1
|
Mukherjee A, Anoop C, Nongthomba U. What a tangled web we weave: crosstalk between JAK-STAT and other signalling pathways during development in Drosophila. FEBS J 2025. [PMID: 39821459 DOI: 10.1111/febs.17391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/26/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling pathway is a key player in animal development and physiology. Although it functions in a variety of processes, the net output of JAK-STAT signalling depends on its spatiotemporal activation, as well as extensive crosstalk with other signalling pathways. Drosophila, with its relatively simple signal transduction pathways and plethora of genetic analysis tools, is an ideal system for dissecting JAK-STAT signalling interactions. In this review, we explore studies in Drosophila revealing that JAK-STAT signalling lies at the nexus of a complex network of interlinked pathways, including epidermal growth factor receptor (EGFR), c-Jun N-terminal kinase (JNK), Notch, Insulin, Hippo, bone morphogenetic protein (BMP), Hedgehog (Hh) and Wingless (Wg). These pathways can synergise with or antagonise one another to produce a variety of outcomes. Given the conserved nature of signal transduction pathways, we conclude with our perspective on the implication of JAK-STAT signalling dysregulation in human diseases, and how studies in Drosophila have the potential to inform and influence clinical research.
Collapse
Affiliation(s)
- Amartya Mukherjee
- Department of Developmental Biology and Genetics, Indian Institute of Science (IISc), Bangalore, India
| | - Chaithra Anoop
- Department of Biological Science, Indian Institute of Science Education and Research (IISER), Mohali, India
| | - Upendra Nongthomba
- Department of Developmental Biology and Genetics, Indian Institute of Science (IISc), Bangalore, India
| |
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
|
Cho B, Shin M, Chang E, Son S, Shin I, Shim J. S-nitrosylation-triggered unfolded protein response maintains hematopoietic progenitors in Drosophila. Dev Cell 2024; 59:1075-1090.e6. [PMID: 38521056 DOI: 10.1016/j.devcel.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/27/2023] [Accepted: 02/29/2024] [Indexed: 03/25/2024]
Abstract
The Drosophila lymph gland houses blood progenitors that give rise to myeloid-like blood cells. Initially, blood progenitors proliferate, but later, they become quiescent to maintain multipotency before differentiation. Despite the identification of various factors involved in multipotency maintenance, the cellular mechanism controlling blood progenitor quiescence remains elusive. Here, we identify the expression of nitric oxide synthase in blood progenitors, generating nitric oxide for post-translational S-nitrosylation of protein cysteine residues. S-nitrosylation activates the Ire1-Xbp1-mediated unfolded protein response, leading to G2 cell-cycle arrest. Specifically, we identify the epidermal growth factor receptor as a target of S-nitrosylation, resulting in its retention within the endoplasmic reticulum and blockade of its receptor function. Overall, our findings highlight developmentally programmed S-nitrosylation as a critical mechanism that induces protein quality control in blood progenitors, maintaining their undifferentiated state by inhibiting cell-cycle progression and rendering them unresponsive to paracrine factors.
Collapse
Affiliation(s)
- Bumsik Cho
- Department of Life Science, College of Natural Science, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Natural Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Mingyu Shin
- Department of Life Science, College of Natural Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunji Chang
- Department of Life Science, College of Natural Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Seogho Son
- Department of Life Science, College of Natural Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Incheol Shin
- Department of Life Science, College of Natural Science, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Natural Science, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Jiwon Shim
- Department of Life Science, College of Natural Science, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Natural Science, Hanyang University, Seoul 04763, Republic of Korea; Research Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Republic of Korea.
| |
Collapse
|
4
|
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: 3] [Impact Index Per Article: 3.0] [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
|
5
|
Sinenko SA. Molecular Mechanisms of Drosophila Hematopoiesis. Acta Naturae 2024; 16:4-21. [PMID: 39188265 PMCID: PMC11345091 DOI: 10.32607/actanaturae.27410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/31/2024] [Indexed: 08/28/2024] Open
Abstract
As a model organism, the fruit fly (Drosophila melanogaster) has assumed a leading position in modern biological research. The Drosophila genetic system has a number of advantages making it a key model in investigating the molecular mechanisms of metazoan developmental processes. Over the past two decades, significant progress has been made in understanding the molecular mechanisms regulating Drosophila hematopoiesis. This review discusses the major advances in investigating the molecular mechanisms involved in maintaining the population of multipotent progenitor cells and their differentiation into mature hemocytes in the hematopoietic organ of the Drosophila larva. The use of the Drosophila hematopoietic organ as a model system for hematopoiesis has allowed to characterize the complex interactions between signaling pathways and transcription factors in regulating the maintenance and differentiation of progenitor cells through the signals from the hematopoietic niche, autocrine and paracrine signals, and the signals emanated by differentiated cells.
Collapse
Affiliation(s)
- S. A. Sinenko
- Institute of Cytology Russian Academy of Sciences, St. Petersburg, 194064 Russian Federation
| |
Collapse
|
6
|
Hof-Michel S, Cigoja L, Huhn S, Bökel C. Rel governs loser elimination during stem cell competition in the Drosophila testis. Eur J Cell Biol 2024; 103:151375. [PMID: 37995529 DOI: 10.1016/j.ejcb.2023.151375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
In the Drosophila testis, a group of stromal cells termed hub provides multiple niche signals for the surrounding germline and somatic stem cells. Stem cells of both populations compete for physical retention in the niche, and clones unable to transduce any one niche signal are rapidly eliminated from the stem cell pool by differentiation. We have mapped the transcriptomes of isolated somatic cyst stem cells and differentiated cyst cells, and found that the stem cells but not their differentiated progeny exhibit the signature of an innate immune response including the NF-κB transcription factor Relish (Rel). Related signalling pathways had previously implicated in cell competition in larval epithelia, prompting the question of whether NF-κB signalling was, despite the clear differences between the two competition scenarios, also involved in stem cell competition in the testis. Here we show i) that in the testis Rel is dispensable for stemness, ii) that loss of Rel or the upstream receptor Toll suppresses loser elimination following a variety of different triggers used to induce loser fate, and iii) that clonal Rel activation is sufficient for the displacement of neutral or winner cells from the niche, even if these cells otherwise retain stem cell properties.
Collapse
Affiliation(s)
- Silvana Hof-Michel
- Dept. of Developmental Genetics, Philipps University Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany
| | - Ljubinka Cigoja
- Dept. of Developmental Genetics, Philipps University Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany
| | - Sabina Huhn
- Dept. of Developmental Genetics, Philipps University Marburg, Karl-von-Frisch-Str. 8, 35043 Marburg, Germany
| | - Christian Bökel
- Core Facility Confocal and Multiphoton Microscopy, Universität Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
7
|
Zhao Y, Johansson E, Duan J, Han Z, Alenius M. Fat- and sugar-induced signals regulate sweet and fat taste perception in Drosophila. Cell Rep 2023; 42:113387. [PMID: 37934669 PMCID: PMC11212107 DOI: 10.1016/j.celrep.2023.113387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/29/2023] [Accepted: 10/22/2023] [Indexed: 11/09/2023] Open
Abstract
In this study, we investigate the interplay between taste perception and macronutrients. While sugar's and protein's self-regulation of taste perception is known, the role of fat remains unclear. We reveal that in Drosophila, fat overconsumption reduces fatty acid taste in favor of sweet perception. Conversely, sugar intake increases fatty acid perception and suppresses sweet taste. Genetic investigations show that the sugar signal, gut-secreted Hedgehog, suppresses sugar taste and enhances fatty acid perception. Fat overconsumption induces unpaired 2 (Upd2) secretion from adipose tissue to the hemolymph. We reveal taste neurons take up Upd2, which triggers Domeless suppression of fatty acid perception. We further show that the downstream JAK/STAT signaling enhances sweet perception and, via Socs36E, fine-tunes Domeless activity and the fatty acid taste perception. Together, our results show that sugar regulates Hedgehog signaling and fat induces Upd2 signaling to balance nutrient intake and to regulate sweet and fat taste perception.
Collapse
Affiliation(s)
- Yunpo Zhao
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | | | - Jianli Duan
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Zhe Han
- Center for Precision Disease Modeling, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mattias Alenius
- Department of Molecular Biology, Umeå University, Umeå, Sweden.
| |
Collapse
|
8
|
Hirschhäuser A, Molitor D, Salinas G, Großhans J, Rust K, Bogdan S. Single-cell transcriptomics identifies new blood cell populations in Drosophila released at the onset of metamorphosis. Development 2023; 150:dev201767. [PMID: 37681301 PMCID: PMC10560556 DOI: 10.1242/dev.201767] [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: 03/13/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
Drosophila blood cells called hemocytes form an efficient barrier against infections and tissue damage. During metamorphosis, hemocytes undergo tremendous changes in their shape and behavior, preparing them for tissue clearance. Yet, the diversity and functional plasticity of pupal blood cells have not been explored. Here, we combine single-cell transcriptomics and high-resolution microscopy to dissect the heterogeneity and plasticity of pupal hemocytes. We identified undifferentiated and specified hemocytes with different molecular signatures associated with distinct functions such as antimicrobial, antifungal immune defense, cell adhesion or secretion. Strikingly, we identified a highly migratory and immune-responsive pupal cell population expressing typical markers of the posterior signaling center (PSC), which is known to be an important niche in the larval lymph gland. PSC-like cells become restricted to the abdominal segments and are morphologically very distinct from typical Hemolectin (Hml)-positive plasmatocytes. G-TRACE lineage experiments further suggest that PSC-like cells can transdifferentiate to lamellocytes triggered by parasitoid wasp infestation. In summary, we present the first molecular description of pupal Drosophila blood cells, providing insights into blood cell functional diversification and plasticity during pupal metamorphosis.
Collapse
Affiliation(s)
- Alexander Hirschhäuser
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Darius Molitor
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University Marburg, Karl-von-Frisch-Strasse 8, 35043 Marburg, Germany
| | - Katja Rust
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Sven Bogdan
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| |
Collapse
|
9
|
Mahanta DK, Bhoi TK, Komal J, Samal I, Nikhil RM, Paschapur AU, Singh G, Kumar PVD, Desai HR, Ahmad MA, Singh PP, Majhi PK, Mukherjee U, Singh P, Saini V, Shahanaz, Srinivasa N, Yele Y. Insect-pathogen crosstalk and the cellular-molecular mechanisms of insect immunity: uncovering the underlying signaling pathways and immune regulatory function of non-coding RNAs. Front Immunol 2023; 14:1169152. [PMID: 37691928 PMCID: PMC10491481 DOI: 10.3389/fimmu.2023.1169152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Multicellular organisms are constantly subjected to pathogens that might be harmful. Although insects lack an adaptive immune system, they possess highly effective anti-infective mechanisms. Bacterial phagocytosis and parasite encapsulation are some forms of cellular responses. Insects often defend themselves against infections through a humoral response. This phenomenon includes the secretion of antimicrobial peptides into the hemolymph. Specific receptors for detecting infection are required for the recognition of foreign pathogens such as the proteins that recognize glucans and peptidoglycans, together referred to as PGRPs and βGRPs. Activation of these receptors leads to the stimulation of signaling pathways which further activates the genes encoding for antimicrobial peptides. Some instances of such pathways are the JAK-STAT, Imd, and Toll. The host immune response that frequently accompanies infections has, however, been circumvented by diseases, which may have assisted insects evolve their own complicated immune systems. The role of ncRNAs in insect immunology has been discussed in several notable studies and reviews. This paper examines the most recent research on the immune regulatory function of ncRNAs during insect-pathogen crosstalk, including insect- and pathogen-encoded miRNAs and lncRNAs, and provides an overview of the important insect signaling pathways and effector mechanisms activated by diverse pathogen invaders.
Collapse
Affiliation(s)
- Deepak Kumar Mahanta
- Department of Entomology, Dr. Rajendra Prasad Central Agricultural University, Samastipur, Bihar, India
| | - Tanmaya Kumar Bhoi
- Forest Protection Division, Indian Council of Forestry Research and Education (ICFRE) - Arid Forest Research Institute (ICFRE-AFRI), Jodhpur, Rajasthan, India
| | - J. Komal
- Department of Entomology, Navsari Agricultural University, Navsari, Gujarat, India
| | - Ipsita Samal
- ICAR-National Research Centre on Litchi, Mushahari, Ramna, Muzaffarpur, Bihar, India
| | - R. M. Nikhil
- Division of Entomology, Indian Agricultural Research Institute, New Delhi, India
| | - Amit Umesh Paschapur
- Crop Protection Division, Indian Council of Agricultural Research (ICAR)-Vivekananda Parvatiya Krishi Anusandhan Sansthan, Almora, Uttarakhand, India
| | - Gaurav Singh
- The Directorate of Research, Maharana Pratap Horticultural University, Karnal, Haryana, India
| | - P. V. Dinesh Kumar
- Department of Plant Pathology University of Agricultural Sciences, Bengaluru, Karnataka, India
| | - H. R. Desai
- Department of Entomology, Main Cotton Research Station, Navsari Agricultural University, Gujarat, India
| | - Mohammad Abbas Ahmad
- Department of Entomology, Dr. Rajendra Prasad Central Agricultural University, Samastipur, Bihar, India
| | - P. P. Singh
- Department of Entomology, Tirhut College of Agriculture, Dr. Rajendra Prasad Central Agricultural University, Samastipur, Bihar, India
| | - Prasanta Kumar Majhi
- Department of Plant Breeding and Genetics, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - U. Mukherjee
- Department of Entomology, Dr. Rajendra Prasad Central Agricultural University, Samastipur, Bihar, India
| | - Pushpa Singh
- Department of Entomology, Dr. Rajendra Prasad Central Agricultural University, Samastipur, Bihar, India
| | - Varun Saini
- Department of Entomology, Chaudhary Charan Singh Haryana Agricultural University, Hisar, Haryana, India
| | - Shahanaz
- Department of Entomology, College of Horticulture Mojerla, Sri Konda Laxman Telengana State Horticultural University, Wanaparthy, Telengana, India
| | - N. Srinivasa
- Department of Entomology and Agricultural Zoology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Yogesh Yele
- School of Crop Health Management Research, Council of Agricultural Research-National Institute of Biotic Stress Management (ICAR)- National Institute of Biotic Stress Management, Raipur, India
| |
Collapse
|
10
|
Benoit I, Di Curzio D, Civetta A, Douville RN. Drosophila as a Model for Human Viral Neuroinfections. Cells 2022; 11:cells11172685. [PMID: 36078091 PMCID: PMC9454636 DOI: 10.3390/cells11172685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
The study of human neurological infection faces many technical and ethical challenges. While not as common as mammalian models, the use of Drosophila (fruit fly) in the investigation of virus–host dynamics is a powerful research tool. In this review, we focus on the benefits and caveats of using Drosophila as a model for neurological infections and neuroimmunity. Through the examination of in vitro, in vivo and transgenic systems, we highlight select examples to illustrate the use of flies for the study of exogenous and endogenous viruses associated with neurological disease. In each case, phenotypes in Drosophila are compared to those in human conditions. In addition, we discuss antiviral drug screening in flies and how investigating virus–host interactions may lead to novel antiviral drug targets. Together, we highlight standardized and reproducible readouts of fly behaviour, motor function and neurodegeneration that permit an accurate assessment of neurological outcomes for the study of viral infection in fly models. Adoption of Drosophila as a valuable model system for neurological infections has and will continue to guide the discovery of many novel virus–host interactions.
Collapse
Affiliation(s)
- Ilena Benoit
- Department of Biology, University of Winnipeg, 599 Portage Avenue, Winnipeg, MB R3B 2G3, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Ave, Winnipeg, MB R2H 2A6, Canada
| | - Domenico Di Curzio
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Ave, Winnipeg, MB R2H 2A6, Canada
| | - Alberto Civetta
- Department of Biology, University of Winnipeg, 599 Portage Avenue, Winnipeg, MB R3B 2G3, Canada
| | - Renée N. Douville
- Department of Biology, University of Winnipeg, 599 Portage Avenue, Winnipeg, MB R3B 2G3, Canada
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, 351 Taché Ave, Winnipeg, MB R2H 2A6, Canada
- Correspondence:
| |
Collapse
|
11
|
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: 59] [Impact Index Per Article: 19.7] [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
|
12
|
Ran XQ, Gao L, Yan M, Kang CJ. Peroxiredoxin 4 Interacts With Domeless and Participates in Antibacterial Immune Response Through the JAK/STAT Pathway. Front Immunol 2022; 13:907183. [PMID: 35711411 PMCID: PMC9195186 DOI: 10.3389/fimmu.2022.907183] [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: 03/29/2022] [Accepted: 04/25/2022] [Indexed: 11/16/2022] Open
Abstract
The JAK/STAT pathway plays an important role in the development and immune responses of animals. In vertebrates, families of cytokines or growth factors act as activators of the JAK/STAT pathway; however, the activators for the JAK/STAT signaling pathway in arthropods are largely unknown. Herein we report a new ligand, peroxiredoxin 4 (Prx4), for the Domeless in the JAK/STAT pathway of shrimp Marsupenaeus japonicus. Prx4 was induced to secrete into the extracellular surroundings upon Vibrio challenge, which then facilitated the anti-Vibrio activity of shrimp by activating the phosphorylation and nuclear translocation of STAT and the expression of STAT-responsive antimicrobial peptides. Blocking the expression of Prx4 in vivo abrogated the activation of the JAK/STAT pathway by Vibrio infection, while injection of Prx4 protein activated the pathway. The interaction between Prx4 and Domeless was proved by immuno-precipitation and protein pull-down assays. Moreover, two cysteine residues in Prx4 that are critical for the interaction and Prx4’s anti-Vibrio role were identified, and the binding site in Domeless for Prx4 was proved to be the cytokine-binding homology module fragment. Taken together, our study revealed a new function for Prx4 enzyme and established a new enzyme-type ligand for the activation of the JAK/STAT pathway in an aquatic arthropod.
Collapse
Affiliation(s)
- Xiao-Qin Ran
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Lin Gao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Meng Yan
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Cui-Jie Kang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| |
Collapse
|
13
|
Goyal M, Tomar A, Madhwal S, Mukherjee T. Blood progenitor redox homeostasis through olfaction-derived systemic GABA in hematopoietic growth control in Drosophila. Development 2022; 149:273541. [PMID: 34850846 PMCID: PMC8733872 DOI: 10.1242/dev.199550] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022]
Abstract
The role of reactive oxygen species (ROS) in myeloid development is well established. However, its aberrant generation alters hematopoiesis. Thus, a comprehensive understanding of events controlling ROS homeostasis forms the central focus of this study. We show that, in homeostasis, myeloid-like blood progenitor cells of the Drosophila larvae, which reside in a specialized hematopoietic organ termed the lymph gland, use TCA to generate ROS. However, excessive ROS production leads to lymph gland growth retardation. Therefore, to moderate blood progenitor ROS, Drosophila larvae rely on olfaction and its downstream systemic GABA. GABA internalization and its breakdown into succinate by progenitor cells activates pyruvate dehydrogenase kinase (PDK), which controls inhibitory phosphorylation of pyruvate dehydrogenase (PDH). PDH is the rate-limiting enzyme that connects pyruvate to the TCA cycle and to oxidative phosphorylation. Thus, GABA metabolism via PDK activation maintains TCA activity and blood progenitor ROS homeostasis, and supports normal lymph gland growth. Consequently, animals that fail to smell also fail to sustain TCA activity and ROS homeostasis, which leads to lymph gland growth retardation. Overall, this study describes the requirement of animal odor-sensing and GABA in myeloid ROS regulation and hematopoietic growth control. Summary: Ablation of olfactory receptor neurons reveals that odor-sensing and GABA are involved in myeloid reactive oxygen species regulation and hematopoietic growth control.
Collapse
Affiliation(s)
- Manisha Goyal
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK, Bellary Road, Bangalore 560065, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka 560064, India
| | - Ajay Tomar
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK, Bellary Road, Bangalore 560065, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, Karnataka 560064, India
| | - Sukanya Madhwal
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK, Bellary Road, Bangalore 560065, India.,Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Tina Mukherjee
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK, Bellary Road, Bangalore 560065, India
| |
Collapse
|
14
|
Koranteng F, Cho B, Shim J. Intrinsic and Extrinsic Regulation of Hematopoiesis in Drosophila. Mol Cells 2022; 45:101-108. [PMID: 35253654 PMCID: PMC8926866 DOI: 10.14348/molcells.2022.2039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 01/12/2022] [Indexed: 11/27/2022] Open
Abstract
Drosophila melanogaster lymph gland, the primary site of hematopoiesis, contains myeloid-like progenitor cells that differentiate into functional hemocytes in the circulation of pupae and adults. Fly hemocytes are dynamic and plastic, and they play diverse roles in the innate immune response and wound healing. Various hematopoietic regulators in the lymph gland ensure the developmental and functional balance between progenitors and mature blood cells. In addition, systemic factors, such as nutrient availability and sensory inputs, integrate environmental variabilities to synchronize the blood development in the lymph gland with larval growth, physiology, and immunity. This review examines the intrinsic and extrinsic factors determining the progenitor states during hemocyte development in the lymph gland and provides new insights for further studies that may extend the frontier of our collective knowledge on hematopoiesis and innate immunity.
Collapse
Affiliation(s)
| | - Bumsik Cho
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Jiwon Shim
- Department of Life Science, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Science, Hanyang University, Seoul 04763, Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
15
|
Hao Y, Pan J, Chen Q, Gu H, Ji G, Yue G, Yang S. Jumu is required for the activation of JAK/STAT in Drosophila lymph gland development and epidermal wounds. Biochem Biophys Res Commun 2022; 591:68-75. [PMID: 34999256 DOI: 10.1016/j.bbrc.2021.12.115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 11/02/2022]
Abstract
The regulatory mechanism of hematopoiesis and innate immunity in Drosophila is highly similar to that in mammals, and Drosophila has become a suitable model to understand vertebrate hematopoiesis and the immune response. JAK-STAT signaling pathway components are widely conserved during evolution, and contribute to hematopoiesis and multiple tissue damage and immune responses. Here, we demonstrate that Stat92E is widely expressed in the lymph gland, and the loss of jumu inhibits the maintenance of the JAK/STAT pathway in the CZ and MZ but not in the PSC of the lymph gland. Furthermore, we found that clean puncture wounding of the larval epidermis can lead to the activation of JAK/STAT signaling and the generation of lamellocytes, and Jumu is required for the activation of JAK/STAT in response to epidermal wounds.
Collapse
Affiliation(s)
- Yangguang Hao
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China.
| | - Jichuan Pan
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China
| | - Qing Chen
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China
| | - Heze Gu
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China
| | - Guanglin Ji
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China
| | - Guanhua Yue
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China
| | - Shuting Yang
- Department of Basic Medical, Shenyang Medical College, Shenyang, 110034, China
| |
Collapse
|
16
|
Järvelä-Stölting M, Vesala L, Maasdorp MK, Ciantar J, Rämet M, Valanne S. Proteasome α6 Subunit Negatively Regulates the JAK/STAT Pathway and Blood Cell Activation in Drosophila melanogaster. Front Immunol 2021; 12:729631. [PMID: 35003057 PMCID: PMC8727353 DOI: 10.3389/fimmu.2021.729631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
JAK/STAT signaling regulates central biological functions such as development, cell differentiation and immune responses. In Drosophila, misregulated JAK/STAT signaling in blood cells (hemocytes) induces their aberrant activation. Using mass spectrometry to analyze proteins associated with a negative regulator of the JAK/STAT pathway, and by performing a genome-wide RNAi screen, we identified several components of the proteasome complex as negative regulators of JAK/STAT signaling in Drosophila. A selected proteasome component, Prosα6, was studied further. In S2 cells, Prosα6 silencing decreased the amount of the known negative regulator of the pathway, ET, leading to enhanced expression of a JAK/STAT pathway reporter gene. Silencing of Prosα6 in vivo resulted in activation of the JAK/STAT pathway, leading to the formation of lamellocytes, a specific hemocyte type indicative of hemocyte activation. This hemocyte phenotype could be partially rescued by simultaneous knockdown of either the Drosophila STAT transcription factor, or MAPKK in the JNK-pathway. Our results suggest a role for the proteasome complex components in the JAK/STAT pathway in Drosophila blood cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Mirva Järvelä-Stölting
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Vesala
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Matthew K. Maasdorp
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Joanna Ciantar
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research Unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Ophthalmology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Susanna Valanne
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- *Correspondence: Susanna Valanne,
| |
Collapse
|
17
|
Morin-Poulard I, Tian Y, Vanzo N, Crozatier M. Drosophila as a Model to Study Cellular Communication Between the Hematopoietic Niche and Blood Progenitors Under Homeostatic Conditions and in Response to an Immune Stress. Front Immunol 2021; 12:719349. [PMID: 34484226 PMCID: PMC8415499 DOI: 10.3389/fimmu.2021.719349] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/26/2021] [Indexed: 12/23/2022] Open
Abstract
In adult mammals, blood cells are formed from hematopoietic stem progenitor cells, which are controlled by a complex cellular microenvironment called "niche". Drosophila melanogaster is a powerful model organism to decipher the mechanisms controlling hematopoiesis, due both to its limited number of blood cell lineages and to the conservation of genes and signaling pathways throughout bilaterian evolution. Insect blood cells or hemocytes are similar to the mammalian myeloid lineage that ensures innate immunity functions. Like in vertebrates, two waves of hematopoiesis occur in Drosophila. The first wave takes place during embryogenesis. The second wave occurs at larval stages, where two distinct hematopoietic sites are identified: subcuticular hematopoietic pockets and a specialized hematopoietic organ called the lymph gland. In both sites, hematopoiesis is regulated by distinct niches. In hematopoietic pockets, sensory neurons of the peripheral nervous system provide a microenvironment that promotes embryonic hemocyte expansion and differentiation. In the lymph gland blood cells are produced from hematopoietic progenitors. A small cluster of cells called Posterior Signaling Centre (PSC) and the vascular system, along which the lymph gland develops, act collectively as a niche, under homeostatic conditions, to control the balance between maintenance and differentiation of lymph gland progenitors. In response to an immune stress such as wasp parasitism, lymph gland hematopoiesis is drastically modified and shifts towards emergency hematopoiesis, leading to increased progenitor proliferation and their differentiation into lamellocyte, a specific blood cell type which will neutralize the parasite. The PSC is essential to control this emergency response. In this review, we summarize Drosophila cellular and molecular mechanisms involved in the communication between the niche and hematopoietic progenitors, both under homeostatic and stress conditions. Finally, we discuss similarities between mechanisms by which niches regulate hematopoietic stem/progenitor cells in Drosophila and mammals.
Collapse
Affiliation(s)
| | - Yushun Tian
- MCD/UMR5077, Centre de Biologie Intégrative (CBI), Toulouse, France
| | - Nathalie Vanzo
- MCD/UMR5077, Centre de Biologie Intégrative (CBI), Toulouse, France
| | | |
Collapse
|
18
|
Cattenoz PB, Monticelli S, Pavlidaki A, Giangrande A. Toward a Consensus in the Repertoire of Hemocytes Identified in Drosophila. Front Cell Dev Biol 2021; 9:643712. [PMID: 33748138 PMCID: PMC7969988 DOI: 10.3389/fcell.2021.643712] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/12/2021] [Indexed: 01/16/2023] Open
Abstract
The catalog of the Drosophila immune cells was until recently limited to three major cell types, based on morphology, function and few molecular markers. Three recent single cell studies highlight the presence of several subgroups, revealing a large diversity in the molecular signature of the larval immune cells. Since these studies rely on somewhat different experimental and analytical approaches, we here compare the datasets and identify eight common, robust subgroups associated to distinct functions such as proliferation, immune response, phagocytosis or secretion. Similar comparative analyses with datasets from different stages and tissues disclose the presence of larval immune cells resembling embryonic hemocyte progenitors and the expression of specific properties in larval immune cells associated with peripheral tissues.
Collapse
Affiliation(s)
- Pierre B. Cattenoz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Sara Monticelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Alexia Pavlidaki
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| |
Collapse
|
19
|
Rodrigues D, Renaud Y, VijayRaghavan K, Waltzer L, Inamdar MS. Differential activation of JAK-STAT signaling reveals functional compartmentalization in Drosophila blood progenitors. eLife 2021; 10:61409. [PMID: 33594977 PMCID: PMC7920551 DOI: 10.7554/elife.61409] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Blood cells arise from diverse pools of stem and progenitor cells. Understanding progenitor heterogeneity is a major challenge. The Drosophila larval lymph gland is a well-studied model to understand blood progenitor maintenance and recapitulates several aspects of vertebrate hematopoiesis. However in-depth analysis has focused on the anterior lobe progenitors (AP), ignoring the posterior progenitors (PP) from the posterior lobes. Using in situ expression mapping and developmental and transcriptome analysis, we reveal PP heterogeneity and identify molecular-genetic tools to study this abundant progenitor population. Functional analysis shows that PP resist differentiation upon immune challenge, in a JAK-STAT-dependent manner. Upon wasp parasitism, AP downregulate JAK-STAT signaling and form lamellocytes. In contrast, we show that PP activate STAT92E and remain undifferentiated, promoting survival. Stat92E knockdown or genetically reducing JAK-STAT signaling permits PP lamellocyte differentiation. We discuss how heterogeneity and compartmentalization allow functional segregation in response to systemic cues and could be widely applicable.
Collapse
Affiliation(s)
- Diana Rodrigues
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.,National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Shanmugha Arts, Science, Technology & Research Academy, Tamil Nadu, India
| | - Yoan Renaud
- University of Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France
| | - K VijayRaghavan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Shanmugha Arts, Science, Technology & Research Academy, Tamil Nadu, India
| | - Lucas Waltzer
- University of Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France
| | - Maneesha S Inamdar
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
20
|
Madhwal S, Shin M, Kapoor A, Goyal M, Joshi MK, Ur Rehman PM, Gor K, Shim J, Mukherjee T. Metabolic control of cellular immune-competency by odors in Drosophila. eLife 2020; 9:60376. [PMID: 33372660 PMCID: PMC7808736 DOI: 10.7554/elife.60376] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022] Open
Abstract
Studies in different animal model systems have revealed the impact of odors on immune cells; however, any understanding on why and how odors control cellular immunity remained unclear. We find that Drosophila employ an olfactory-immune cross-talk to tune a specific cell type, the lamellocytes, from hematopoietic-progenitor cells. We show that neuronally released GABA derived upon olfactory stimulation is utilized by blood-progenitor cells as a metabolite and through its catabolism, these cells stabilize Sima/HIFα protein. Sima capacitates blood-progenitor cells with the ability to initiate lamellocyte differentiation. This systemic axis becomes relevant for larvae dwelling in wasp-infested environments where chances of infection are high. By co-opting the olfactory route, the preconditioned animals elevate their systemic GABA levels leading to the upregulation of blood-progenitor cell Sima expression. This elevates their immune-potential and primes them to respond rapidly when infected with parasitic wasps. The present work highlights the importance of the olfaction in immunity and shows how odor detection during animal development is utilized to establish a long-range axis in the control of blood-progenitor competency and immune-priming.
Collapse
Affiliation(s)
- Sukanya Madhwal
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Mingyu Shin
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Ankita Kapoor
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Manisha Goyal
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India.,The University of Trans-Disciplinary Health Sciences & Technology (TDU), Bengaluru, India
| | - Manish K Joshi
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India
| | | | - Kavan Gor
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India
| | - Jiwon Shim
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea.,Research Institute for Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Tina Mukherjee
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India
| |
Collapse
|
21
|
Nunes C, Sucena É, Koyama T. Endocrine regulation of immunity in insects. FEBS J 2020; 288:3928-3947. [PMID: 33021015 DOI: 10.1111/febs.15581] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Organisms have constant contact with potentially harmful agents that can compromise their fitness. However, most of the times these agents fail to cause serious disease by virtue of the rapid and efficient immune responses elicited in the host that can range from behavioural adaptations to immune system triggering. The immune system of insects does not comprise the adaptive arm, making it less complex than that of vertebrates, but key aspects of the activation and regulation of innate immunity are conserved across different phyla. This is the case for the hormonal regulation of immunity as a part of the broad organismal responses to external conditions under different internal states. In insects, depending on the physiological circumstances, distinct hormones either enhance or suppress the immune response integrating individual (and often collective) responses physiologically and behaviourally. In this review, we provide an overview of our current knowledge on the endocrine regulation of immunity in insects, its mechanisms and implications on metabolic adaptation and behaviour. We highlight the importance of this multilayered regulation of immunity in survival and reproduction (fitness) and its dependence on the hormonal integration with other mechanisms and life-history traits.
Collapse
Affiliation(s)
| | - Élio Sucena
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Moore R, Vogt K, Acosta-Martin AE, Shire P, Zeidler M, Smythe E. Integration of JAK/STAT receptor-ligand trafficking, signalling and gene expression in Drosophila melanogaster cells. J Cell Sci 2020; 133:jcs246199. [PMID: 32917740 DOI: 10.1242/jcs.246199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
The JAK/STAT pathway is an essential signalling cascade required for multiple processes during development and for adult homeostasis. A key question in understanding this pathway is how it is regulated in different cell contexts. Here, we have examined how endocytic processing contributes to signalling by the single cytokine receptor in Drosophila melanogaster cells, Domeless. We identify an evolutionarily conserved di-leucine (di-Leu) motif that is required for Domeless internalisation and show that endocytosis is required for activation of a subset of Domeless targets. Our data indicate that endocytosis both qualitatively and quantitatively regulates Domeless signalling. STAT92E, the single STAT transcription factor in Drosophila, appears to be the target of endocytic regulation, and our studies show that phosphorylation of STAT92E on Tyr704, although necessary, is not always sufficient for target transcription. Finally, we identify a conserved residue, Thr702, which is essential for Tyr704 phosphorylation. Taken together, our findings identify previously unknown aspects of JAK/STAT pathway regulation likely to play key roles in the spatial and temporal regulation of signalling in vivo.
Collapse
Affiliation(s)
- Rachel Moore
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Katja Vogt
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Adelina E Acosta-Martin
- biOMICS Facility, Faculty of Science Mass Spectrometry Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Patrick Shire
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Martin Zeidler
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Elizabeth Smythe
- Centre for Membrane Interactions and Dynamics, Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
23
|
Luo F, Yu S, Jin LH. The Posterior Signaling Center Is an Important Microenvironment for Homeostasis of the Drosophila Lymph Gland. Front Cell Dev Biol 2020; 8:382. [PMID: 32509789 PMCID: PMC7253591 DOI: 10.3389/fcell.2020.00382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/28/2020] [Indexed: 01/02/2023] Open
Abstract
Hematopoiesis is a necessary process for development and immune defense in Drosophila from the embryonic period to adulthood. There are two main stages in this process: the first stage occurs in the head mesoderm during the embryonic stage, and the second occurs in a specialized hematopoietic organ along the dorsal vessel, the lymph gland, during the larval stage. The lymph gland consists of paired lobes, each of which has distinct regions: the cortical zone (CZ), which contains mature hemocytes; the medullary zone (MZ), which contains hematopoietic progenitors; and the posterior signaling center (PSC), which specifically expresses the early B-cell factor (EBF) transcription factor Collier (Col) and the HOX factor Antennapedia (Antp) to form a microenvironment similar to that of the mammalian bone marrow hematopoietic stem cell niche. The PSC plays a key role in regulating hematopoietic progenitor differentiation. Moreover, the PSC contributes to the cellular immune response to wasp parasitism triggered by elevated ROS levels. Two recent studies have revealed that hematopoietic progenitor maintenance is directly regulated by Col expressed in the MZ and is independent of the PSC, challenging the traditional model. In this review, we summarize the regulatory networks of PSC cell proliferation, the controversy regarding PSC-mediated regulation of hematopoietic progenitor differentiation, and the wasp egg infection response. In addition, we discuss why the PSC is an ideal model for investigating mammalian hematopoietic stem cell niches and leukemia.
Collapse
Affiliation(s)
| | | | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| |
Collapse
|
24
|
Fu Y, Huang X, Zhang P, van de Leemput J, Han Z. Single-cell RNA sequencing identifies novel cell types in Drosophila blood. J Genet Genomics 2020; 47:175-186. [PMID: 32487456 DOI: 10.1016/j.jgg.2020.02.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/14/2022]
Abstract
Drosophila has been extensively used to model the human blood-immune system, as both systems share many developmental and immune response mechanisms. However, while many human blood cell types have been identified, only three were found in flies: plasmatocytes, crystal cells and lamellocytes. To better understand the complexity of fly blood system, we used single-cell RNA sequencing technology to generate comprehensive gene expression profiles for Drosophila circulating blood cells. In addition to the known cell types, we identified two new Drosophila blood cell types: thanacytes and primocytes. Thanacytes, which express many stimulus response genes, are involved in distinct responses to different types of bacteria. Primocytes, which express cell fate commitment and signaling genes, appear to be involved in keeping stem cells in the circulating blood. Furthermore, our data revealed four novel plasmatocyte subtypes (Ppn+, CAH7+, Lsp+ and reservoir plasmatocytes), each with unique molecular identities and distinct predicted functions. We also identified cross-species markers from Drosophila hemocytes to human blood cells. Our analysis unveiled a more complex Drosophila blood system and broadened the scope of using Drosophila to model human blood system in development and disease.
Collapse
Affiliation(s)
- Yulong Fu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Xiaohu Huang
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA; Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Peng Zhang
- Divisions of Immunotherapy, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA; Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA; Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
25
|
Paddibhatla I, Gautam DK, Mishra RK. SETDB1 modulates the differentiation of both the crystal cells and the lamellocytes in Drosophila. Dev Biol 2019; 456:74-85. [DOI: 10.1016/j.ydbio.2019.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 01/10/2023]
|
26
|
Kim-Jo C, Gatti JL, Poirié M. Drosophila Cellular Immunity Against Parasitoid Wasps: A Complex and Time-Dependent Process. Front Physiol 2019; 10:603. [PMID: 31156469 PMCID: PMC6529592 DOI: 10.3389/fphys.2019.00603] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/29/2019] [Indexed: 11/13/2022] Open
Abstract
Host-parasitoid interactions are among the most studied interactions between invertebrates because of their fundamental interest - the evolution of original traits in parasitoids - and applied, parasitoids being widely used in biological control. Immunity, and in particular cellular immunity, is central in these interactions, the host encapsulation response being specific for large foreign bodies such as parasitoid eggs. Although already well studied in this species, recent data on Drosophila melanogaster have unquestionably improved knowledge of invertebrate cellular immunity. At the same time, the venomics of parasitoids has expanded, notably those of Drosophila. Here, we summarize and discuss these advances, with a focus on an emerging "time-dependent" view of interactions outcome at the intra- and interspecific level. We also present issues still in debate and prospects for study. Data on the Drosophila-parasitoid model paves the way to new concepts in insect immunity as well as parasitoid wasp strategies to overcome it.
Collapse
Affiliation(s)
| | | | - Marylène Poirié
- INRA, CNRS, Institut Sophia Agrobiotech, Université Côte d’Azur, Sophia Antipolis, France
| |
Collapse
|
27
|
Powers N, Srivastava A. JAK/STAT signaling is involved in air sac primordium development of Drosophila melanogaster. FEBS Lett 2019; 593:658-669. [PMID: 30854626 DOI: 10.1002/1873-3468.13355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/17/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022]
Abstract
The dorsal thoracic air sacs in fruit flies (Drosophila melanogaster) are functionally and developmentally comparable to human lungs. The progenitors of these structures, air sac primordia (ASPs), invasively propagate into wing imaginal disks, employing mechanisms similar to those that promote metastasis in malignant tumors. We investigated whether Janus kinase/signal transducer and activator of transcription JAK/STAT signaling plays a role in the directed morphogenesis of ASPs. We find that JAK/STAT signaling occurs in ASP tip cells and misexpression of core components in the JAK/STAT signaling cascade significantly impedes ASP development. We further identify Upd2 as an activating ligand for JAK/STAT activity in the ASP. Together, these data constitute a considerable step forward in understanding the role of JAK/STAT signaling in ASPs and similar structures in mammalian models.
Collapse
Affiliation(s)
- Nathan Powers
- Department of Biology and Biotechnology Center, Western Kentucky University, Bowling Green, KY, USA
| | - Ajay Srivastava
- Department of Biology and Biotechnology Center, Western Kentucky University, Bowling Green, KY, USA
| |
Collapse
|
28
|
Headcase is a Repressor of Lamellocyte Fate in Drosophila melanogaster. Genes (Basel) 2019; 10:genes10030173. [PMID: 30841641 PMCID: PMC6470581 DOI: 10.3390/genes10030173] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 01/12/2023] Open
Abstract
Due to the evolutionary conservation of the regulation of hematopoiesis, Drosophila provides an excellent model organism to study blood cell differentiation and hematopoietic stem cell (HSC) maintenance. The larvae of Drosophila melanogaster respond to immune induction with the production of special effector blood cells, the lamellocytes, which encapsulate and subsequently kill the invader. Lamellocytes differentiate as a result of a concerted action of all three hematopoietic compartments of the larva: the lymph gland, the circulating hemocytes, and the sessile tissue. Within the lymph gland, the communication of the functional zones, the maintenance of HSC fate, and the differentiation of effector blood cells are regulated by a complex network of signaling pathways. Applying gene conversion, mutational analysis, and a candidate based genetic interaction screen, we investigated the role of Headcase (Hdc), the homolog of the tumor suppressor HECA in the hematopoiesis of Drosophila. We found that naive loss-of-function hdc mutant larvae produce lamellocytes, showing that Hdc has a repressive role in effector blood cell differentiation. We demonstrate that hdc genetically interacts with the Hedgehog and the Decapentaplegic pathways in the hematopoietic niche of the lymph gland. By adding further details to the model of blood cell fate regulation in the lymph gland of the larva, our findings contribute to the better understanding of HSC maintenance.
Collapse
|
29
|
Banerjee U, Girard JR, Goins LM, Spratford CM. Drosophila as a Genetic Model for Hematopoiesis. Genetics 2019; 211:367-417. [PMID: 30733377 PMCID: PMC6366919 DOI: 10.1534/genetics.118.300223] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
In this FlyBook chapter, we present a survey of the current literature on the development of the hematopoietic system in Drosophila The Drosophila blood system consists entirely of cells that function in innate immunity, tissue integrity, wound healing, and various forms of stress response, and are therefore functionally similar to myeloid cells in mammals. The primary cell types are specialized for phagocytic, melanization, and encapsulation functions. As in mammalian systems, multiple sites of hematopoiesis are evident in Drosophila and the mechanisms involved in this process employ many of the same molecular strategies that exemplify blood development in humans. Drosophila blood progenitors respond to internal and external stress by coopting developmental pathways that involve both local and systemic signals. An important goal of these Drosophila studies is to develop the tools and mechanisms critical to further our understanding of human hematopoiesis during homeostasis and dysfunction.
Collapse
Affiliation(s)
- Utpal Banerjee
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
- Molecular Biology Institute, University of California, Los Angeles, California 90095
- Department of Biological Chemistry, University of California, Los Angeles, California 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| | - Juliet R Girard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Lauren M Goins
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Carrie M Spratford
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| |
Collapse
|
30
|
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: 53] [Impact Index Per Article: 7.6] [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
|
31
|
Bazzi W, Cattenoz PB, Delaporte C, Dasari V, Sakr R, Yuasa Y, Giangrande A. Embryonic hematopoiesis modulates the inflammatory response and larval hematopoiesis in Drosophila. eLife 2018; 7:e34890. [PMID: 29992900 PMCID: PMC6040882 DOI: 10.7554/elife.34890] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/18/2018] [Indexed: 11/25/2022] Open
Abstract
Recent lineage tracing analyses have significantly improved our understanding of immune system development and highlighted the importance of the different hematopoietic waves. The current challenge is to understand whether these waves interact and whether this affects the function of the immune system. Here we report a molecular pathway regulating the immune response and involving the communication between embryonic and larval hematopoietic waves in Drosophila. Down-regulating the transcription factor Gcm specific to embryonic hematopoiesis enhances the larval phenotypes induced by over-expressing the pro-inflammatory Jak/Stat pathway or by wasp infestation. Gcm works by modulating the transduction of the Upd cytokines to the site of larval hematopoiesis and hence the response to chronic (Jak/Stat over-expression) and acute (wasp infestation) immune challenges. Thus, homeostatic interactions control the function of the immune system in physiology and pathology. Our data also indicate that a transiently expressed developmental pathway has a long-lasting effect on the immune response.
Collapse
Affiliation(s)
- Wael Bazzi
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Pierre B Cattenoz
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Claude Delaporte
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Vasanthi Dasari
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Rosy Sakr
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Yoshihiro Yuasa
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| |
Collapse
|
32
|
Fisher KH, Fragiadaki M, Pugazhendhi D, Bausek N, Arredondo MA, Thomas SJ, Brown S, Zeidler MP. A genome-wide RNAi screen identifies MASK as a positive regulator of cytokine receptor stability. J Cell Sci 2018; 131:jcs.209551. [PMID: 29848658 DOI: 10.1242/jcs.209551] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 05/16/2018] [Indexed: 01/01/2023] Open
Abstract
Cytokine receptors often act via the Janus kinase and signal transducer and activator of transcription (JAK/STAT) pathway to form a signalling cascade that is essential for processes such as haematopoiesis, immune responses and tissue homeostasis. In order to transduce ligand activation, cytokine receptors must dimerise. However, mechanisms regulating their dimerisation are poorly understood. In order to better understand the processes regulating cytokine receptor levels, and their activity and dimerisation, we analysed the highly conserved JAK/STAT pathway in Drosophila, which acts via a single receptor, known as Domeless. We performed a genome-wide RNAi screen in Drosophila cells, identifying MASK as a positive regulator of Domeless dimerisation and protein levels. We show that MASK is able to regulate receptor levels and JAK/STAT signalling both in vitro and in vivo We also show that its human homologue, ANKHD1, is also able to regulate JAK/STAT signalling and the levels of a subset of pathway receptors in human cells. Taken together, our results identify MASK as a novel regulator of cytokine receptor levels, and suggest functional conservation, which may have implications for human health.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Katherine H Fisher
- The Bateson Centre, Department of Biomedical Science, The University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| | - Maria Fragiadaki
- The Bateson Centre, Department of Biomedical Science, The University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| | - Dhamayanthi Pugazhendhi
- The Bateson Centre, Department of Biomedical Science, The University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| | - Nina Bausek
- The Bateson Centre, Department of Biomedical Science, The University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| | - Maria A Arredondo
- Department of Oncology & Human Metabolism, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Sally J Thomas
- Department of Oncology & Human Metabolism, The University of Sheffield, Sheffield, S10 2RX, UK
| | - Stephen Brown
- The Sheffield RNAi Screening Facility, Department of Biomedical Science, The University of Sheffield, S10 2TN, UK
| | - Martin P Zeidler
- The Bateson Centre, Department of Biomedical Science, The University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| |
Collapse
|
33
|
Yu S, Luo F, Jin LH. The Drosophila lymph gland is an ideal model for studying hematopoiesis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:60-69. [PMID: 29191551 DOI: 10.1016/j.dci.2017.11.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/30/2017] [Accepted: 11/26/2017] [Indexed: 06/07/2023]
Abstract
Hematopoiesis in Drosophila melanogaster occurs throughout the entire life cycle, from the embryo to adulthood. The healthy lymph gland, as a hematopoietic organ during the larval stage, can give rise to two mature types of hemocytes, plasmatocytes and crystal cells, which persist into the pupal and adult stages. Homeostasis of the lymph gland is tightly controlled by a series of conserved factors and signaling pathways, which also play key roles in mammalian hematopoiesis. Thus, revealing the hematopoietic mechanisms in Drosophila will advance our understanding of hematopoietic stem cells and their niche as well as leukemia in mammals. In addition, the lymph gland employs a battery of strategies to produce lamellocytes, another type of mature hemocyte, to fight against parasitic wasp eggs, making the lymph gland an important immunological organ. In this review, the developmental process of the lymph gland and the regulatory networks of hematopoiesis are summarized. Moreover, we outline the current knowledge and novel insight into homeostasis of the lymph gland.
Collapse
Affiliation(s)
- Shichao Yu
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Fangzhou Luo
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China.
| |
Collapse
|
34
|
The human Smoothened inhibitor PF-04449913 induces exit from quiescence and loss of multipotent Drosophila hematopoietic progenitor cells. Oncotarget 2018; 7:55313-55327. [PMID: 27486815 PMCID: PMC5342419 DOI: 10.18632/oncotarget.10879] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/26/2016] [Indexed: 11/25/2022] Open
Abstract
The efficient treatment of hematological malignancies as Acute Myeloid Leukemia, myelofibrosis and Chronic Myeloid Leukemia, requires the elimination of cancer-initiating cells and the prevention of disease relapse through targeting pathways that stimulate generation and maintenance of these cells. In mammals, inhibition of Smoothened, the key mediator of the Hedgehog signaling pathway, reduces Chronic Myeloid Leukemia progression and propagation. These findings make Smo a candidate target to inhibit maintenance of leukemia-initiating cells. In Drosophila melanogaster the same pathway maintains the hematopoietic precursor cells of the lymph gland, the hematopoietic organ that develops in the larva. Using Drosophila as an in vivo model, we investigated the mode of action of PF-04449913, a small-molecule inhibitor of the human Smo protein. Drosophila larvae fed with PF-04449913 showed traits of altered hematopoietic homeostasis. These include the development of melanotic nodules, increase of circulating hemocytes, the size increase of the lymph gland and accelerated differentiation of blood cells likely due to the exit of multi-potent precursors from quiescence. Importantly, the Smo inhibition can lead to the complete loss of hematopoietic precursors. We conclude that PF-04449913 inhibits Drosophila Smo blocking the Hh signaling pathway and causing the loss of hematopoietic precursor cells. Interestingly, this is the effect expected in patients treated with PF-04449913: number decrease of cancer initiating cells in the bone marrow to reduce the risk of leukemia relapse. Altogether our results indicate that Drosophila comprises a model system for the in vivo study of molecules that target evolutionary conserved pathways implicated in human hematological malignancies.
Collapse
|
35
|
Louradour I, Sharma A, Morin-Poulard I, Letourneau M, Vincent A, Crozatier M, Vanzo N. Reactive oxygen species-dependent Toll/NF-κB activation in the Drosophila hematopoietic niche confers resistance to wasp parasitism. eLife 2017; 6:25496. [PMID: 29091025 PMCID: PMC5681226 DOI: 10.7554/elife.25496] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem/progenitor cells in the adult mammalian bone marrow ensure blood cell renewal. Their cellular microenvironment, called 'niche', regulates hematopoiesis both under homeostatic and immune stress conditions. In the Drosophila hematopoietic organ, the lymph gland, the posterior signaling center (PSC) acts as a niche to regulate the hematopoietic response to immune stress such as wasp parasitism. This response relies on the differentiation of lamellocytes, a cryptic cell type, dedicated to pathogen encapsulation and killing. Here, we establish that Toll/NF-κB pathway activation in the PSC in response to wasp parasitism non-cell autonomously induces the lymph gland immune response. Our data further establish a regulatory network where co-activation of Toll/NF-κB and EGFR signaling by ROS levels in the PSC/niche controls lymph gland hematopoiesis under parasitism. Whether a similar regulatory network operates in mammals to control emergency hematopoiesis is an open question.
Collapse
Affiliation(s)
- Isabelle Louradour
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Anurag Sharma
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Ismael Morin-Poulard
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Manon Letourneau
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Alain Vincent
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Vanzo
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
36
|
Sun JJ, Lan JF, Zhao XF, Vasta GR, Wang JX. Binding of a C-type lectin's coiled-coil domain to the Domeless receptor directly activates the JAK/STAT pathway in the shrimp immune response to bacterial infection. PLoS Pathog 2017; 13:e1006626. [PMID: 28931061 PMCID: PMC5645147 DOI: 10.1371/journal.ppat.1006626] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 10/17/2017] [Accepted: 09/03/2017] [Indexed: 11/28/2022] Open
Abstract
C-type lectins (CTLs) are characterized by the presence of a C-type carbohydrate recognition domain (CTLD) that by recognizing microbial glycans, is responsible for their roles as pattern recognition receptors in the immune response to bacterial infection. In addition to the CTLD, however, some CTLs display additional domains that can carry out effector functions, such as the collagenous domain of the mannose-binding lectin. While in vertebrates, the mechanisms involved in these effector functions have been characterized in considerable detail, in invertebrates they remain poorly understood. In this study, we identified in the kuruma shrimp (Marsupenaeus japonicus) a structurally novel CTL (MjCC-CL) that in addition to the canonical CTLD, contains a coiled-coil domain (CCD) responsible for the effector functions that are key to the shrimp's antibacterial response mediated by antimicrobial peptides (AMPs). By the use of in vitro and in vivo experimental approaches we elucidated the mechanism by which the recognition of bacterial glycans by the CTLD of MjCC-CL leads to activation of the JAK/STAT pathway via interaction of the CCD with the surface receptor Domeless, and upregulation of AMP expression. Thus, our study of the shrimp MjCC-CL revealed a striking functional difference with vertebrates, in which the JAK/STAT pathway is indirectly activated by cell death and stress signals through cytokines or growth factors. Instead, by cross-linking microbial pathogens with the cell surface receptor Domeless, a lectin directly activates the JAK/STAT pathway, which plays a central role in the shrimp antibacterial immune responses by upregulating expression of selected AMPs.
Collapse
Affiliation(s)
- Jie-Jie Sun
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China
| | - Jiang-Feng Lan
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China
| | - Gerardo R. Vasta
- Department of Microbiology and Immunology, School of Medicine, University of Maryland Baltimore and Institute of Marine and Environmental Technology, Baltimore, Maryland, United States of America
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
37
|
Regulation of phagocyte triglyceride by a STAT-ATG2 pathway controls mycobacterial infection. Nat Commun 2017; 8:14642. [PMID: 28262681 PMCID: PMC5343520 DOI: 10.1038/ncomms14642] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/18/2017] [Indexed: 01/27/2023] Open
Abstract
Mycobacterium tuberculosis remains a global threat to human health, yet the molecular mechanisms regulating immunity remain poorly understood. Cytokines can promote or inhibit mycobacterial survival inside macrophages and the underlying mechanisms represent potential targets for host-directed therapies. Here we show that cytokine-STAT signalling promotes mycobacterial survival within macrophages by deregulating lipid droplets via ATG2 repression. In Drosophila infected with Mycobacterium marinum, mycobacterium-induced STAT activity triggered by unpaired-family cytokines reduces Atg2 expression, permitting deregulation of lipid droplets. Increased Atg2 expression or reduced macrophage triglyceride biosynthesis, normalizes lipid deposition in infected phagocytes and reduces numbers of viable intracellular mycobacteria. In human macrophages, addition of IL-6 promotes mycobacterial survival and BCG-induced lipid accumulation by a similar, but probably not identical, mechanism. Our results reveal Atg2 regulation as a mechanism by which cytokines can control lipid droplet homeostasis and consequently resistance to mycobacterial infection in Drosophila. Cytokines and their associated pathways can affect survival of Mycobacterium tuberculosis in macrophages, representing potential targets for host-directed therapies. Here, Péan et al. show that cytokine-STAT signalling promotes mycobacterial survival within macrophages by deregulating lipid droplet homeostasis.
Collapse
|
38
|
Terriente-Félix A, Pérez L, Bray SJ, Nebreda AR, Milán M. A Drosophila model of myeloproliferative neoplasm reveals a feed-forward loop in the JAK pathway mediated by p38 MAPK signalling. Dis Model Mech 2017; 10:399-407. [PMID: 28237966 PMCID: PMC5399568 DOI: 10.1242/dmm.028118] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/01/2017] [Indexed: 12/25/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) of the Philadelphia-negative class comprise polycythaemia vera, essential thrombocythaemia and primary myelofibrosis (PMF). They are associated with aberrant numbers of myeloid lineage cells in the blood, and in the case of overt PMF, with development of myelofibrosis in the bone marrow and failure to produce normal blood cells. These diseases are usually caused by gain-of-function mutations in the kinase JAK2. Here, we use Drosophila to investigate the consequences of activation of the JAK2 orthologue in haematopoiesis. We have identified maturing haemocytes in the lymph gland, the major haematopoietic organ in the fly, as the cell population susceptible to induce hypertrophy upon targeted overexpression of JAK. We show that JAK activates a feed-forward loop, including the cytokine-like ligand Upd3 and its receptor, Domeless, which are required to induce lymph gland hypertrophy. Moreover, we present evidence that p38 MAPK signalling plays a key role in this process by inducing expression of the ligand Upd3. Interestingly, we also show that forced activation of the p38 MAPK pathway in maturing haemocytes suffices to generate hypertrophic organs and the appearance of melanotic tumours. Our results illustrate a novel pro-tumourigenic crosstalk between the p38 MAPK pathway and JAK signalling in a Drosophila model of MPNs. Based on the shared molecular mechanisms underlying MPNs in flies and humans, the interplay between Drosophila JAK and p38 signalling pathways unravelled in this work might have translational relevance for human MPNs. Summary: Pro-tumourigenic crosstalk occurs between the p38 MAPK pathway and JAK signalling in a Drosophila model of myeloproliferative neoplasm.
Collapse
Affiliation(s)
- Ana Terriente-Félix
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Lidia Pérez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Sarah J Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain .,ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain
| | - Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain .,ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain
| |
Collapse
|
39
|
Letourneau M, Lapraz F, Sharma A, Vanzo N, Waltzer L, Crozatier M. Drosophila hematopoiesis under normal conditions and in response to immune stress. FEBS Lett 2016; 590:4034-4051. [PMID: 27455465 DOI: 10.1002/1873-3468.12327] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/07/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
The emergence of hematopoietic progenitors and their differentiation into various highly specialized blood cell types constitute a finely tuned process. Unveiling the genetic cascades that control blood cell progenitor fate and understanding how they are modulated in response to environmental changes are two major challenges in the field of hematopoiesis. In the last 20 years, many studies have established important functional analogies between blood cell development in vertebrates and in the fruit fly, Drosophila melanogaster. Thereby, Drosophila has emerged as a powerful genetic model for studying mechanisms that control hematopoiesis during normal development or in pathological situations. Moreover, recent advances in Drosophila have highlighted how intricate cell communication networks and microenvironmental cues regulate blood cell homeostasis. They have also revealed the striking plasticity of Drosophila mature blood cells and the presence of different sites of hematopoiesis in the larva. This review provides an overview of Drosophila hematopoiesis during development and summarizes our current knowledge on the molecular processes controlling larval hematopoiesis, both under normal conditions and in response to an immune challenge, such as wasp parasitism.
Collapse
Affiliation(s)
- Manon Letourneau
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Francois Lapraz
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Anurag Sharma
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France.,Department of Biomedical Sciences, NU Centre for Science Education & Research, Nitte University, Mangalore-18, India
| | - Nathalie Vanzo
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Lucas Waltzer
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| |
Collapse
|
40
|
Wu M, Wu Y, Lan T, Jiang L, Qian H, Chen Y. Type II cGMP-dependent protein kinase inhibits EGF-induced JAK/STAT signaling in gastric cancer cells. Mol Med Rep 2016; 14:1849-56. [DOI: 10.3892/mmr.2016.5452] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 06/07/2016] [Indexed: 11/06/2022] Open
|
41
|
Oyallon J, Vanzo N, Krzemień J, Morin-Poulard I, Vincent A, Crozatier M. Two Independent Functions of Collier/Early B Cell Factor in the Control of Drosophila Blood Cell Homeostasis. PLoS One 2016; 11:e0148978. [PMID: 26866694 PMCID: PMC4750865 DOI: 10.1371/journal.pone.0148978] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/26/2016] [Indexed: 11/18/2022] Open
Abstract
Blood cell production in the Drosophila hematopoietic organ, the lymph gland, is controlled by intrinsic factors and extrinsic signals. Initial analysis of Collier/Early B Cell Factor function in the lymph gland revealed the role of the Posterior Signaling Center (PSC) in mounting a dedicated cellular immune response to wasp parasitism. Further, premature blood cell differentiation when PSC specification or signaling was impaired, led to assigning the PSC a role equivalent to the vertebrate hematopoietic niche. We report here that Collier is expressed in a core population of lymph gland progenitors and cell autonomously maintains this population. The PSC contributes to lymph gland homeostasis by regulating blood cell differentiation, rather than by maintaining core progenitors. In addition to PSC signaling, switching off Collier expression in progenitors is required for efficient immune response to parasitism. Our data show that two independent sites of Collier/Early B Cell Factor expression, hematopoietic progenitors and the PSC, achieve control of hematopoiesis.
Collapse
Affiliation(s)
- Justine Oyallon
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), University Paul Sabatier (UPS), CNRS, 118 Route de Narbonne 31062 Toulouse cedex 09, France
| | - Nathalie Vanzo
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), University Paul Sabatier (UPS), CNRS, 118 Route de Narbonne 31062 Toulouse cedex 09, France
| | - Joanna Krzemień
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), University Paul Sabatier (UPS), CNRS, 118 Route de Narbonne 31062 Toulouse cedex 09, France
| | - Ismaël Morin-Poulard
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), University Paul Sabatier (UPS), CNRS, 118 Route de Narbonne 31062 Toulouse cedex 09, France
| | - Alain Vincent
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), University Paul Sabatier (UPS), CNRS, 118 Route de Narbonne 31062 Toulouse cedex 09, France
| | - Michèle Crozatier
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), University Paul Sabatier (UPS), CNRS, 118 Route de Narbonne 31062 Toulouse cedex 09, France
| |
Collapse
|
42
|
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
|
43
|
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
|
44
|
Yang H, Kronhamn J, Ekström JO, Korkut GG, Hultmark D. JAK/STAT signaling in Drosophila muscles controls the cellular immune response against parasitoid infection. EMBO Rep 2015; 16:1664-72. [PMID: 26412855 PMCID: PMC4687419 DOI: 10.15252/embr.201540277] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 08/28/2015] [Indexed: 11/09/2022] Open
Abstract
The role of JAK/STAT signaling in the cellular immune response of Drosophila is not well understood. Here, we show that parasitoid wasp infection activates JAK/STAT signaling in somatic muscles of the Drosophila larva, triggered by secretion of the cytokines Upd2 and Upd3 from circulating hemocytes. Deletion of upd2 or upd3, but not the related os (upd1) gene, reduced the cellular immune response, and suppression of the JAK/STAT pathway in muscle cells reduced the encapsulation of wasp eggs and the number of circulating lamellocyte effector cells. These results suggest that JAK/STAT signaling in muscles participates in a systemic immune defense against wasp infection.
Collapse
Affiliation(s)
- Hairu Yang
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jesper Kronhamn
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jens-Ola Ekström
- Department of Molecular Biology, Umeå University, Umeå, Sweden Institute of Biomedical Technology BMT Tampere University, Tampere, Finland
| | | | - Dan Hultmark
- Department of Molecular Biology, Umeå University, Umeå, Sweden Institute of Biomedical Technology BMT Tampere University, Tampere, Finland
| |
Collapse
|
45
|
Bajgar A, Kucerova K, Jonatova L, Tomcala A, Schneedorferova I, Okrouhlik J, Dolezal T. Extracellular adenosine mediates a systemic metabolic switch during immune response. PLoS Biol 2015; 13:e1002135. [PMID: 25915062 PMCID: PMC4411001 DOI: 10.1371/journal.pbio.1002135] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/18/2015] [Indexed: 12/20/2022] Open
Abstract
Immune defense is energetically costly, and thus an effective response requires metabolic adaptation of the organism to reallocate energy from storage, growth, and development towards the immune system. We employ the natural infection of Drosophila with a parasitoid wasp to study energy regulation during immune response. To combat the invasion, the host must produce specialized immune cells (lamellocytes) that destroy the parasitoid egg. We show that a significant portion of nutrients are allocated to differentiating lamellocytes when they would otherwise be used for development. This systemic metabolic switch is mediated by extracellular adenosine released from immune cells. The switch is crucial for an effective immune response. Preventing adenosine transport from immune cells or blocking adenosine receptor precludes the metabolic switch and the deceleration of development, dramatically reducing host resistance. Adenosine thus serves as a signal that the “selfish” immune cells send during infection to secure more energy at the expense of other tissues. A study of the fruit fly's response to parasitoid wasp eggs reveals that immune cells selfishly release adenosine as a signal to trigger a systemic metabolic switch, thereby suppressing nonimmune processes and securing energy and nutrients for immune activity. Read the Primer. The immune response is energetically costly and often requires adaption of the whole organism to ensure it receives enough energy. It is not well understood how distribution of energy resources within the organism is regulated during an immune response. To understand this better, we used parasitoid wasp infection of fruit fly larvae—the host larvae have 48 h before they pupate to destroy the infecting “alien” or face destruction by the parasitoid that will consume the developing pupa. Here we find a signal, generated by the host immune cells, which mediates a systemic energy switch. This signal—adenosine—suppresses processes driving larval to pupal development of the host, thereby freeing up energy for the immune system. We show that the resulting developmental delay in the fruit fly larvae is crucial for an efficient immune response; without the adenosine signal, resistance to the parasitoid drops drastically. Generation of this signal by immune cells demonstrates that in response to external stressors, the immune system can mobilize reallocation to itself of energy and nutrients from the rest of the organism.
Collapse
Affiliation(s)
- Adam Bajgar
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czech Republic
| | - Katerina Kucerova
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czech Republic
| | - Lucie Jonatova
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czech Republic
| | - Ales Tomcala
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Ceske Budejovice, Czech Republic
| | - Ivana Schneedorferova
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czech Republic
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Ceske Budejovice, Czech Republic
| | - Jan Okrouhlik
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czech Republic
| | - Tomas Dolezal
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czech Republic
- * E-mail:
| |
Collapse
|
46
|
Chen XY, Yang YS, Chen K, Chen LS, Xie WR, Wang H. JAK-STAT signaling pathway and acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2015; 23:932-937. [DOI: 10.11569/wcjd.v23.i6.932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of acute pancreatitis has long been an important research topic. In acute pancreatitis, cytokines and growth factors bind to Janus kinase (JAK) related receptors, and activate JAKs. The activated JAKs phosphorylate the tyrosine residues of the receptor. The downstream signal transducers and activators of transcription (STAT) then bind to the specific site of the phosphorylated JAK receptor complexes, leading to the activation of STATs. The activated STATs detach from the receptor complexes and translocate to the nucleus to regulate the expression of Bcl-2, Bcl-X(L), Mcl-1 and other genes, thereby participating in the pathogenesis of pancreatitis. Such signal transduction can be terminated by the dephosphorylation of STATs. At present, more and more clinical experiments and animal studies have shown that the JAK-STAT pathway is closely related with acute pancreatitis. In this article, we will review the structure, distribution, and function of JAK-STAT signaling pathway as well as the role of JAK-STAT signaling pathway in the pathogenesis of acute pancreatitis.
Collapse
|
47
|
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
|
48
|
Amoyel M, Bach EA. Functions of the Drosophila JAK-STAT pathway: Lessons from stem cells. JAKSTAT 2014; 1:176-83. [PMID: 24058767 PMCID: PMC3670241 DOI: 10.4161/jkst.21621] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 07/13/2012] [Accepted: 07/25/2012] [Indexed: 01/06/2023] Open
Abstract
JAK-STAT signaling has been proposed to act in numerous stem cells in a variety of organisms. Here we provide an overview of its roles in three well characterized stem cell populations in Drosophila, in the intestine, lymph gland and testis. In flies, there is a single JAK and a single STAT, which has made the genetic dissection of pathway function considerably easier and facilitated the analysis of communication between stem cells, their niches and offspring. Studies in flies have revealed roles for this pathway as diverse as regulating bona fide intrinsic self-renewal, integrating response to environmental cues that control quiescence and promoting mitogenic responses to stress.
Collapse
Affiliation(s)
- Marc Amoyel
- Department of Biochemistry and Molecular Pharmacology; New York University School of Medicine; New York, NY USA
| | | |
Collapse
|
49
|
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
|
50
|
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
|