valanne S, Yang H, Kronhamn J, Rämet M, Rusten TE, Hultmark D. Genetic Screen in Drosophila Larvae Links ird1 Function to Toll Signaling in the Fat Body and Hemocyte Motility. PLoS One 2016; 11:e0159473. [PMID: 27467079 PMCID: PMC4965076 DOI: 10.1371/journal.pone.0159473] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/05/2016] [Indexed: 12/26/2022] Open
Abstract
To understand how Toll signaling controls the activation of a cellular immune response in Drosophila blood cells (hemocytes), we carried out a genetic modifier screen, looking for deletions that suppress or enhance the mobilization of sessile hemocytes by the gain-of-function mutation Toll10b (Tl10b). Here we describe the results from chromosome arm 3R, where five regions strongly suppressed this phenotype. We identified the specific genes immune response deficient 1 (ird1), headcase (hdc) and possibly Rab23 as suppressors, and we studied the role of ird1 in more detail. An ird1 null mutant and a mutant that truncates the N-terminal kinase domain of the encoded Ird1 protein affected the Tl10b phenotype, unlike mutations that affect the C-terminal part of the protein. The ird1 null mutant suppressed mobilization of sessile hemocytes, but enhanced other Tl10b hemocyte phenotypes, like the formation of melanotic nodules and the increased number of circulating hemocytes. ird1 mutants also had blood cell phenotypes on their own. They lacked crystal cells and showed aberrant formation of lamellocytes. ird1 mutant plasmatocytes had a reduced ability to spread on an artificial substrate by forming protrusions, which may explain why they did not go into circulation in response to Toll signaling. The effect of the ird1 mutation depended mainly on ird1 expression in hemocytes, but ird1-dependent effects in other tissues may contribute. Specifically, the Toll receptor was translocated from the cell membrane to intracellular vesicles in the fat body of the ird1 mutant, and Toll signaling was activated in that tissue, partially explaining the Tl10b-like phenotype. As ird1 is otherwise known to control vesicular transport, we conclude that the vesicular transport system may be of particular importance during an immune response.
Collapse
Affiliation(s)
| | - Ines Anderl
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- BioMediTech, University of Tampere, Tampere, Finland
| | - Hoa T. M. Vo
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | - Hairu Yang
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jesper Kronhamn
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Mika Rämet
- BioMediTech, University of Tampere, Tampere, Finland
- PEDEGO Research Center, and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tor Erik Rusten
- Department of Molecular Cell Biology, Oslo University Hospital, Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Dan Hultmark
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- BioMediTech, University of Tampere, Tampere, Finland
| |
Collapse
|
49
|
The Friend of GATA Transcriptional Co-Regulator, U-Shaped, Is a Downstream Antagonist of Dorsal-Driven Prohemocyte Differentiation in Drosophila. PLoS One 2016; 11:e0155372. [PMID: 27163255 PMCID: PMC4862636 DOI: 10.1371/journal.pone.0155372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 04/27/2016] [Indexed: 12/11/2022] Open
Abstract
Recent studies suggest that mammalian hematopoietic stem and progenitor cells (HSPCs) respond directly to infection and inflammatory signaling. These signaling pathways also regulate HSPCs during steady-state conditions (absence of infection), and dysregulation may lead to cancer or age-related loss of progenitor repopulation capacity. Toll-like receptors (TLRs) are a major class of pathogen recognition receptors, and are expressed on the surface of immune effector cells and HSPCs. TLR/NF-κB activation promotes HSPCs differentiation; however, the mechanisms by which this signaling pathway alters the intrinsic transcriptional landscape are not well understood. Although Drosophila prohemocytes are the functional equivalent of mammalian HSPCs, a prohemocyte-specific function for Toll signaling has not been reported. Using Drosophila transgenics, we identified prohemocyte-specific roles for Toll pathway members, Dorsal and Cactus. We showed that Dorsal is required to limit the size of the progenitor pool. Additionally, we showed that activation of Toll signaling in prohemocytes drives differentiation in a manner that is analogous to TLR/NF-κB-driven HSPC differentiation. This was accomplished by showing that over-expression of Dorsal, or knockdown of Cactus, promotes differentiation. We also investigated whether Dorsal and Cactus control prohemocyte differentiation by regulating a key intrinsic prohemocyte factor, U-shaped (Ush), which is known to promote multipotency and block differentiation. We showed that Dorsal repressed Ush expression levels to promote differentiation, whereas Cactus maintained Ush levels to block differentiation. Additionally, we showed that another Toll antagonist, Lesswright, also maintained the level of Ush to block differentiation and promote proliferative quiescence. Collectively, these results identify a novel role for Ush as a downstream target of Toll signaling.
Collapse
|
50
|
Espín-Palazón R, Traver D. The NF-κB family: Key players during embryonic development and HSC emergence. Exp Hematol 2016; 44:519-27. [PMID: 27132652 DOI: 10.1016/j.exphem.2016.03.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 03/24/2016] [Accepted: 03/26/2016] [Indexed: 02/07/2023]
Abstract
The nuclear factor-κB (NF-κB) family is a crucial transcription factor group known mainly for its role in the regulation of the immune system and its response to infection in vertebrates. The signaling pathway leading to NF-κB activation and translocation to the nucleus to exert its function as a transcription factor is well conserved among Kingdom Animalia, which has helped to elucidate other roles that NF-κB plays in other biological contexts such as developmental biology. The manipulation of NF-κB members in a diverse range of animal models results in severe developmental defects during embryogenesis, very often leading to embryonic lethality. Defects include dorsal-ventral patterning and limb, liver, skin, lung, neural, notochord, muscle, skeletal, and hematopoietic defects. Here, we recapitulate the research that has been done to address the role that NF-κB plays during embryonic development, in particular to emphasize its recently discovered role in the specification of hematopoietic stem cells (HSCs), the foundation of the hematopoietic system in vertebrates.
Collapse
Affiliation(s)
- Raquel Espín-Palazón
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA.
| |
Collapse
|