1
|
Aalto AL, Luukkonen V, Meinander A. Ubiquitin signalling in Drosophila innate immune responses. FEBS J 2024; 291:4397-4413. [PMID: 38069549 DOI: 10.1111/febs.17028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Cells respond to invading pathogens and danger signals from the environment by adapting gene expression to meet the need for protective effector molecules. While this innate immune response is required for the cell and the organism to recover, excess immune activation may lead to loss of homeostasis, thereby promoting chronic inflammation and cancer progression. The molecular basis of innate immune defence is comprised of factors promoting survival and proliferation, such as cytokines, antimicrobial peptides and anti-apoptotic proteins. As the molecular mechanisms regulating innate immune responses are conserved through evolution, the fruit fly Drosophila melanogaster serves as a convenient, affordable and ethical model organism to enhance understanding of immune signalling. Fly immunity against bacterial infection is built up by both cellular and humoral responses, where the latter is regulated by the Imd and Toll pathways activating NF-κB transcription factors Relish, Dorsal and Dif, as well as JNK activation and JAK/STAT signalling. As in mammals, the Drosophila innate immune signalling pathways are characterised by ubiquitination of signalling molecules followed by ubiquitin receptors binding to the ubiquitin chains, as well as by rapid changes in protein levels by ubiquitin-mediated targeted proteasomal and lysosomal degradation. In this review, we summarise the molecular signalling pathways regulating immune responses to pathogen infection in Drosophila, with a focus on ubiquitin-dependent control of innate immunity and inflammatory signalling.
Collapse
Affiliation(s)
- Anna L Aalto
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Veera Luukkonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| |
Collapse
|
2
|
Brantley SE, Stouthamer CM, Kr P, Fischer ML, Hill J, Schlenke TA, Mortimer NT. Host JAK-STAT activity is a target of parasitoid wasp virulence strategies. PLoS Pathog 2024; 20:e1012349. [PMID: 38950076 PMCID: PMC11244843 DOI: 10.1371/journal.ppat.1012349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/12/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
Innate immune responses that allow hosts to survive infection depend on the action of multiple conserved signaling pathways. Pathogens and parasites in turn have evolved virulence factors to target these immune signaling pathways in an attempt to overcome host immunity. Consequently, the interactions between host immune molecules and pathogen virulence factors play an important role in determining the outcome of an infection. The immune responses of Drosophila melanogaster provide a valuable model to understand immune signaling and host-pathogen interactions. Flies are commonly infected by parasitoid wasps and mount a coordinated cellular immune response following infection. This response is characterized by the production of specialized blood cells called lamellocytes that form a tight capsule around wasp eggs in the host hemocoel. The conserved JAK-STAT signaling pathway has been implicated in lamellocyte proliferation and is required for successful encapsulation of wasp eggs. Here we show that activity of Stat92E, the D. melanogaster STAT ortholog, is induced in immune tissues following parasitoid infection. Virulent wasp species are able to suppress Stat92E activity during infection, suggesting they target JAK-STAT pathway activation as a virulence strategy. Furthermore, two wasp species (Leptopilina guineaensis and Ganaspis xanthopoda) suppress phenotypes associated with a gain-of-function mutation in hopscotch, the D. melanogaster JAK ortholog, indicating that they inhibit the activity of the core signaling components of the JAK-STAT pathway. Our data suggest that parasitoid wasp virulence factors block JAK-STAT signaling to overcome fly immune defenses.
Collapse
Affiliation(s)
- Susanna E Brantley
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Corinne M Stouthamer
- Department of Entomology, University of Arizona, Tucson, Arizona, United States of America
| | - Pooja Kr
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Mary L Fischer
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Joshua Hill
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Todd A Schlenke
- Department of Entomology, University of Arizona, Tucson, Arizona, United States of America
| | - Nathan T Mortimer
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
3
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
4
|
Laureti M, Lee RX, Bennett A, Wilson LA, Sy VE, Kohl A, Dietrich I. Rift Valley Fever Virus Primes Immune Responses in Aedes aegypti Cells. Pathogens 2023; 12:563. [PMID: 37111448 PMCID: PMC10146816 DOI: 10.3390/pathogens12040563] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/25/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
The ongoing global emergence of arthropod-borne (arbo) viruses has accelerated research into the interactions of these viruses with the immune systems of their vectors. Only limited information exists on how bunyaviruses, such as Rift Valley fever virus (RVFV), are sensed by mosquito immunity or escape detection. RVFV is a zoonotic phlebovirus (Bunyavirales; Phenuiviridae) of veterinary and human public health and economic importance. We have shown that the infection of mosquitoes with RVFV triggers the activation of RNA interference pathways, which moderately restrict viral replication. Here, we aimed to better understand the interactions between RVFV and other vector immune signaling pathways that might influence RVFV replication and transmission. For this, we used the immunocompetent Aedes aegypti Aag2 cell line as a model. We found that bacteria-induced immune responses restricted RVFV replication. However, virus infection alone did not alter the gene expression levels of immune effectors. Instead, it resulted in the marked enhancement of immune responses to subsequent bacterial stimulation. The gene expression levels of several mosquito immune pattern recognition receptors were altered by RVFV infection, which may contribute to this immune priming. Our findings imply that there is a complex interplay between RVFV and mosquito immunity that could be targeted in disease prevention strategies.
Collapse
Affiliation(s)
| | - Rui-Xue Lee
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Amelia Bennett
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
- Department of Life Sciences, Faculty of Science, Claverton Down, University of Bath, Bath BA2 7AY, UK
| | - Lucas Aladar Wilson
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | | | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Isabelle Dietrich
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| |
Collapse
|
5
|
Wang Y, Zhou L, Liang W, Dang Z, Wang S, Zhang Y, Zhao P, Lu Z. Cytokine receptor DOME controls wing disc development in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 148:103828. [PMID: 36002096 DOI: 10.1016/j.ibmb.2022.103828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
In multicellular organisms, the JAK/STAT signaling pathway is involved in cell proliferation, differentiation, apoptosis, and immune regulation. Through activation of the Stat92E transcription factor, JAK/STAT signaling induced proper wing development in Drosophila. Domeless (DOME) was the first identified invertebrate JAK/STAT receptor. However, the function of DOME in Bombyx mori development remains unclear, especially in wing morphogenesis. In this study, we isolated the cytokine receptor DOME gene in B. mori and evaluated its function in DOME-knockout models. We found that overexpression of DOME at the cellular level upregulated the expression of JAK/STAT pathway-related genes, promoted proliferation, and inhibited apoptosis. The results of the interference with DOME had the opposite effects with those of overexpression at the cellular level. Using CRISPR/Cas9 technology, we constructed a DOME-knockout transgenic silkworm strain (KO-DOME) and found that the wings of the pupa and moth stages were vesicle-shaped and smaller than those of the wild-type silkworm. Some KO-DOME silkworms were unable to extend their wings from the pupal case after eclosion. We detected the expression of cyclin and apoptosis-related genes in the wing disc of the moth stage and found that some cyclin genes, such as CyclinA, CyclinB, and CyclinD, were downregulated, whereas apoptotic genes, such as Caspase1, Caspase3, and Caspase8, were upregulated. We propose that DOME regulates cell proliferation and apoptosis by affecting the JAK/STAT signaling pathway, ultimately influencing the development of wing discs. Our study provides empirical evidence for the biological function of the silkworm DOME gene, which is essential for the normal development of wings.
Collapse
Affiliation(s)
- Yaping Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China.
| | - Li Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Wenjuan Liang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Zhuo Dang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Shiyuan Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Yan Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715, PR China
| | - Ping Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715, PR China
| | - Zhongyan Lu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
6
|
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: 45] [Impact Index Per Article: 15.0] [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
|
7
|
Neophytou C, Pitsouli C. Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep 2022; 38:110505. [PMID: 35263602 DOI: 10.1016/j.celrep.2022.110505] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/11/2021] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Diet is a key regulator of metabolism and interacts with the intestinal microbiome. Here, we study the role of the Drosophila intestinal stem cell (ISC)-specific biotin transporter Smvt in midgut homeostasis, infection-induced regeneration, and tumorigenesis. We show that Smvt-transported biotin in ISCs is necessary for ISC mitosis. Smvt deficiency impairs intestinal maintenance, which can be rescued by the human Smvt, encoded by SLC5A6. ISC-specific, Smvt-silenced flies exhibit microbial dysbiosis, whereby the growth of Providencia sneebia, an opportunistic pathogen, is favored. Dysbiosis correlates with increased Nox expression, reactive oxygen species (ROS), and enterocyte apoptosis. Flies acquire biotin from their diet and microbiota. We show that, when dietary biotin is scarce, biotin-producing commensals, e.g., E. coli, can rescue reduced ISC mitosis. Smvt and commensals also control intestinal tumor growth. Our findings suggest that direct modification of the gut microbiome by biotin can serve as an approach for the treatment of dysbiosis-promoted diseases and tumorigenesis control.
Collapse
Affiliation(s)
- Constantina Neophytou
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus.
| |
Collapse
|
8
|
Cao X, Rojas M, Pastor-Pareja JC. Intrinsic and damage-induced JAK/STAT signaling regulate developmental timing by the Drosophila prothoracic gland. Dis Model Mech 2021; 15:273570. [PMID: 34842272 PMCID: PMC8807578 DOI: 10.1242/dmm.049160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
Abstract
Development involves tightly paced, reproducible sequences of events, yet it must adjust to conditions external to it, such as resource availability and organismal damage. A major mediator of damage-induced immune responses in vertebrates and insects is JAK/STAT signaling. At the same time, JAK/STAT activation by the Drosophila Upd cytokines is pleiotropically involved in normal development of multiple organs. Whether inflammatory and developmental JAK/STAT roles intersect is unknown. Here, we show that JAK/STAT is active during development of the prothoracic gland (PG), which controls metamorphosis onset through ecdysone production. Reducing JAK/STAT signaling decreased PG size and advanced metamorphosis. Conversely, JAK/STAT hyperactivation by overexpression of pathway components or SUMOylation loss caused PG hypertrophy and metamorphosis delay. Tissue damage and tumors, known to secrete Upd cytokines, also activated JAK/STAT in the PG and delayed metamorphosis, at least in part by inducing expression of the JAK/STAT target Apontic. JAK/STAT damage signaling, therefore, regulates metamorphosis onset by co-opting its developmental role in the PG. Our findings in Drosophila provide insights on how systemic effects of damage and cancer can interfere with hormonally controlled development and developmental transitions. Summary: Damage signaling from tumors mediated by JAK/STAT-activating Upd cytokines delays the Drosophila larva–pupa transition through co-option of a JAK/STAT developmental role in the prothoracic gland.
Collapse
Affiliation(s)
- Xueya Cao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Marta Rojas
- School of Medicine, Tsinghua University, Beijing, China
| | - José Carlos Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
9
|
Scholl A, Ndoja I, Jiang L. Drosophila Trachea as a Novel Model of COPD. Int J Mol Sci 2021; 22:ijms222312730. [PMID: 34884534 PMCID: PMC8658011 DOI: 10.3390/ijms222312730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
COPD, a chronic obstructive pulmonary disease, is one of the leading causes of death worldwide. Clinical studies and research in rodent models demonstrated that failure of repair mechanisms to cope with increased ROS and inflammation in the lung leads to COPD. Despite this progress, the molecular mechanisms underlying the development of COPD remain poorly understood, resulting in a lack of effective treatments. Thus, an informative, simple model is highly valued and desired. Recently, the cigarette smoke-induced Drosophila COPD model showed a complex set of pathological phenotypes that resemble those seen in human COPD patients. The Drosophila trachea has been used as a premier model to reveal the mechanisms of tube morphogenesis. The association of these mechanisms to structural changes in COPD can be analyzed by using Drosophila trachea. Additionally, the timeline of structural damage, ROS, and inflammation can be studied in live organisms using fluorescently-tagged proteins. The related function of human COPD genes identified by GWAS can be screened using respective fly homologs. Finally, the Drosophila trachea can be used as a high-throughput drug screening platform to identify novel treatments for COPD. Therefore, Drosophila trachea is an excellent model that is complementary to rodent COPD models.
Collapse
|
10
|
Rosendo Machado S, van der Most T, Miesen P. Genetic determinants of antiviral immunity in dipteran insects - Compiling the experimental evidence. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104010. [PMID: 33476667 DOI: 10.1016/j.dci.2021.104010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 06/12/2023]
Abstract
The genetic basis of antiviral immunity in dipteran insects is extensively studied in Drosophila melanogaster and advanced technologies for genetic manipulation allow a better characterization of immune responses also in non-model insect species. Especially, immunity in vector mosquitoes is recently in the spotlight, due to the medical impact that these insects have by transmitting viruses and other pathogens. Here, we review the current state of experimental evidence that supports antiviral functions for immune genes acting in different cellular pathways. We discuss the well-characterized RNA interference mechanism along with the less well-defined JAK-STAT, Toll, and IMD signaling pathways. Furthermore, we highlight the initial evidence for antiviral activity observed for the autophagy pathway, transcriptional pausing, as well as piRNA production from endogenous viral elements. We focus our review on studies from Drosophila and mosquito species from the lineages Aedes, Culex, and Anopheles, which contain major vector species responsible for virus transmission.
Collapse
Affiliation(s)
- Samara Rosendo Machado
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Tom van der Most
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands.
| |
Collapse
|
11
|
Wang F. Sending Out Alarms: A Perspective on Intercellular Communications in Insect Antiviral Immune Response. Front Immunol 2021; 12:613729. [PMID: 33708207 PMCID: PMC7940532 DOI: 10.3389/fimmu.2021.613729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/12/2021] [Indexed: 11/13/2022] Open
Abstract
Viral infection triggers insect immune response, including RNA interference, apoptosis and autophagy, and profoundly changes the gene expression profiles in infected cells. Although intracellular degradation is crucial for restricting viral infection, intercellular communication is required to mount a robust systemic immune response. This review focuses on recent advances in understanding the intercellular communications in insect antiviral immunity, including protein-based and virus-derived RNA based cell-cell communications, with emphasis on the signaling pathway that induces the production of the potential cytokines. The prospects and challenges of future work are also discussed.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China.,Biological Science Research Center, Southwest University, Chongqing, China
| |
Collapse
|
12
|
Benoist L, Corre E, Bernay B, Henry J, Zatylny-Gaudin C. -Omic Analysis of the Sepia officinalis White Body: New Insights into Multifunctionality and Haematopoiesis Regulation. J Proteome Res 2020; 19:3072-3087. [PMID: 32643382 DOI: 10.1021/acs.jproteome.0c00100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cephalopods, like other protostomes, lack an adaptive immune system and only rely on an innate immune system. The main immune cells are haemocytes (Hcts), which are able to respond to pathogens and external attacks. First reports based on morphological observations revealed that the white body (WB) located in the optic sinuses of cuttlefish was the origin of Hcts. Combining transcriptomic and proteomic analyses, we identified several factors known to be involved in haematopoiesis in vertebrate species in cuttlefish WB. Among these factors, members of the JAK-STAT signaling pathway were identified, some of them for the first time in a molluscan transcriptome and proteome. Immune factors, such as members of the Toll/NF-κB signaling pathway, pattern recognition proteins and receptors, and members of the oxidative stress responses, were also identified, and support an immune role of the WB. Both transcriptome and proteome analyses revealed that the WB harbors an intense metabolism concurrent with the haematopoietic function. Finally, a comparative analysis of the WB and Hct proteomes revealed many proteins in common, confirming previous morphological studies on the origin of Hcts in cuttlefish. This molecular work demonstrates that the WB is multifunctional and provides bases for haematopoiesis regulation in cuttlefish.
Collapse
Affiliation(s)
- Louis Benoist
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France.,Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France
| | - Erwan Corre
- Plateforme ABiMS, Station Biologique de Roscoff (CNRS-Sorbonne Université), 29688 Roscoff, France
| | - Benoit Bernay
- Plateforme PROTEOGEN, SF 4206 ICORE, Normandie université, Esplanade de la Paix, 14032 Caen Cedex, France
| | - Joel Henry
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France.,Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France
| | - Céline Zatylny-Gaudin
- NORMANDIE UNIV, UNICAEN, CNRS, BOREA, 14000 Caen, France.,Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen-Normandie, MNHN, SU, UA, CNRS, IRD, Esplanade de la paix, 14032 Caen Cedex, France
| |
Collapse
|
13
|
Sreejith P, Jang W, To V, Hun Jo Y, Biteau B, Kim C. Lin28 is a critical factor in the function and aging of Drosophila testis stem cell niche. Aging (Albany NY) 2020; 11:855-873. [PMID: 30713156 PMCID: PMC6382437 DOI: 10.18632/aging.101765] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/05/2019] [Indexed: 12/21/2022]
Abstract
Age-related decline in stem cell function is observed in many tissues from invertebrates to humans. While cell intrinsic alterations impair stem cells, aging of the stem cell niche also significantly contributes to the loss of tissue homeostasis associated with reduced regenerative capacity. Hub cells, which constitute the stem cell niche in the Drosophila testis, exhibit age-associated decline in number and activities, yet underlying mechanisms are not fully understood. Here we show that Lin28, a highly conserved RNA binding protein, is expressed in hub cells and its expression dramatically declines in old testis. lin28 mutant testes exhibit hub cell loss and defective hub architecture, recapitulating the normal aging process. Importantly, maintained expression of Lin28 prolongs hub integrity and function in aged testes, suggesting that Lin28 decline is a driver of hub cell aging. Mechanistically, the level of unpaired (upd), a stem cell self-renewal factor, is reduced in lin28 mutant testis and Lin28 protein directly binds and stabilizes upd transcripts, in a let-7 independent manner. Altogether, our results suggest that Lin28 acts to protect upd transcripts in hub cells, and reduction of Lin28 in old testis leads to decreased upd levels, hub cell aging and loss of the stem cell niche.
Collapse
Affiliation(s)
- Perinthottathil Sreejith
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Wijeong Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Van To
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong Hun Jo
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 500-757, Republic of Korea
| | - Benoit Biteau
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Changsoo Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
14
|
Kierdorf K, Hersperger F, Sharrock J, Vincent CM, Ustaoglu P, Dou J, Gyoergy A, Groß O, Siekhaus DE, Dionne MS. Muscle function and homeostasis require cytokine inhibition of AKT activity in Drosophila. eLife 2020; 9:e51595. [PMID: 31944178 PMCID: PMC6996930 DOI: 10.7554/elife.51595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/10/2020] [Indexed: 12/20/2022] Open
Abstract
Unpaired ligands are secreted signals that act via a GP130-like receptor, domeless, to activate JAK/STAT signalling in Drosophila. Like many mammalian cytokines, unpaireds can be activated by infection and other stresses and can promote insulin resistance in target tissues. However, the importance of this effect in non-inflammatory physiology is unknown. Here, we identify a requirement for unpaired-JAK signalling as a metabolic regulator in healthy adult Drosophila muscle. Adult muscles show basal JAK-STAT signalling activity in the absence of any immune challenge. Plasmatocytes (Drosophila macrophages) are an important source of this tonic signal. Loss of the dome receptor on adult muscles significantly reduces lifespan and causes local and systemic metabolic pathology. These pathologies result from hyperactivation of AKT and consequent deregulation of metabolism. Thus, we identify a cytokine signal that must be received in muscle to control AKT activity and metabolic homeostasis.
Collapse
Affiliation(s)
- Katrin Kierdorf
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUnited Kingdom
- Department of Life SciencesImperial College LondonLondonUnited Kingdom
| | - Fabian Hersperger
- Institute of Neuropathology, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Jessica Sharrock
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUnited Kingdom
- Department of Life SciencesImperial College LondonLondonUnited Kingdom
| | - Crystal M Vincent
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUnited Kingdom
- Department of Life SciencesImperial College LondonLondonUnited Kingdom
| | - Pinar Ustaoglu
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUnited Kingdom
- Department of Life SciencesImperial College LondonLondonUnited Kingdom
| | - Jiawen Dou
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUnited Kingdom
| | - Attila Gyoergy
- Institute of Science and TechnologyKlosterneuburgAustria
| | - Olaf Groß
- Institute of Neuropathology, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Centre for Integrative Biological Signalling Studies (CIBSS)University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | | | - Marc S Dionne
- MRC Centre for Molecular Bacteriology and InfectionImperial College LondonLondonUnited Kingdom
| |
Collapse
|
15
|
Cell Adhesion-Mediated Actomyosin Assembly Regulates the Activity of Cubitus Interruptus for Hematopoietic Progenitor Maintenance in Drosophila. Genetics 2019; 212:1279-1300. [PMID: 31138608 PMCID: PMC6707476 DOI: 10.1534/genetics.119.302209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
The actomyosin network is involved in crucial cellular processes including morphogenesis, cell adhesion, apoptosis, proliferation, differentiation, and collective cell migration in Drosophila, Caenorhabditiselegans, and mammals. Here, we demonstrate that Drosophila larval blood stem-like progenitors require actomyosin activity for their maintenance. Genetic loss of the actomyosin network from progenitors caused a decline in their number. Likewise, the progenitor population increased upon sustained actomyosin activation via phosphorylation by Rho-associated kinase. We show that actomyosin positively regulates larval blood progenitors by controlling the maintenance factor Cubitus interruptus (Ci). Overexpression of the maintenance signal via a constitutively activated construct (ci.HA) failed to sustain Ci-155 in the absence of actomyosin components like Zipper (zip) and Squash (sqh), thus favoring protein kinase A (PKA)-independent regulation of Ci activity. Furthermore, we demonstrate that a change in cortical actomyosin assembly mediated by DE-cadherin modulates Ci activity, thereby determining progenitor status. Thus, loss of cell adhesion and downstream actomyosin activity results in desensitization of the progenitors to Hh signaling, leading to their differentiation. Our data reveal how cell adhesion and the actomyosin network cooperate to influence patterning, morphogenesis, and maintenance of the hematopoietic stem-like progenitor pool in the developing Drosophila hematopoietic organ.
Collapse
|
16
|
Wang W, Pan C, Huang Z, Yuan H, Chen J. WSV181 inhibits JAK/STAT signaling and promotes viral replication in Drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:20-28. [PMID: 30414403 DOI: 10.1016/j.dci.2018.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/06/2018] [Accepted: 11/06/2018] [Indexed: 06/08/2023]
Abstract
The Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway plays a critical role in host defense against viral infections. Here, we report the use of the Drosophila model system to investigate the modulation of the JAK/STAT pathway by the white spot syndrome virus (WSSV) protein WSV181. WSV181 overexpression in transgenic flies resulted in the downregulation of STAT92E and STAT92E-targeted genes. This result indicates that WSV181 can suppress JAK/STAT signaling by controlling STAT92E expression. An infection experiment was carried out on transgenic Drosophila infected with Drosophila C virus and on Litopenaeus vannamei injected with recombinant WSV181 and WSSV. The increased viral load and suppressed transcript levels of JAK/STAT pathway components indicate that WSV181 can promote viral proliferation by inhibiting the JAK/STAT pathway. This study provided evidence for the role of WSV181 in viral replication and revealed a new mechanism through which WSSV evades host immunity to maintain persistent infection.
Collapse
Affiliation(s)
- Wei Wang
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian, 350108, China.
| | - Changkun Pan
- Technology and Data Department of Technology Center, PoolingMed Co., Ltd., Hangzhou, Zhejiang, 310053, China
| | - Zongliang Huang
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Huifang Yuan
- School of Biological Sciences, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Jianming Chen
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian, 350108, China.
| |
Collapse
|
17
|
Herrera SC, Bach EA. JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates. Development 2019; 146:dev167643. [PMID: 30696713 PMCID: PMC6361132 DOI: 10.1242/dev.167643] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
The JAK/STAT pathway is a conserved metazoan signaling system that transduces cues from extracellular cytokines into transcriptional changes in the nucleus. JAK/STAT signaling is best known for its roles in immunity. However, recent work has demonstrated that it also regulates critical homeostatic processes in germline and somatic stem cells, as well as regenerative processes in several tissues, including the gonad, intestine and appendages. Here, we provide an overview of JAK/STAT signaling in stem cells and regeneration, focusing on Drosophila and highlighting JAK/STAT pathway functions in proliferation, survival and cell competition that are conserved between Drosophila and vertebrates.
Collapse
Affiliation(s)
- Salvador C Herrera
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Erika A Bach
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
18
|
Trivedi S, Starz-Gaiano M. Drosophila Jak/STAT Signaling: Regulation and Relevance in Human Cancer and Metastasis. Int J Mol Sci 2018; 19:ijms19124056. [PMID: 30558204 PMCID: PMC6320922 DOI: 10.3390/ijms19124056] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/08/2018] [Accepted: 12/11/2018] [Indexed: 12/26/2022] Open
Abstract
Over the past three-decades, Janus kinase (Jak) and signal transducer and activator of transcription (STAT) signaling has emerged as a paradigm to understand the involvement of signal transduction in development and disease pathology. At the molecular level, cytokines and interleukins steer Jak/STAT signaling to transcriptional regulation of target genes, which are involved in cell differentiation, migration, and proliferation. Jak/STAT signaling is involved in various types of blood cell disorders and cancers in humans, and its activation is associated with carcinomas that are more invasive or likely to become metastatic. Despite immense information regarding Jak/STAT regulation, the signaling network has numerous missing links, which is slowing the progress towards developing drug therapies. In mammals, many components act in this cascade, with substantial cross-talk with other signaling pathways. In Drosophila, there are fewer pathway components, which has enabled significant discoveries regarding well-conserved regulatory mechanisms. Work across species illustrates the relevance of these regulators in humans. In this review, we showcase fundamental Jak/STAT regulation mechanisms in blood cells, stem cells, and cell motility. We examine the functional relevance of key conserved regulators from Drosophila to human cancer stem cells and metastasis. Finally, we spotlight less characterized regulators of Drosophila Jak/STAT signaling, which stand as promising candidates to be investigated in cancer biology. These comparisons illustrate the value of using Drosophila as a model for uncovering the roles of Jak/STAT signaling and the molecular means by which the pathway is controlled.
Collapse
Affiliation(s)
- Sunny Trivedi
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
19
|
Abstract
Like humans, insects face the threat of viral infection. Despite having repercussions on human health and disease, knowledge gaps exist for how insects cope with viral pathogens. Drosophila melanogaster serves as an ideal insect model due to its genetic tractability. When encountering a pathogen, two major approaches to fight disease are resistance strategies and tolerance strategies. Disease resistance strategies promote the health of the infected host by reducing pathogen load. Multiple disease resistance mechanisms have been identified in Drosophila: RNA interference, Jak/STAT signaling, Toll signaling, IMD signaling, and autophagy. Disease tolerance mechanisms, in contrast, do not reduce pathogen load directly, but rather mitigate the stress and damage incurred by infection. The main benefit of tolerance mechanisms may therefore be to provide the host with time to engage antiviral resistance mechanisms that eliminate the threat. In this review, antiviral resistance mechanisms used by Drosophila will be described and compared to mammalian antiviral mechanisms. Disease tolerance will then be explained in a broader context as this is a burgeoning field of study.
Collapse
Affiliation(s)
- Jonathan Chow
- Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jonathan C Kagan
- Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States.
| |
Collapse
|
20
|
Bao R, Dia SE, Issa HA, Alhusein D, Friedrich M. Comparative Evidence of an Exceptional Impact of Gene Duplication on the Developmental Evolution of Drosophila and the Higher Diptera. Front Ecol Evol 2018. [DOI: 10.3389/fevo.2018.00063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
21
|
Sahu A, Ghosh R, Deshpande G, Prasad M. A Gap Junction Protein, Inx2, Modulates Calcium Flux to Specify Border Cell Fate during Drosophila oogenesis. PLoS Genet 2017; 13:e1006542. [PMID: 28114410 PMCID: PMC5256874 DOI: 10.1371/journal.pgen.1006542] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/15/2016] [Indexed: 01/09/2023] Open
Abstract
Intercellular communication mediated by gap junction (GJ) proteins is indispensable during embryogenesis, tissue regeneration and wound healing. Here we report functional analysis of a gap junction protein, Innexin 2 (Inx2), in cell type specification during Drosophila oogenesis. Our data reveal a novel involvement of Inx2 in the specification of Border Cells (BCs), a migratory cell type, whose identity is determined by the cell autonomous STAT activity. We show that Inx2 influences BC fate specification by modulating STAT activity via Domeless receptor endocytosis. Furthermore, detailed experimental analysis has uncovered that Inx2 also regulates a calcium flux that transmits across the follicle cells. We propose that Inx2 mediated calcium flux in the follicle cells stimulates endocytosis by altering Dynamin (Shibire) distribution which is in turn critical for careful calibration of STAT activation and, thus for BC specification. Together our data provide unprecedented molecular insights into how gap junction proteins can regulate cell-type specification. Gap junction mediated intercellular communication modulates several processes during development, morphogenesis and normal tissue homeostasis. While gap junction proteins play an important role during intercellular communication, the underlying molecular mechanism(s) as to how they regulate diverse signaling cascades are unclear. By employing the Drosophila melanogaster oogenesis model we have characterized the role of gap junction protein, Innexin 2 (Inx2), in cell fate specification during Drosophila oogenesis. Our data demonstrate that loss of inx2 affects border cell specification. Border cells are a small group of 6–8 follicle cells that acquire migratory fate in response to the activation of JAK-STAT signaling. We show that perturbing Inx2 levels in the follicle cells inhibits JAK-STAT signaling thereby adversely influencing border cell fate specification. Using live cell imaging and molecular genetic analysis, we have elucidated the molecular mechanism underlying Inx2 function in this process. We show that Inx2 mediates inter-follicular calcium flux that is critical for border cell fate determination. Furthermore, our observations indicate that Inx2 regulates Domeless receptor internalization possibly via influencing distribution of Drosophila Dynamin, Shibire in the follicle cells. Taken together these results suggest a functional link between Inx2, calcium flux and receptor endocytosis during border cell fate specification in Drosophila oogenesis.
Collapse
Affiliation(s)
- Aresh Sahu
- Department of Biological Sciences Indian Institute of Science Education & Research Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
| | - Ritabrata Ghosh
- Department of Biological Sciences Indian Institute of Science Education & Research Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
| | - Girish Deshpande
- Department of Molecular Biology Princeton University, Princeton, NJ, United States of America
- Indian Institute of Science Education and Research Pune. Pune Maharashtra, India
| | - Mohit Prasad
- Department of Biological Sciences Indian Institute of Science Education & Research Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
- * E-mail:
| |
Collapse
|
22
|
β-Arrestin 1's Interaction with TC45 Attenuates Stat signaling by dephosphorylating Stat to inhibit antimicrobial peptide expression. Sci Rep 2016; 6:35808. [PMID: 27782165 PMCID: PMC5080627 DOI: 10.1038/srep35808] [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: 06/13/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022] Open
Abstract
Impaired phosphatase activity leads to the persistent activation of signal transducers and activators of transcription (Stat). In mammals, Stat family members are often phosphorylated or dephosphorylated by the same enzymes. To date, only one Stat similar to mammalian Stat5a/b has been found in crustaceans and there have been few studies in Stat signal regulation in crustaceans. Here, we report that β-arrestin1 interacts with TC45 (45-kDa form of T cell protein tyrosine phosphatase) in the nucleus to attenuate Stat signaling by promoting dephosphorylation of Stat. Initially, we showed that Stat translocates into the nucleus to induce antimicrobial peptide (AMP) expression after bacterial infection. βArr1 enters the nucleus of hemocytes and recruits TC45 to form the βarr1-TC45-Stat complex, which dephosphorylates Stat efficiently. The interaction of TC45 with Stat decreased and Stat phosphorylation increased in βarr1-silenced shrimp (Marsupenaeus japonicus) after challenge with Vibrio anguillarum. βArr1 directly interacts with Stat in nucleus and accelerates Stat dephosphorylation by recruiting TC45 after V. anguillarum challenge. Further study showed that βarr1 and TC45 also affect AMP expression, which is regulated by Stat. Therefore, βarr1 and TC45 are involved in the anti-V. anguillarum immune response by regulating Stat activity negatively to decrease AMP expression in shrimp.
Collapse
|
23
|
Vanha-Aho LM, Valanne S, Rämet M. Cytokines in Drosophila immunity. Immunol Lett 2015; 170:42-51. [PMID: 26730849 DOI: 10.1016/j.imlet.2015.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
Cytokines are a large and diverse group of small proteins that can affect many biological processes, but most commonly cytokines are known as mediators of the immune response. In the event of an infection, cytokines are produced in response to an immune stimulus, and they function as key regulators of the immune response. Cytokines come in many shapes and sizes, and although they vary greatly in structure, their functions have been well conserved in evolution. The immune signaling pathways that respond to cytokines are remarkably conserved from fly to man. Therefore, Drosophila melanogaster, provides an excellent platform for studying the biology and function of cytokines. In this review, we will describe the cytokines and cytokine-like molecules found in the fly and discuss their roles in host immunity.
Collapse
Affiliation(s)
- Leena-Maija Vanha-Aho
- Laboratory of Experimental Immunology, BioMediTech, 33014 University of Tampere, Finland.
| | - Susanna Valanne
- Laboratory of Experimental Immunology, BioMediTech, 33014 University of Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech, 33014 University of Tampere, Finland; PEDEGO Research Unit, and Medical Research Center Oulu, University of Oulu and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
24
|
Fisher KH, Stec W, Brown S, Zeidler MP. Mechanisms of JAK/STAT pathway negative regulation by the short coreceptor Eye Transformer/Latran. Mol Biol Cell 2015; 27:434-41. [PMID: 26658615 PMCID: PMC4751595 DOI: 10.1091/mbc.e15-07-0546] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/01/2015] [Indexed: 11/30/2022] Open
Abstract
The short receptor Et/Lat negatively regulates Drosophila JAK/STAT signaling. It binds to intracellular components and the Domeless receptor but cannot bind ligands, thus generating a signaling-incompetent complex. Et/Lat is also more stable than Dome. The study provides insights into how short receptors negatively regulate signaling. Transmembrane receptors interact with extracellular ligands to transduce intracellular signaling cascades, modulate target gene expression, and regulate processes such as proliferation, apoptosis, differentiation, and homeostasis. As a consequence, aberrant signaling events often underlie human disease. Whereas the vertebrate JAK/STAT signaling cascade is transduced via multiple receptor combinations, the Drosophila pathway has only one full-length signaling receptor, Domeless (Dome), and a single negatively acting receptor, Eye Transformer/Latran (Et/Lat). Here we investigate the molecular mechanisms underlying Et/Lat activity. We demonstrate that Et/Lat negatively regulates the JAK/STAT pathway activity and can bind to Dome, thus reducing Dome:Dome homodimerization by creating signaling-incompetent Dome:Et/Lat heterodimers. Surprisingly, we find that Et/Lat is able to bind to both JAK and STAT92E but, despite the presence of putative cytokine-binding motifs, does not detectably interact with pathway ligands. We find that Et/Lat is trafficked through the endocytic machinery for lysosomal degradation but at a much slower rate than Dome, a difference that may enhance its ability to sequester Dome into signaling-incompetent complexes. Our data offer new insights into the molecular mechanism and regulation of Et/Lat in Drosophila that may inform our understanding of how short receptors function in other organisms.
Collapse
Affiliation(s)
- Katherine H Fisher
- Bateson Centre, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Wojciech Stec
- Bateson Centre, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Stephen Brown
- Sheffield RNAi Screening Facility, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Martin P Zeidler
- Bateson Centre, University of Sheffield, Sheffield S10 2TN, United Kingdom
| |
Collapse
|
25
|
Monahan AJ, Starz-Gaiano M. Socs36E limits STAT signaling via Cullin2 and a SOCS-box independent mechanism in the Drosophila egg chamber. Mech Dev 2015; 138 Pt 3:313-27. [PMID: 26277564 DOI: 10.1016/j.mod.2015.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/31/2015] [Accepted: 08/06/2015] [Indexed: 01/30/2023]
Abstract
The Suppressor of Cytokine Signaling (SOCS) proteins are critical, highly conserved feedback inhibitors of signal transduction cascades. The family of SOCS proteins is divided into two groups: ancestral and vertebrate-specific SOCS proteins. Vertebrate-specific SOCS proteins have been heavily studied as a result of their strong mutant phenotypes. However, the ancestral clade remains less studied, a potential result of genetic redundancies in mammals. Use of the genetically tractable organism Drosophila melanogaster enables in vivo assessment of signaling components and mechanisms with less concern about the functional redundancy observed in mammals. In this study, we investigated how the SOCS family member Suppressor of Cytokine Signaling at 36E (Socs36E) attenuates Janus Kinase/Signal Transducer and Activator of Transcription (Jak/STAT) activation during specification of motile border cells in Drosophila oogenesis. We found that Socs36E genetically interacts with the Cullin2 (Cul2) scaffolding protein. Like Socs36E, Cul2 is required to limit the number of motile cells in egg chambers. We demonstrated that loss of Cul2 in the follicle cells significantly increased nuclear STAT protein levels, which resulted in additional cells acquiring invasive properties. Further, reduction of Cul2 suppressed border cell migration defects that occur in a Stat92E-sensitized genetic background. Our data incorporated Cul2 into a previously described Jak/STAT-directed genetic regulatory network that is required to generate a discrete boundary between cell fates. We also found that Socs36E is able to attenuate STAT activity in the egg chamber when it does not have a functional SOCS box. Collectively, this work contributes mechanistic insight to a Jak/STAT regulatory genetic circuit, and suggests that Socs36E regulates Jak/STAT signaling via a Cul2-dependent mechanism, as well as by a Cullin-independent manner, in vivo.
Collapse
Affiliation(s)
- Amanda J Monahan
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD 21250, USA.
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
26
|
Shapiro-Kulnane L, Smolko AE, Salz HK. Maintenance of Drosophila germline stem cell sexual identity in oogenesis and tumorigenesis. Development 2015; 142:1073-82. [PMID: 25758221 DOI: 10.1242/dev.116590] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adult stem cells maintain tissue homeostasis by balancing self-renewal and differentiation. In Drosophila females, germline stem cells (GSCs) require Sex lethal (Sxl) to exit the stem cell state and to enter the differentiation pathway. Without Sxl GSCs do not differentiate and instead form tumors. Previous studies have shown that these tumors are not caused by a failure in the self-renewal/differentiation switch. Here, we show that Sxl is also necessary for the cell-autonomous maintenance of germ cell female identity and demonstrate that tumors are caused by the acquisition of male characteristics. Germ cells without Sxl protein exhibit a global derepression of testis genes, including Phf7, a male germline sexual identity gene. Phf7 is a key effector of the tumor-forming pathway, as it is both necessary and sufficient for tumor formation. In the absence of Sxl protein, inappropriate Phf7 expression drives tumor formation through a cell-autonomous mechanism that includes sex-inappropriate activation of Jak/Stat signaling. Remarkably, tumor formation requires a novel response to external signals emanating from the GSC niche, highlighting the importance of interactions between mutant cells and the surrounding normal cells that make up the tumor microenvironment. Derepression of testis genes, and inappropriate Phf7 expression, is also observed in germ cell tumors arising from the loss of bag of marbles (bam), demonstrating that maintenance of female sexual identity requires the concerted actions of Sxl and bam. Our work reveals that GSCs must maintain their sexual identity as they are reprogrammed into a differentiated cell, or risk tumorigenesis.
Collapse
Affiliation(s)
- Laura Shapiro-Kulnane
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4955, USA
| | - Anne Elizabeth Smolko
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4955, USA
| | - Helen Karen Salz
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4955, USA
| |
Collapse
|
27
|
Ren W, Zhang Y, Li M, Wu L, Wang G, Baeg GH, You J, Li Z, Lin X. Windpipe controls Drosophila intestinal homeostasis by regulating JAK/STAT pathway via promoting receptor endocytosis and lysosomal degradation. PLoS Genet 2015; 11:e1005180. [PMID: 25923769 PMCID: PMC4414558 DOI: 10.1371/journal.pgen.1005180] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/27/2015] [Indexed: 01/12/2023] Open
Abstract
The adult intestinal homeostasis is tightly controlled by proper proliferation and differentiation of intestinal stem cells. The JAK/STAT (Janus Kinase/Signal Transducer and Activator of Transcription) signaling pathway is essential for the regulation of adult stem cell activities and maintenance of intestinal homeostasis. Currently, it remains largely unknown how JAK/STAT signaling activities are regulated in these processes. Here we have identified windpipe (wdp) as a novel component of the JAK/STAT pathway. We demonstrate that Wdp is positively regulated by JAK/STAT signaling in Drosophila adult intestines. Loss of wdp activity results in the disruption of midgut homeostasis under normal and regenerative conditions. Conversely, ectopic expression of Wdp inhibits JAK/STAT signaling activity. Importantly, we show that Wdp interacts with the receptor Domeless (Dome), and promotes its internalization for subsequent lysosomal degradation. Together, these data led us to propose that Wdp acts as a novel negative feedback regulator of the JAK/STAT pathway in regulating intestinal homeostasis. Effective tissue homeostasis requires a proper balance between the removal of dead cells and production of new cells. Due to environmental challenges, the Drosophila midgut epithelial cells are damaged from time to time and intestinal stem cells (ISC) can accelerate their proliferative rate to replace the lost midgut epithelium. The JAK/STAT pathway plays essential roles in these progresses. Upon damage, Upd ligands produced by dying enterocytes (ECs) activate JAK/STAT signaling in ISCs to promote their proliferation and differentiation. However, after damage how JAK/STAT signaling is switched from a highly active state to a homeostatic state is not yet fully understood. In this study, we identified the leucine rich repeats (LRR) protein Windpipe (Wdp) as a novel negative feedback regulator of JAK/STAT signaling during intestinal development. Wdp expression was induced by high levels of JAK/STAT signaling in intestines. And loss of Wdp leads to midgut homeostasis loss and increased ISC proliferation. Furthermore, we found Wdp in turn negatively regulates JAK/STAT signaling activity through promoting Domeless receptor endocytosis and lysosomal degradation. In this way, high levels of JAK/STAT signaling is switched off by Wdp, which ensure ISCs return to the homeostatic state after tissue damage.
Collapse
Affiliation(s)
- Wenyan Ren
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Min Li
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical College, Wenzhou, China
| | - Longfei Wu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Guolun Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gyeong-Hun Baeg
- Department of Anatomy, National University of Singapore, Singapore
| | - Jia You
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Zhouhua Li
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Capital Normal University, Beijing, China
- * E-mail: (ZL); (XL)
| | - Xinhua Lin
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail: (ZL); (XL)
| |
Collapse
|
28
|
Chabu C, Xu T. Oncogenic Ras stimulates Eiger/TNF exocytosis to promote growth. Development 2014; 141:4729-39. [PMID: 25411211 DOI: 10.1242/dev.108092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Oncogenic mutations in Ras deregulate cell death and proliferation to cause cancer in a significant number of patients. Although normal Ras signaling during development has been well elucidated in multiple organisms, it is less clear how oncogenic Ras exerts its effects. Furthermore, cancers with oncogenic Ras mutations are aggressive and generally resistant to targeted therapies or chemotherapy. We identified the exocytosis component Sec15 as a synthetic suppressor of oncogenic Ras in an in vivo Drosophila mosaic screen. We found that oncogenic Ras elevates exocytosis and promotes the export of the pro-apoptotic ligand Eiger (Drosophila TNF). This blocks tumor cell death and stimulates overgrowth by activating the JNK-JAK-STAT non-autonomous proliferation signal from the neighboring wild-type cells. Inhibition of Eiger/TNF exocytosis or interfering with the JNK-JAK-STAT non-autonomous proliferation signaling at various steps suppresses oncogenic Ras-mediated overgrowth. Our findings highlight important cell-intrinsic and cell-extrinsic roles of exocytosis during oncogenic growth and provide a new class of synthetic suppressors for targeted therapy approaches.
Collapse
Affiliation(s)
- Chiswili Chabu
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, Boyer Center for Molecular Medicine, 295 Congress Avenue, New Haven, CT 06536, USA
| | - Tian Xu
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, Boyer Center for Molecular Medicine, 295 Congress Avenue, New Haven, CT 06536, USA
| |
Collapse
|
29
|
Doherty J, Sheehan AE, Bradshaw R, Fox AN, Lu TY, Freeman MR. PI3K signaling and Stat92E converge to modulate glial responsiveness to axonal injury. PLoS Biol 2014; 12:e1001985. [PMID: 25369313 PMCID: PMC4219656 DOI: 10.1371/journal.pbio.1001985] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/22/2014] [Indexed: 11/18/2022] Open
Abstract
Activation of glial cells following axon injury is mediated by a positive feedback loop downstream of the glial phagocytic receptor Draper, allowing the strength of the response to match the severity of injury. Glial cells are exquisitely sensitive to neuronal injury but mechanisms by which glia establish competence to respond to injury, continuously gauge neuronal health, and rapidly activate reactive responses remain poorly defined. Here, we show glial PI3K signaling in the uninjured brain regulates baseline levels of Draper, a receptor essential for Drosophila glia to sense and respond to axonal injury. After injury, Draper levels are up-regulated through a Stat92E-modulated, injury-responsive enhancer element within the draper gene. Surprisingly, canonical JAK/STAT signaling does not regulate draper expression. Rather, we find injury-induced draper activation is downstream of the Draper/Src42a/Shark/Rac1 engulfment signaling pathway. Thus, PI3K signaling and Stat92E are critical in vivo regulators of glial responsiveness to axonal injury. We provide evidence for a positive auto-regulatory mechanism whereby signaling through the injury-responsive Draper receptor leads to Stat92E-dependent, transcriptional activation of the draper gene. We propose that Drosophila glia use this auto-regulatory loop as a mechanism to adjust their reactive state following injury. Acute injuries of the central nervous system (CNS) trigger a robust reaction from glial cells—a non-neuronal population of cells that regulate and support neural development and physiology. Although this process occurs after all types of CNS trauma in mammals, how it is activated and its precise role in recovery remain poorly understood. Using the fruit fly Drosophila melanogaster as a model, we previously identified a cell surface receptor called Draper, which is required for the activation of glia after local axon injury (“axotomy”) and for the removal of degenerating axonal debris by phagocytosis. Here, we show that regulation of Draper protein levels and glial activation through the Draper signaling pathway are mediated by the well-conserved PI3K and signal transducer and activator of transcription (STAT) signaling cascades. We find that STAT transcriptional activity is activated in glia in response to axotomy, and identify an injury-responsive regulatory element within the draper gene that appears to be directly modulated by STAT. Interestingly, the intensity of STAT activity in glial cells after axotomy correlates tightly with the number of local severed axons, indicating that Drosophila glia are able to fine-tune their response to neuronal injury according to its severity. In summary, we propose that the initial phagocytic competence of glia is regulated by setting Draper baseline levels (via PI3K), whereas injury-activated glial phagocytic activity is modulated through a positive feedback loop that requires STAT-dependent activation of draper. We speculate that the level of activation of this cascade is determined by glial cell recognition of Draper ligands present on degenerating axon material, thereby matching the levels of glial reactivity to the amount of injured axonal material.
Collapse
Affiliation(s)
- Johnna Doherty
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Amy E. Sheehan
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Rachel Bradshaw
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - A. Nicole Fox
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Tsai-Yi Lu
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Marc R. Freeman
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
30
|
Amoyel M, Simons BD, Bach EA. Neutral competition of stem cells is skewed by proliferative changes downstream of Hh and Hpo. EMBO J 2014; 33:2295-313. [PMID: 25092766 PMCID: PMC4253521 DOI: 10.15252/embj.201387500] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Neutral competition, an emerging feature of stem cell homeostasis, posits that individual stem cells can be lost and replaced by their neighbors stochastically, resulting in chance dominance of a clone at the niche. A single stem cell with an oncogenic mutation could bias this process and clonally spread the mutation throughout the stem cell pool. The Drosophila testis provides an ideal system for testing this model. The niche supports two stem cell populations that compete for niche occupancy. Here, we show that cyst stem cells (CySCs) conform to the paradigm of neutral competition and that clonal deregulation of either the Hedgehog (Hh) or Hippo (Hpo) pathway allows a single CySC to colonize the niche. We find that the driving force behind such behavior is accelerated proliferation. Our results demonstrate that a single stem cell colonizes its niche through oncogenic mutation by co-opting an underlying homeostatic process.
Collapse
Affiliation(s)
- Marc Amoyel
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK Wellcome Trust-CRUK Gurdon Institute, University of Cambridge, Cambridge, UK Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Erika A Bach
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA The Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
31
|
Chen Q, Giedt M, Tang L, Harrison DA. Tools and methods for studying the Drosophila JAK/STAT pathway. Methods 2014; 68:160-72. [DOI: 10.1016/j.ymeth.2014.03.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/17/2014] [Accepted: 03/19/2014] [Indexed: 12/29/2022] Open
|
32
|
Myllymäki H, Rämet M. JAK/STAT Pathway inDrosophilaImmunity. Scand J Immunol 2014; 79:377-85. [DOI: 10.1111/sji.12170] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 03/21/2014] [Indexed: 12/24/2022]
Affiliation(s)
- H. Myllymäki
- Laboratory of Experimental Immunology; BioMediTech; University of Tampere; Tampere Finland
| | - M. Rämet
- Laboratory of Experimental Immunology; BioMediTech; University of Tampere; Tampere Finland
- Department of Pediatrics; Tampere University Hospital; Tampere Finland
- Department of Pediatrics; Medical Research Center Oulu; University of Oulu; Oulu Finland
- Department of Children and Adolescents; Oulu University Hospital; Oulu Finland
| |
Collapse
|
33
|
A comparison of midline and tracheal gene regulation during Drosophila development. PLoS One 2014; 9:e85518. [PMID: 24465586 PMCID: PMC3896416 DOI: 10.1371/journal.pone.0085518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 11/28/2013] [Indexed: 11/19/2022] Open
Abstract
Within the Drosophila embryo, two related bHLH-PAS proteins, Single-minded and Trachealess, control development of the central nervous system midline and the trachea, respectively. These two proteins are bHLH-PAS transcription factors and independently form heterodimers with another bHLH-PAS protein, Tango. During early embryogenesis, expression of Single-minded is restricted to the midline and Trachealess to the trachea and salivary glands, whereas Tango is ubiquitously expressed. Both Single-minded/Tango and Trachealess/Tango heterodimers bind to the same DNA sequence, called the CNS midline element (CME) within cis-regulatory sequences of downstream target genes. While Single-minded/Tango and Trachealess/Tango activate some of the same genes in their respective tissues during embryogenesis, they also activate a number of different genes restricted to only certain tissues. The goal of this research is to understand how these two related heterodimers bind different enhancers to activate different genes, thereby regulating the development of functionally diverse tissues. Existing data indicates that Single-minded and Trachealess may bind to different co-factors restricted to various tissues, causing them to interact with the CME only within certain sequence contexts. This would lead to the activation of different target genes in different cell types. To understand how the context surrounding the CME is recognized by different bHLH-PAS heterodimers and their co-factors, we identified and analyzed novel enhancers that drive midline and/or tracheal expression and compared them to previously characterized enhancers. In addition, we tested expression of synthetic reporter genes containing the CME flanked by different sequences. Taken together, these experiments identify elements overrepresented within midline and tracheal enhancers and suggest that sequences immediately surrounding a CME help dictate whether a gene is expressed in the midline or trachea.
Collapse
|
34
|
Xu J, Cherry S. Viruses and antiviral immunity in Drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 42:67-84. [PMID: 23680639 PMCID: PMC3826445 DOI: 10.1016/j.dci.2013.05.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/26/2013] [Accepted: 05/02/2013] [Indexed: 05/10/2023]
Abstract
Viral pathogens present many challenges to organisms, driving the evolution of a myriad of antiviral strategies to combat infections. A wide variety of viruses infect invertebrates, including both natural pathogens that are insect-restricted, and viruses that are transmitted to vertebrates. Studies using the powerful tools in the model organism Drosophila have expanded our understanding of antiviral defenses against diverse viruses. In this review, we will cover three major areas. First, we will describe the tools used to study viruses in Drosophila. Second, we will survey the major viruses that have been studied in Drosophila. And lastly, we will discuss the well-characterized mechanisms that are active against these diverse pathogens, focusing on non-RNAi mediated antiviral mechanisms. Antiviral RNAi is discussed in another paper in this issue.
Collapse
Affiliation(s)
- Jie Xu
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
35
|
Morillo Prado JR, Srinivasan S, Fuller MT. The histone variant His2Av is required for adult stem cell maintenance in the Drosophila testis. PLoS Genet 2013; 9:e1003903. [PMID: 24244183 PMCID: PMC3820763 DOI: 10.1371/journal.pgen.1003903] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 09/08/2013] [Indexed: 12/19/2022] Open
Abstract
Many tissues are sustained by adult stem cells, which replace lost cells by differentiation and maintain their own population through self-renewal. The mechanisms through which adult stem cells maintain their identity are thus important for tissue homeostasis and repair throughout life. Here, we show that a histone variant, His2Av, is required cell autonomously for maintenance of germline and cyst stem cells in the Drosophila testis. The ATP-dependent chromatin-remodeling factor Domino is also required in this tissue for adult stem cell maintenance possibly by regulating the incorporation of His2Av into chromatin. Interestingly, although expression of His2Av was ubiquitous, its function was dispensable for germline and cyst cell differentiation, suggesting a specific role for this non-canonical histone in maintaining the stem cell state in these lineages.
Collapse
Affiliation(s)
- Jose Rafael Morillo Prado
- Department of Developmental Biology, Stanford University, School of Medicine Stanford, California, United States of America
| | | | | |
Collapse
|
36
|
Zeidler MP, Bausek N. The Drosophila JAK-STAT pathway. JAKSTAT 2013; 2:e25353. [PMID: 24069564 PMCID: PMC3772116 DOI: 10.4161/jkst.25353] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/23/2013] [Indexed: 02/03/2023] Open
Abstract
The conservation of signaling cascades between humans and Drosophila, over more than 500 million years of evolutionary time, means that the genetic tractability of the fly can be used to its full advantage to understand the functional requirements for JAK-STAT pathway signaling across species. Here we review the background to how the pathway was first identified and the first characterization of JAK-STAT pathway phenotypes in the Drosophila system, highlighting the molecular, functional, and disease-related conservation of the pathway.
Collapse
Affiliation(s)
- Martin P Zeidler
- MRC Centre for Development and Biomedical Genetics and the Department of Biomedical Science; The University of Sheffield; Sheffield, UK
| | | |
Collapse
|
37
|
Baumgartner R, Stocker H, Hafen E. The RNA-binding proteins FMR1, rasputin and caprin act together with the UBA protein lingerer to restrict tissue growth in Drosophila melanogaster. PLoS Genet 2013; 9:e1003598. [PMID: 23874212 PMCID: PMC3708825 DOI: 10.1371/journal.pgen.1003598] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 05/15/2013] [Indexed: 12/19/2022] Open
Abstract
Appropriate expression of growth-regulatory genes is essential to ensure normal animal development and to prevent diseases like cancer. Gene regulation at the levels of transcription and translational initiation mediated by the Hippo and Insulin signaling pathways and by the TORC1 complex, respectively, has been well documented. Whether translational control mediated by RNA-binding proteins contributes to the regulation of cellular growth is less clear. Here, we identify Lingerer (Lig), an UBA domain-containing protein, as growth suppressor that associates with the RNA-binding proteins Fragile X mental retardation protein 1 (FMR1) and Caprin (Capr) and directly interacts with and regulates the RNA-binding protein Rasputin (Rin) in Drosophila melanogaster. lig mutant organs overgrow due to increased proliferation, and a reporter for the JAK/STAT signaling pathway is upregulated in a lig mutant situation. rin, Capr or FMR1 in combination as double mutants, but not the respective single mutants, display lig like phenotypes, implicating a redundant function of Rin, Capr and FMR1 in growth control in epithelial tissues. Thus, Lig regulates cell proliferation during development in concert with Rin, Capr and FMR1. Animal growth is orchestrated by controlled expression of growth-regulatory factors. This regulation is achieved at different molecular levels like transcription, translation initiation, and translational regulation. Whereas transcriptional control and translation initiation of growth components have been well studied, the role of translational control in this process is less well understood. Here, we describe Lingerer (Lig), an UBA domain-containing protein, as a new growth suppressor that associates with the three RNA-binding proteins Fragile X mental retardation protein 1 (FMR1), Rasputin (Rin) and Caprin (Capr). Drosophila FMR1, Rin and Capr orthologs are known translational regulators. In lig mutants and in FMR1, Capr and rin in combination as double mutants, organ size is increased due to excess proliferation. These data unveil a growth-regulatory function of Lig, and a redundant function of the RNA-binding proteins FMR1, Capr and Rin. Our findings demonstrate the involvement of mRNA-binding proteins in epithelial growth control and may also contribute to a better molecular understanding of the Fragile X mental retardation syndrome.
Collapse
Affiliation(s)
- Roland Baumgartner
- Institute of Molecular Systems Biology, ETH Zürich, Wolfgang-Pauli-Strasse, Zürich, Switzerland
| | - Hugo Stocker
- Institute of Molecular Systems Biology, ETH Zürich, Wolfgang-Pauli-Strasse, Zürich, Switzerland
| | - Ernst Hafen
- Institute of Molecular Systems Biology, ETH Zürich, Wolfgang-Pauli-Strasse, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
38
|
Localised JAK/STAT pathway activation is required for Drosophila wing hinge development. PLoS One 2013; 8:e65076. [PMID: 23741461 PMCID: PMC3669132 DOI: 10.1371/journal.pone.0065076] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/22/2013] [Indexed: 01/02/2023] Open
Abstract
Extensive morphogenetic remodelling takes place during metamorphosis from a larval to an adult insect body plan. These changes are particularly intricate in the generation of the dipteran wing hinge, a complex structure that is derived from an apparently simple region of the wing imaginal disc. Using the characterisation of original outstretched alleles of the unpaired locus as a starting point, we demonstrate the role of JAK/STAT pathway signalling in the process of wing hinge development. We show that differences in JAK/STAT signalling within the proximal most of three lateral folds present in the wing imaginal disc is required for fold morphology and the subsequent differentiation of the first and second auxiliary sclerites as well as the posterior notal wing process. Changes in these domains are consistent with the established fate map of the wing disc. We show that outstretched wing posture phenotypes arise from the loss of a region of Unpaired expression in the proximal wing fold and demonstrate that this results in a decrease in JAK/STAT pathway activity. Finally we show that reduction of JAK/STAT pathway activity within the proximal wing fold is sufficient to phenocopy the outstretched phenotype. Taken together, we suggest that localised Unpaired expression and hence JAK/STAT pathway activity, is required for the morphogenesis of the adult wing hinge, providing new insights into the link between signal transduction pathways, patterning and development.
Collapse
|
39
|
Oldefest M, Nowinski J, Hung CW, Neelsen D, Trad A, Tholey A, Grötzinger J, Lorenzen I. Upd3--an ancestor of the four-helix bundle cytokines. Biochem Biophys Res Commun 2013; 436:66-72. [PMID: 23707937 DOI: 10.1016/j.bbrc.2013.04.107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 04/29/2013] [Indexed: 11/19/2022]
Abstract
The unpaired-like protein 3 (Upd3) is one of the three cytokines of Drosophila melanogaster supposed to activate the JAK/STAT signaling pathway (Janus tyrosine kinases/signal transducer and activator of transcription). This activation occurs via the type-I cytokine receptor domeless, an orthologue of gp130, the common signal transducer of all four-helix bundle interleukin-6 (IL-6) type cytokines. Both receptors are known to exist as preformed dimers in the plasma membrane and initiate the acute-phase response. These facts indicate an evolutionary relation between vertebrate IL-6 and the Drosophila protein Upd3. Here we presented data which strengthen this notion. Upd3 was recombinantly expressed, a renaturation and purification protocol was established which allows to obtain high amounts of biological active protein. This protein is, like human IL-6, a monomeric-α helical cytokine, implicating that Upd3 is an "ancestor" of the four-helix bundle cytokines.
Collapse
Affiliation(s)
- Mirja Oldefest
- Biochemisches Institut der Christian-Albrechts-Universität Kiel, Olshausenstr. 40, 24118 Kiel, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Sotillos S, Krahn M, Espinosa-Vázquez JM, Hombría JCG. Src kinases mediate the interaction of the apical determinant Bazooka/PAR3 with STAT92E and increase signalling efficiency in Drosophila ectodermal cells. Development 2013; 140:1507-16. [DOI: 10.1242/dev.092320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Intercellular communication depends on the correct organization of the signal transduction complexes. In many signalling pathways, the mechanisms controlling the overall cell polarity also localize components of these pathways to different domains of the plasma membrane. In the Drosophila ectoderm, the JAK/STAT pathway components are highly polarized with apical localization of the receptor, the associated kinase and the STAT92E protein itself. The apical localization of STAT92E is independent of the receptor complex and is due to its direct association with the apical determining protein Bazooka (Baz). Here, we find that Baz-STAT92E interaction depends on the presence of the Drosophila Src kinases. In the absence of Src, STAT92E cannot bind to Baz in cells or in whole embryos, and this correlates with an impairment of JAK/STAT signalling function. We believe that the requirement of Src proteins for STAT92E apical localization is mediated through Baz, as we can co-precipitate Src with Baz but not with STAT92E. This is the first time that a functional link between cell polarity, the JAK/STAT signalling pathway and the Src kinases has been established in a whole organism.
Collapse
Affiliation(s)
- Sol Sotillos
- Centro Andaluz de Biología del Desarrollo, CSIC/JA/UPO, Ctra de Utrera Km1, 41013 Sevilla, Spain
| | - Michael Krahn
- Stem Cell Biology, Department of Anatomy and Cell Biology, University of Goettingen, Justus-von-Liebig-Weg 11, 37 077 Goettingen, Germany
| | | | | |
Collapse
|
41
|
Zhao S, Chen D, Geng Q, Wang Z. The highly conserved LAMMER/CLK2 protein kinases prevent germ cell overproliferation in Drosophila. Dev Biol 2013; 376:163-70. [DOI: 10.1016/j.ydbio.2013.01.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/14/2013] [Accepted: 01/23/2013] [Indexed: 10/27/2022]
|
42
|
Amoyel M, Sanny J, Burel M, Bach EA. Hedgehog is required for CySC self-renewal but does not contribute to the GSC niche in the Drosophila testis. Development 2012; 140:56-65. [PMID: 23175633 DOI: 10.1242/dev.086413] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Drosophila testis harbors two types of stem cells: germ line stem cells (GSCs) and cyst stem cells (CySCs). Both stem cell types share a physical niche called the hub, located at the apical tip of the testis. The niche produces the JAK/STAT ligand Unpaired (Upd) and BMPs to maintain CySCs and GSCs, respectively. However, GSCs also require BMPs produced by CySCs, and as such CySCs are part of the niche for GSCs. Here we describe a role for another secreted ligand, Hedgehog (Hh), produced by niche cells, in the self-renewal of CySCs. Hh signaling cell-autonomously regulates CySC number and maintenance. The Hh and JAK/STAT pathways act independently and non-redundantly in CySC self-renewal. Finally, Hh signaling does not contribute to the niche function of CySCs, as Hh-sustained CySCs are unable to maintain GSCs in the absence of Stat92E. Therefore, the extended niche function of CySCs is solely attributable to JAK/STAT pathway function.
Collapse
Affiliation(s)
- Marc Amoyel
- Department of Biochemistry and Molecular Biology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | |
Collapse
|
43
|
Abstract
The compound eye of the fruit fly, Drosophila melanogaster, has for decades been used extensively to study a number of critical developmental processes including tissue development, pattern formation, cell fate specification, and planar cell polarity. To a lesser degree it has been used to examine the cell cycle and tissue proliferation. Discovering the mechanisms that balance tissue growth and cell death in developing epithelia has traditionally been the realm of those using the wing disc. However, over the last decade a series of observations has demonstrated that the eye is a suitable and maybe even preferable tissue for studying tissue growth. This review will focus on how growth of the retina is controlled by the genes and pathways that govern the specification of tissue fate, the division of the epithelium into dorsal-ventral compartments, the initiation, and progression of the morphogenetic furrow and the second mitotic wave.
Collapse
Affiliation(s)
- Justin P Kumar
- Department of Biology, Indiana University, Bloomington, USA.
| |
Collapse
|
44
|
Srinivasan S, Mahowald AP, Fuller MT. The receptor tyrosine phosphatase Lar regulates adhesion between Drosophila male germline stem cells and the niche. Development 2012; 139:1381-90. [PMID: 22378638 DOI: 10.1242/dev.070052] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The stem cell niche provides a supportive microenvironment to maintain adult stem cells in their undifferentiated state. Adhesion between adult stem cells and niche cells or the local basement membrane ensures retention of stem cells in the niche environment. Drosophila male germline stem cells (GSCs) attach to somatic hub cells, a component of their niche, through E-cadherin-mediated adherens junctions, and orient their centrosomes toward these localized junctional complexes to carry out asymmetric divisions. Here we show that the transmembrane receptor tyrosine phosphatase Leukocyte-antigen-related-like (Lar), which is best known for its function in axonal migration and synapse morphogenesis in the nervous system, helps maintain GSCs at the hub by promoting E-cadherin-based adhesion between hub cells and GSCs. Lar is expressed in GSCs and early spermatogonial cells and localizes to the hub-GSC interface. Loss of Lar function resulted in a reduced number of GSCs at the hub. Lar function was required cell-autonomously in germ cells for proper localization of Adenomatous polyposis coli 2 and E-cadherin at the hub-GSC interface and for the proper orientation of centrosomes in GSCs. Ultrastructural analysis revealed that in Lar mutants the adherens junctions between hub cells and GSCs lack the characteristic dense staining seen in wild-type controls. Thus, the Lar receptor tyrosine phosphatase appears to polarize and retain GSCs through maintenance of localized E-cadherin-based adherens junctions.
Collapse
Affiliation(s)
- Shrividhya Srinivasan
- Department of Developmental Biology, Stanford University, School of Medicine, Stanford, CA 94305, USA
| | | | | |
Collapse
|
45
|
Abstract
The deleterious and sometimes fatal outcomes of bacterial infectious diseases are the net result of the interactions between the pathogen and the host, and the genetically tractable fruit fly, Drosophila melanogaster, has emerged as a valuable tool for modeling the pathogen-host interactions of a wide variety of bacteria. These studies have revealed that there is a remarkable conservation of bacterial pathogenesis and host defence mechanisms between higher host organisms and Drosophila. This review presents an in-depth discussion of the Drosophila immune response, the Drosophila killing model, and the use of the model to examine bacterial-host interactions. The recent introduction of the Drosophila model into the oral microbiology field is discussed, specifically the use of the model to examine Porphyromonas gingivalis-host interactions, and finally the potential uses of this powerful model system to further elucidate oral bacterial-host interactions are addressed.
Collapse
Affiliation(s)
- Christina O Igboin
- Division of Oral Biology, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
46
|
Grönholm J, Kaustio M, Myllymäki H, Kallio J, Saarikettu J, Kronhamn J, Valanne S, Silvennoinen O, Rämet M. Not4 enhances JAK/STAT pathway‐dependent gene expression in
Drosophila
and in human cells. FASEB J 2011; 26:1239-50. [DOI: 10.1096/fj.11-195875] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Juha Grönholm
- Laboratory of Molecular Immunology and Cytokine Receptor SignalingInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
| | - Meri Kaustio
- Laboratory of Experimental ImmunologyInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
| | - Henna Myllymäki
- Laboratory of Experimental ImmunologyInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
| | - Jenni Kallio
- Laboratory of Experimental ImmunologyInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
| | - Juha Saarikettu
- Laboratory of Molecular Immunology and Cytokine Receptor SignalingInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
| | | | - Susanna Valanne
- Laboratory of Molecular Immunology and Cytokine Receptor SignalingInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
| | - Olli Silvennoinen
- Laboratory of Molecular Immunology and Cytokine Receptor SignalingInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
- Science Center, Centre for Laboratory MedicineTampere University HospitalTampereFinland
| | - Mika Rämet
- Laboratory of Experimental ImmunologyInstitute of Biomedical TechnologyUniversity of TampereTampereFinland
- Department of PediatricsTampere University HospitalTampereFinland
| |
Collapse
|
47
|
Maruyama R, Andrew DJ. Drosophila as a model for epithelial tube formation. Dev Dyn 2011; 241:119-35. [PMID: 22083894 DOI: 10.1002/dvdy.22775] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2011] [Indexed: 12/17/2022] Open
Abstract
Epithelial tubular organs are essential for life in higher organisms and include the pancreas and other secretory organs that function as biological factories for the synthesis and delivery of secreted enzymes, hormones, and nutrients essential for tissue homeostasis and viability. The lungs, which are necessary for gas exchange, vocalization, and maintaining blood pH, are organized as highly branched tubular epithelia. Tubular organs include arteries, veins, and lymphatics, high-speed passageways for delivery and uptake of nutrients, liquids, gases, and immune cells. The kidneys and components of the reproductive system are also epithelial tubes. Both the heart and central nervous system of many vertebrates begin as epithelial tubes. Thus, it is not surprising that defects in tube formation and maintenance underlie many human diseases. Accordingly, a thorough understanding how tubes form and are maintained is essential to developing better diagnostics and therapeutics. Among the best-characterized tubular organs are the Drosophila salivary gland and trachea, organs whose relative simplicity have allowed for in depth analysis of gene function, yielding key mechanistic insight into tube initiation, remodeling and maintenance. Here, we review our current understanding of salivary gland and trachea formation - highlighting recent discoveries into how these organs attain their final form and function.
Collapse
Affiliation(s)
- Rika Maruyama
- The Johns Hopkins University School of Medicine, Department of Cell Biology, Baltimore, Maryland 21205-2196, USA
| | | |
Collapse
|
48
|
Sheng XR, Matunis E. Live imaging of the Drosophila spermatogonial stem cell niche reveals novel mechanisms regulating germline stem cell output. Development 2011; 138:3367-76. [PMID: 21752931 DOI: 10.1242/dev.065797] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adult stem cells modulate their output by varying between symmetric and asymmetric divisions, but have rarely been observed in living intact tissues. Germline stem cells (GSCs) in the Drosophila testis are anchored to somatic hub cells and were thought to exclusively undergo oriented asymmetric divisions, producing one stem cell that remains hub-anchored and one daughter cell displaced out of the stem cell-maintaining micro-environment (niche). We developed extended live imaging of the Drosophila testis niche, allowing us to track individual germline cells. Surprisingly, new wild-type GSCs are generated in the niche during steady-state tissue maintenance by a previously undetected event we term 'symmetric renewal', where interconnected GSC-daughter cell pairs swivel such that both cells contact the hub. We also captured GSCs undergoing direct differentiation by detaching from the hub. Following starvation-induced GSC loss, GSC numbers are restored by symmetric renewals. Furthermore, upon more severe (genetically induced) GSC loss, both symmetric renewal and de-differentiation (where interconnected spermatogonia fragment into pairs while moving towards then establishing contact with the hub) occur simultaneously to replenish the GSC pool. Thus, stereotypically oriented stem cell divisions are not always correlated with an asymmetric outcome in cell fate, and changes in stem cell output are governed by altered signals in response to tissue requirements.
Collapse
Affiliation(s)
- X Rebecca Sheng
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
49
|
Dinardo S, Okegbe T, Wingert L, Freilich S, Terry N. lines and bowl affect the specification of cyst stem cells and niche cells in the Drosophila testis. Development 2011; 138:1687-96. [PMID: 21486923 DOI: 10.1242/dev.057364] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To function properly, tissue-specific stem cells must reside in a niche. The Drosophila testis niche is one of few niches studied in vivo. Here, a single niche, comprising ten hub cells, maintains both germline stem cells (GSC) and somatic stem cells (CySC). Here, we show that lines is an essential CySC factor. Surprisingly, lines-depleted CySCs adopted several characteristics of hub cells, including the recruitment of new CySCs. This led us to examine the developmental relationship between CySCs and hub cells. In contrast to a previous report, we did not observe significant conversion of steady-state CySC progeny to hub fate. However, we found that these two cell types derive from a common precursor pool during gonadogenesis. Furthermore, lines mutant embryos exhibited gonads containing excess hub cells, indicating that lines represses hub cell fate during gonadogenesis. In many tissues, lines acts antagonistically to bowl, and we found that this is true for hub specification, establishing bowl as a positively acting factor in the development of the testis niche.
Collapse
Affiliation(s)
- Stephen Dinardo
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine and the Penn Institute for Regenerative Medicine, Philadelphia, PA 19104-6048, USA.
| | | | | | | | | |
Collapse
|
50
|
Zheng Q, Wang Y, Vargas E, DiNardo S. magu is required for germline stem cell self-renewal through BMP signaling in the Drosophila testis. Dev Biol 2011; 357:202-10. [PMID: 21723859 DOI: 10.1016/j.ydbio.2011.06.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Revised: 06/11/2011] [Accepted: 06/16/2011] [Indexed: 12/30/2022]
Abstract
Understanding how stem cells are maintained in their microenvironment (the niche) is vital for their application in regenerative medicine. Studies of Drosophila male germline stem cells (GSCs) have served as a paradigm in niche-stem cell biology. It is known that the BMP and JAK-STAT pathways are necessary for the maintenance of GSCs in the testis (Kawase et al., 2004; Kiger et al., 2001; Schulz et al., 2004; Shivdasani and Ingham, 2003; Tulina and Matunis, 2001). However, our recent work strongly suggests that BMP signaling is the primary pathway leading to GSC self-renewal (Leatherman and DiNardo, 2010). Here we show that magu controls GSC maintenance by modulating the BMP pathway. We found that magu was specifically expressed from hub cells, and accumulated at the testis tip. Testes from magu mutants exhibited a reduced number of GSCs, yet maintained a normal population of somatic stem cells and hub cells. Additionally, BMP pathway activity was reduced, whereas JAK-STAT activation was retained in mutant testes. Finally, GSC loss caused by the magu mutation could be suppressed by overactivating the BMP pathway in the germline.
Collapse
Affiliation(s)
- Qi Zheng
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, 19104, USA
| | | | | | | |
Collapse
|