1
|
Qin B, Xue H, Wang X, Kim H, Jin LH. Atg2 controls Drosophila hematopoiesis through the PVR/TOR signaling pathways. FEBS J 2025; 292:294-312. [PMID: 39513270 DOI: 10.1111/febs.17288] [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: 11/05/2023] [Revised: 06/01/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024]
Abstract
The hematopoietic system of Drosophila is a well-established genetic model for studying hematopoiesis mechanisms, which are strictly regulated by multiple signaling pathways. Autophagy-related 2 (Atg2) protein is involved in autophagosome formation through its lipid transfer function; however, other functions in animal development, especially the role of Atg2 in maintaining hematopoietic homeostasis, are unclear. Here, we show that Atg2 knockdown in the cortical zone (CZ) induced the proliferation and differentiation of mature plasmatocytes and disrupted progenitor maintenance in the medullary zone (MZ). We also observed the differentiation of lamellocytes among circulating hemocytes and in the lymph gland, which is rarely observed in healthy larvae. The above results on hematopoiesis disorders are due to Atg2 regulating the Drosophila PDGF/VEGF receptor (PVR) and target of rapamycin (TOR) in the CZ of lymph gland. In conclusion, we identified Atg2 as a previously undescribed regulator of hematopoiesis. Understanding the mechanism of maintenance of hematopoietic homeostasis in Drosophila will help us better evaluate human blood disorder-related diseases.
Collapse
Affiliation(s)
- Bo Qin
- College of Life Sciences, Northeast Forestry University, Harbin, China
- Institute of Crop Breeding, Heilongjiang Academy of Agricultural Sciences, Harbin, China
| | - Hongmei Xue
- Peking University People's Hospital, Qingdao, China
- Women and Children's Hospital, Qingdao University, China
| | - Xiaoran Wang
- College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Hyonil Kim
- College of Life Sciences, Northeast Forestry University, Harbin, China
- College of Life Science, Kim Il Sung University, Pyongyang, Korea
| | - Li Hua Jin
- College of Life Sciences, Northeast Forestry University, Harbin, China
| |
Collapse
|
2
|
Luo F, Sui L, Sun Y, Lai Z, Zhang C, Zhang G, Bi B, Yu S, Jin LH. Rab1 and Syntaxin 17 regulate hematopoietic homeostasis through β-integrin trafficking in Drosophila. J Genet Genomics 2025; 52:51-65. [PMID: 39542172 DOI: 10.1016/j.jgg.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Hematopoiesis is crucial for organismal health, and Drosophila serves as an effective genetic model due to conserved regulatory mechanisms with vertebrates. In larvae, hematopoiesis primarily occurs in the lymph gland, which contains distinct zones, including the cortical zone, intermediate zone, medullary zone, and posterior signaling center (PSC). Rab1 is vital for membrane trafficking and maintaining the localization of cell adhesion molecules, yet its role in hematopoietic homeostasis is not fully understood. This study investigates the effects of Rab1 dysfunction on β-integrin trafficking within circulating hemocytes and lymph gland cells. Rab1 impairment disrupts the endosomal trafficking of β-integrin, leading to its abnormal localization on cell membranes, which promotes lamellocyte differentiation and alters progenitor dynamics in circulating hemocytes and lymph glands, respectively. We also show that the mislocalization of β-integrin is dependent on the adhesion protein DE-cadherin. The reduction of β-integrin at cell boundaries in PSC cells leads to fewer PSC cells and lamellocyte differentiation. Furthermore, Rab1 regulates the trafficking of β-integrin via the Q-SNARE protein Syntaxin 17 (Syx17). Our findings indicate that Rab1 and Syx17 regulate distinct trafficking pathways for β-integrin in different hematopoietic compartments and maintain hematopoietic homeostasis of Drosophila.
Collapse
Affiliation(s)
- Fangzhou Luo
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Luwei Sui
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Ying Sun
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Zhixian Lai
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Chengcheng Zhang
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Gaoqun Zhang
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Bing Bi
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Shichao Yu
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China.
| | - Li Hua Jin
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China.
| |
Collapse
|
3
|
Ho KYL, Ou AYJ, Samuelson N, Tanentzapf G. Novel features of Drosophila hematopoiesis uncovered by long-term live imaging. Dev Biol 2025; 517:286-300. [PMID: 39536928 DOI: 10.1016/j.ydbio.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Stem cells are subject to continuous regulation to ensure that the correct balance between stem cell differentiation and self-renewal is maintained. The dynamic and ongoing nature of stem cell regulation, as well as the complex signaling microenvironment in which stem cells are typically found, means that studying them in their endogenous environment in real time has multiple advantages over static fixed-sample approaches. We recently described a method for long-term, ex-vivo, live imaging of the blood progenitors in the Drosophila larval hematopoietic organ, the Lymph Gland (LG). This methodology has allowed us to analyze multiple aspects of fly hematopoiesis, in real time, in a manner that could not be carried out previously. Here, we describe novel insights derived from our quantitative live imaging approach. These insights include: the identification of extensive filopodia in the progenitors and description of their morphology and dynamics; visualization and quantitative analysis of JAK/STAT signaling in progenitors by the simultaneous tracking of thousands of vesicles containing internalized Domeless receptors; quantitative analysis of the location, morphology, and dynamics of mitochondria in blood progenitors; long-term tracking of patterns of cell division and migration of mature blood cell in the LG; long-term tracking of multiple cell behaviors in the distal committed progenitors; analysis of Ca2+ signaling of blood progenitors in the secondary lobes of the LG. Together, these observations illustrate the power of imaging fly hematopoiesis in real time and identify many previously undescribed processes and behaviors in the LG that are likely to play important roles in the regulation of progenitor differentiation and self-renewal.
Collapse
Affiliation(s)
- Kevin Y L Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Annie Y J Ou
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada; School of Kinesiology, University of British Columbia, Vancouver, V6T 1Z1, Canada; Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Nicholas Samuelson
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
4
|
Kharrat B, Gábor E, Virág N, Sinka R, Jankovics F, Kristó I, Vilmos P, Csordás G, Honti V. Dual role for Headcase in hemocyte progenitor fate determination in Drosophila melanogaster. PLoS Genet 2024; 20:e1011448. [PMID: 39466810 PMCID: PMC11515969 DOI: 10.1371/journal.pgen.1011448] [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: 03/11/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
The hematopoietic organ of the Drosophila larva, the lymph gland, is a simplified representation of mammalian hematopoietic compartments, with the presence of hemocyte progenitors in the medullary zone (MZ), differentiated hemocytes in the cortical zone (CZ), and a hematopoietic niche called the posterior signaling centre (PSC) that orchestrates progenitor differentiation. Our previous work has demonstrated that the imaginal cell factor Headcase (Hdc, Heca) is required in the hematopoietic niche to control the differentiation of hemocyte progenitors. However, the downstream mechanisms of Hdc-mediated hematopoietic control remained unknown. Here we show that Hdc exerts this function by negatively regulating the insulin/mTOR signaling in the niche. When Hdc is depleted in the PSC, the overactivation of this pathway triggers reactive oxygen species (ROS) accumulation and, in turn, the differentiation of effector lamellocytes non-cell-autonomously. Although overactivation of insulin/mTOR signaling normally leads to an increase in the size of the hematopoietic niche, this effect is concealed by cell death caused by hdc loss-of-function. Moreover, we describe here that hdc silencing in progenitors causes cell-autonomous ROS elevation and JNK pathway activation, resulting in decreased MZ size and differentiation of lamellocytes. Similarly to the PSC niche, knocking down hdc in the MZ also leads to caspase activation. Notably, depleting Hdc in the progenitors triggers proliferation, an opposing effect to what is observed in the niche. These findings further our understanding of how progenitor maintenance in the larval lymph gland is controlled autonomously and non-cell-autonomously, and point towards new mechanisms potentially regulating HSC maintenance across vertebrates.
Collapse
Affiliation(s)
- Bayan Kharrat
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Erika Gábor
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Nikolett Virág
- Department of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Rita Sinka
- Department of Genetics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Jankovics
- Laboratory of Drosophila Germ Cell Differentiation, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Ildikó Kristó
- Drosophila Nuclear Actin Laboratory, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Péter Vilmos
- Drosophila Nuclear Actin Laboratory, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gábor Csordás
- Lysosomal Degradation Research Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Viktor Honti
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| |
Collapse
|
5
|
Goins LM, Girard JR, Mondal BC, Buran S, Su CC, Tang R, Biswas T, Kissi JA, Banerjee U. Wnt signaling couples G2 phase control with differentiation during hematopoiesis in Drosophila. Dev Cell 2024; 59:2477-2496.e5. [PMID: 38866012 PMCID: PMC11421984 DOI: 10.1016/j.devcel.2024.05.023] [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: 07/20/2023] [Revised: 03/27/2024] [Accepted: 05/17/2024] [Indexed: 06/14/2024]
Abstract
During homeostasis, a critical balance is maintained between myeloid-like progenitors and their differentiated progeny, which function to mitigate stress and innate immune challenges. The molecular mechanisms that help achieve this balance are not fully understood. Using genetic dissection in Drosophila, we show that a Wnt6/EGFR-signaling network simultaneously controls progenitor growth, proliferation, and differentiation. Unlike G1-quiescence of stem cells, hematopoietic progenitors are blocked in G2 phase by a β-catenin-independent (Wnt/STOP) Wnt6 pathway that restricts Cdc25 nuclear entry and promotes cell growth. Canonical β-catenin-dependent Wnt6 signaling is spatially confined to mature progenitors through localized activation of the tyrosine kinases EGFR and Abelson kinase (Abl), which promote nuclear entry of β-catenin and facilitate exit from G2. This strategy combines transcription-dependent and -independent forms of both Wnt6 and EGFR pathways to create a direct link between cell-cycle control and differentiation. This unique combinatorial strategy employing conserved components may underlie homeostatic balance and stress response in mammalian hematopoiesis.
Collapse
Affiliation(s)
- Lauren M Goins
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Juliet R Girard
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Bama Charan Mondal
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sausan Buran
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chloe C Su
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ruby Tang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Titash Biswas
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jessica A Kissi
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Utpal Banerjee
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Ho KYL, An K, Carr RL, Dvoskin AD, Ou AYJ, Vogl W, Tanentzapf G. Maintenance of hematopoietic stem cell niche homeostasis requires gap junction-mediated calcium signaling. Proc Natl Acad Sci U S A 2023; 120:e2303018120. [PMID: 37903259 PMCID: PMC10636368 DOI: 10.1073/pnas.2303018120] [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: 02/22/2023] [Accepted: 09/11/2023] [Indexed: 11/01/2023] Open
Abstract
Regulation of stem cells requires coordination of the cells that make up the stem cell niche. Here, we describe a mechanism that allows communication between niche cells to coordinate their activity and shape the signaling environment surrounding resident stem cells. Using the Drosophila hematopoietic organ, the lymph gland, we show that cells of the hematopoietic niche, the posterior signaling center (PSC), communicate using gap junctions (GJs) and form a signaling network. This network allows PSC cells to exchange Ca2+ signals repetitively which regulate the hematopoietic niche. Disruption of Ca2+ signaling in the PSC or the GJ-mediated network connecting niche cells causes dysregulation of the PSC and blood progenitor differentiation. Analysis of PSC-derived cell signaling shows that the Hedgehog pathway acts downstream of GJ-mediated Ca2+ signaling to modulate the niche microenvironment. These data show that GJ-mediated communication between hematopoietic niche cells maintains their homeostasis and consequently controls blood progenitor behavior.
Collapse
Affiliation(s)
- Kevin Y. L. Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Kevin An
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Rosalyn L. Carr
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
- British Columbia Children’s Hospital Research Institute, British Columbia Children’s Hospital, Vancouver, BCV5Z 4H4, Canada
| | - Alexandra D. Dvoskin
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Annie Y. J. Ou
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
- School of Kinesiology, University of British Columbia, Vancouver, BCV6T 1Z1, Canada
| | - Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BCV6T 1Z3, Canada
| |
Collapse
|
9
|
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
|
10
|
Tian Y, Morin-Poulard I, Liu X, Vanzo N, Crozatier M. A mechanosensitive vascular niche for Drosophila hematopoiesis. Proc Natl Acad Sci U S A 2023; 120:e2217862120. [PMID: 37094122 PMCID: PMC10160988 DOI: 10.1073/pnas.2217862120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
Hematopoietic stem and progenitor cells maintain blood cell homeostasis by integrating various cues provided by specialized microenvironments or niches. Biomechanical forces are emerging as key regulators of hematopoiesis. Here, we report that mechanical stimuli provided by blood flow in the vascular niche control Drosophila hematopoiesis. In vascular niche cells, the mechanosensitive channel Piezo transduces mechanical forces through intracellular calcium upregulation, leading to Notch activation and repression of FGF ligand transcription, known to regulate hematopoietic progenitor maintenance. Our results provide insight into how the vascular niche integrates mechanical stimuli to regulate hematopoiesis.
Collapse
Affiliation(s)
- Yushun Tian
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Ismaël Morin-Poulard
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Xiaohui Liu
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Nathalie Vanzo
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Michèle Crozatier
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| |
Collapse
|
11
|
Ho KYL, Carr RL, Dvoskin AD, Tanentzapf G. Kinetics of blood cell differentiation during hematopoiesis revealed by quantitative long-term live imaging. eLife 2023; 12:e84085. [PMID: 37000163 PMCID: PMC10065797 DOI: 10.7554/elife.84085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/08/2023] [Indexed: 04/01/2023] Open
Abstract
Stem cells typically reside in a specialized physical and biochemical environment that facilitates regulation of their behavior. For this reason, stem cells are ideally studied in contexts that maintain this precisely constructed microenvironment while still allowing for live imaging. Here, we describe a long-term organ culture and imaging strategy for hematopoiesis in flies that takes advantage of powerful genetic and transgenic tools available in this system. We find that fly blood progenitors undergo symmetric cell divisions and that their division is both linked to cell size and is spatially oriented. Using quantitative imaging to simultaneously track markers for stemness and differentiation in progenitors, we identify two types of differentiation that exhibit distinct kinetics. Moreover, we find that infection-induced activation of hematopoiesis occurs through modulation of the kinetics of cell differentiation. Overall, our results show that even subtle shifts in proliferation and differentiation kinetics can have large and aggregate effects to transform blood progenitors from a quiescent to an activated state.
Collapse
Affiliation(s)
- Kevin Yueh Lin Ho
- Department of Cellular and Physiological Sciences, University of British ColumbiaVancouverCanada
| | - Rosalyn Leigh Carr
- Department of Cellular and Physiological Sciences, University of British ColumbiaVancouverCanada
- School of Biomedical Engineering, University of British ColumbiaVancouverCanada
- British Columbia Children’s HospitalVancouverCanada
| | | | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British ColumbiaVancouverCanada
| |
Collapse
|
12
|
D'Souza LC, Kuriakose N, Raghu SV, Kabekkodu SP, Sharma A. ROS-directed activation of Toll/NF-κB in the hematopoietic niche triggers benzene-induced emergency hematopoiesis. Free Radic Biol Med 2022; 193:190-201. [PMID: 36216301 DOI: 10.1016/j.freeradbiomed.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 09/17/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022]
Abstract
Hematopoietic stem cells/progenitor cells (HSC/HPCs) orchestrate the hematopoietic process, effectively regulated by the hematopoietic niche under normal and stressed conditions. The hematopoietic niche provides various soluble factors which influence the differentiation and self-renewal of HSC/HSPs. Unceasing differentiation/proliferation/high metabolic activity of HSC/HPCs makes them susceptible to damage by environmental toxicants like benzene. Oxidative stress, epigenetic modifications, and DNA damage in the HSC/HPCs are the key factors of benzene-induced hematopoietic injury. However, the role of the hematopoietic niche in benzene-induced hematopoietic injury/response is still void. Therefore, the current study aims to unravel the role of the hematopoietic niche in benzene-induced hematotoxicity using a genetically tractable model, Drosophila melanogaster. The lymph gland is a dedicated hematopoietic organ in Drosophila larvae. A group of 30-45 cells called the posterior signaling center (PSC) in the lymph gland acts as a niche that regulates Drosophila HSC/HPCs maintenance. Benzene exposure to Drosophila larvae (48 h) resulted in aberrant hemocyte production, especially hyper-differentiation of lamellocytes followed by premature lymph gland dispersal and reduced adult emergence upon developmental exposure. Subsequent genetic experiments revealed that benzene-induced lamellocyte production and premature lymph gland dispersal were PSC mediated. The genetic experiments further showed that benzene generates Dual oxidase (Duox)-dependent Reactive Oxygen Species (ROS) in the PSC, activating Toll/NF-κB signaling, which is essential for the aberrant hemocyte production, lymph gland dispersal, and larval survival. Together, the study establishes a functional perspective of the hematopoietic niche in a benzene-induced hematopoietic emergency in a genetic model, Drosophila, which might be relevant to higher organisms.
Collapse
Affiliation(s)
- Leonard Clinton D'Souza
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Nithin Kuriakose
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India; Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Proteomics and Cancer Biology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Shamprasad Varija Raghu
- Neurogenetics Lab, Department of Applied Zoology, Mangalore University, Mangalagangothri, Konaje, Karnataka, 574199, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Science, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Anurag Sharma
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
13
|
Hultmark D, Andó I. Hematopoietic plasticity mapped in Drosophila and other insects. eLife 2022; 11:e78906. [PMID: 35920811 PMCID: PMC9348853 DOI: 10.7554/elife.78906] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/20/2022] [Indexed: 12/12/2022] Open
Abstract
Hemocytes, similar to vertebrate blood cells, play important roles in insect development and immunity, but it is not well understood how they perform their tasks. New technology, in particular single-cell transcriptomic analysis in combination with Drosophila genetics, may now change this picture. This review aims to make sense of recently published data, focusing on Drosophila melanogaster and comparing to data from other drosophilids, the malaria mosquito, Anopheles gambiae, and the silkworm, Bombyx mori. Basically, the new data support the presence of a few major classes of hemocytes: (1) a highly heterogenous and plastic class of professional phagocytes with many functions, called plasmatocytes in Drosophila and granular cells in other insects. (2) A conserved class of cells that control melanin deposition around parasites and wounds, called crystal cells in D. melanogaster, and oenocytoids in other insects. (3) A new class of cells, the primocytes, so far only identified in D. melanogaster. They are related to cells of the so-called posterior signaling center of the larval hematopoietic organ, which controls the hematopoiesis of other hemocytes. (4) Different kinds of specialized cells, like the lamellocytes in D. melanogaster, for the encapsulation of parasites. These cells undergo rapid evolution, and the homology relationships between such cells in different insects are uncertain. Lists of genes expressed in the different hemocyte classes now provide a solid ground for further investigation of function.
Collapse
Affiliation(s)
- Dan Hultmark
- Department of Molecular Biology, Umeå UniversityUmeåSweden
| | - István Andó
- Biological Research Centre, Institute of Genetics, Innate Immunity Group, Eötvös Loránd Research NetworkSzegedHungary
| |
Collapse
|
14
|
Kharrat B, Csordás G, Honti V. Peeling Back the Layers of Lymph Gland Structure and Regulation. Int J Mol Sci 2022; 23:7767. [PMID: 35887113 PMCID: PMC9319083 DOI: 10.3390/ijms23147767] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 12/18/2022] Open
Abstract
During the past 60 years, the fruit fly, Drosophila melanogaster, has proven to be an excellent model to study the regulation of hematopoiesis. This is not only due to the evolutionarily conserved signalling pathways and transcription factors contributing to blood cell fate, but also to convergent evolution that led to functional similarities in distinct species. An example of convergence is the compartmentalization of blood cells, which ensures the quiescence of hematopoietic stem cells and allows for the rapid reaction of the immune system upon challenges. The lymph gland, a widely studied hematopoietic organ of the Drosophila larva, represents a microenvironment with similar features and functions to classical hematopoietic stem cell niches of vertebrates. Lymph gland studies were effectively supported by the unparalleled toolkit developed in Drosophila, which enabled the high-resolution investigation of the cellular composition and regulatory interaction networks of the lymph gland. In this review, we summarize how our understanding of lymph gland structure and hematopoietic cell-to-cell communication evolved during the past decades and compare their analogous features to those of the vertebrate hematopoietic stem cell niche.
Collapse
Affiliation(s)
- Bayan Kharrat
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, Biological Research Centre, P.O. Box 521, H-6701 Szeged, Hungary;
- Faculty of Science and Informatics, Doctoral School of Biology, University of Szeged, P.O. Box 427, H-6720 Szeged, Hungary
| | - Gábor Csordás
- Lysosomal Degradation Research Group, Institute of Genetics, Biological Research Centre, P.O. Box 521, H-6701 Szeged, Hungary;
| | - Viktor Honti
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, Biological Research Centre, P.O. Box 521, H-6701 Szeged, Hungary;
| |
Collapse
|
15
|
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
|
16
|
Morin-Poulard I, Destalminil-Letourneau M, Bataillé L, Frendo JL, Lebreton G, Vanzo N, Crozatier M. Identification of Bipotential Blood Cell/Nephrocyte Progenitors in Drosophila: Another Route for Generating Blood Progenitors. Front Cell Dev Biol 2022; 10:834720. [PMID: 35237606 PMCID: PMC8883574 DOI: 10.3389/fcell.2022.834720] [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: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
The Drosophila lymph gland is the larval hematopoietic organ and is aligned along the anterior part of the cardiovascular system, composed of cardiac cells, that form the cardiac tube and its associated pericardial cells or nephrocytes. By the end of embryogenesis the lymph gland is composed of a single pair of lobes. Two additional pairs of posterior lobes develop during larval development to contribute to the mature lymph gland. In this study we describe the ontogeny of lymph gland posterior lobes during larval development and identify the genetic basis of the process. By lineage tracing we show here that each posterior lobe originates from three embryonic pericardial cells, thus establishing a bivalent blood cell/nephrocyte potential for a subset of embryonic pericardial cells. The posterior lobes of L3 larvae posterior lobes are composed of heterogeneous blood progenitors and their diversity is progressively built during larval development. We further establish that in larvae, homeotic genes and the transcription factor Klf15 regulate the choice between blood cell and nephrocyte fates. Our data underline the sequential production of blood cell progenitors during larval development.
Collapse
Affiliation(s)
- Ismaël Morin-Poulard
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France
| | - Manon Destalminil-Letourneau
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France
| | - Laetitia Bataillé
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France.,CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes), UMR6290, ERL U1305, Rennes, France
| | - Jean-Louis Frendo
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France.,INSERM U1301, CNRS 5070, Université de Toulouse, Toulouse, France
| | - Gaëlle Lebreton
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France
| | - Nathalie Vanzo
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France
| | - Michèle Crozatier
- Unité de Biologie Moléculaire et Cellulaire et du Développement (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse UMR 5077/CNRS, Toulouse, France
| |
Collapse
|
17
|
Spratford CM, Goins LM, Chi F, Girard JR, Macias SN, Ho VW, Banerjee U. Intermediate progenitor cells provide a transition between hematopoietic progenitors and their differentiated descendants. Development 2021; 148:273785. [PMID: 34918741 PMCID: PMC8722385 DOI: 10.1242/dev.200216] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Genetic and genomic analysis in Drosophila suggests that hematopoietic progenitors likely transition into terminal fates via intermediate progenitors (IPs) with some characteristics of either, but perhaps maintaining IP-specific markers. In the past, IPs have not been directly visualized and investigated owing to lack of appropriate genetic tools. Here, we report a Split GAL4 construct, CHIZ-GAL4, that identifies IPs as cells physically juxtaposed between true progenitors and differentiating hemocytes. IPs are a distinct cell type with a unique cell-cycle profile and they remain multipotent for all blood cell fates. In addition, through their dynamic control of the Notch ligand Serrate, IPs specify the fate of direct neighbors. The Ras pathway controls the number of IP cells and promotes their transition into differentiating cells. This study suggests that it would be useful to characterize such intermediate populations of cells in mammalian hematopoietic systems.
Collapse
Affiliation(s)
- Carrie M Spratford
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA.,Molecular Biology Institute, University of California, Los Angeles, USA
| | - Lauren M Goins
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA.,Molecular Biology Institute, University of California, Los Angeles, USA
| | - Fangtao Chi
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA.,Molecular Biology Institute, University of California, Los Angeles, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, USA
| | - Juliet R Girard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA.,Molecular Biology Institute, University of California, Los Angeles, USA
| | - Savannah N Macias
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA
| | - Vivien W Ho
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA
| | - Utpal Banerjee
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, USA.,Molecular Biology Institute, University of California, Los Angeles, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, USA.,Department of Biological Chemistry, University of California, Los Angeles, USA
| |
Collapse
|
18
|
Ho KYL, Khadilkar RJ, Carr RL, Tanentzapf G. A gap-junction-mediated, calcium-signaling network controls blood progenitor fate decisions in hematopoiesis. Curr Biol 2021; 31:4697-4712.e6. [PMID: 34480855 DOI: 10.1016/j.cub.2021.08.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/28/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022]
Abstract
Stem cell homeostasis requires coordinated fate decisions among stem cells that are often widely distributed within a tissue at varying distances from their stem cell niche. This requires a mechanism to ensure robust fate decisions within a population of stem cells. Here, we show that, in the Drosophila hematopoietic organ, the lymph gland (LG), gap junctions form a network that coordinates fate decisions between blood progenitors. Using live imaging of calcium signaling in intact LGs, we find that blood progenitors are connected through a signaling network. Blocking gap junction function disrupts this network, alters the pattern of encoded calcium signals, and leads to loss of progenitors and precocious blood cell differentiation. Ectopic and uniform activation of the calcium-signaling mediator CaMKII restores progenitor homeostasis when gap junctions are disrupted. Overall, these data show that gap junctions equilibrate cell signals between blood progenitors to coordinate fate decisions and maintain hematopoietic homeostasis.
Collapse
Affiliation(s)
- Kevin Y L Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Rohan J Khadilkar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Advanced Centre for Treatment, Research and Education in Cancer-Tata Memorial Centre (ACTREC-TMC), Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Rosalyn L Carr
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
19
|
Boulet M, Renaud Y, Lapraz F, Benmimoun B, Vandel L, Waltzer L. Characterization of the Drosophila Adult Hematopoietic System Reveals a Rare Cell Population With Differentiation and Proliferation Potential. Front Cell Dev Biol 2021; 9:739357. [PMID: 34722521 PMCID: PMC8550105 DOI: 10.3389/fcell.2021.739357] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023] Open
Abstract
While many studies have described Drosophila embryonic and larval blood cells, the hematopoietic system of the imago remains poorly characterized and conflicting data have been published concerning adult hematopoiesis. Using a combination of blood cell markers, we show that the adult hematopoietic system is essentially composed of a few distinct mature blood cell types. In addition, our transcriptomics results indicate that adult and larval blood cells have both common and specific features and it appears that adult hemocytes reactivate many genes expressed in embryonic blood cells. Interestingly, we identify a small set of blood cells that does not express differentiation markers but rather maintains the expression of the progenitor marker domeMeso. Yet, we show that these cells are derived from the posterior signaling center, a specialized population of cells present in the larval lymph gland, rather than from larval blood cell progenitors, and that their maintenance depends on the EBF transcription factor Collier. Furthermore, while these cells are normally quiescent, we find that some of them can differentiate and proliferate in response to bacterial infection. In sum, our results indicate that adult flies harbor a small population of specialized cells with limited hematopoietic potential and further support the idea that no substantial hematopoiesis takes place during adulthood.
Collapse
Affiliation(s)
- Manon Boulet
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
| | - Yoan Renaud
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
| | - François Lapraz
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Billel Benmimoun
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Laurence Vandel
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
| | - Lucas Waltzer
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France.,Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
20
|
Eleftherianos I, Heryanto C, Bassal T, Zhang W, Tettamanti G, Mohamed A. Haemocyte-mediated immunity in insects: Cells, processes and associated components in the fight against pathogens and parasites. Immunology 2021; 164:401-432. [PMID: 34233014 PMCID: PMC8517599 DOI: 10.1111/imm.13390] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
The host defence of insects includes a combination of cellular and humoral responses. The cellular arm of the insect innate immune system includes mechanisms that are directly mediated by haemocytes (e.g., phagocytosis, nodulation and encapsulation). In addition, melanization accompanying coagulation, clot formation and wound healing, nodulation and encapsulation processes leads to the formation of cytotoxic redox-cycling melanin precursors and reactive oxygen and nitrogen species. However, demarcation between cellular and humoral immune reactions as two distinct categories is not straightforward. This is because many humoral factors affect haemocyte functions and haemocytes themselves are an important source of many humoral molecules. There is also a considerable overlap between cellular and humoral immune functions that span from recognition of foreign intruders to clot formation. Here, we review these immune reactions starting with the cellular mechanisms that limit haemolymph loss and participate in wound healing and clot formation and advancing to cellular functions that are critical in restricting pathogen movement and replication. This information is important because it highlights that insect cellular immunity is controlled by a multilayered system, different components of which are activated by different pathogens or during the different stages of the infection.
Collapse
Affiliation(s)
- Ioannis Eleftherianos
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Christa Heryanto
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Taha Bassal
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| | - Wei Zhang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural BioengineeringKey Laboratory of Green Pesticide and Agricultural BioengineeringMinistry of EducationGuizhou UniversityGuiyangChina
| | - Gianluca Tettamanti
- Department of Biotechnology and Life SciencesUniversity of InsubriaVareseItaly
- BAT Center‐Interuniversity Center for Studies on Bioinspired Agro‐Environmental TechnologyUniversity of Napoli Federico IINapoliItaly
| | - Amr Mohamed
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| |
Collapse
|
21
|
Girard JR, Goins LM, Vuu DM, Sharpley MS, Spratford CM, Mantri SR, Banerjee U. Paths and pathways that generate cell-type heterogeneity and developmental progression in hematopoiesis. eLife 2021; 10:e67516. [PMID: 34713801 PMCID: PMC8610493 DOI: 10.7554/elife.67516] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022] Open
Abstract
Mechanistic studies of Drosophila lymph gland hematopoiesis are limited by the availability of cell-type-specific markers. Using a combination of bulk RNA-Seq of FACS-sorted cells, single-cell RNA-Seq, and genetic dissection, we identify new blood cell subpopulations along a developmental trajectory with multiple paths to mature cell types. This provides functional insights into key developmental processes and signaling pathways. We highlight metabolism as a driver of development, show that graded Pointed expression allows distinct roles in successive developmental steps, and that mature crystal cells specifically express an alternate isoform of Hypoxia-inducible factor (Hif/Sima). Mechanistically, the Musashi-regulated protein Numb facilitates Sima-dependent non-canonical, and inhibits canonical, Notch signaling. Broadly, we find that prior to making a fate choice, a progenitor selects between alternative, biologically relevant, transitory states allowing smooth transitions reflective of combinatorial expressions rather than stepwise binary decisions. Increasingly, this view is gaining support in mammalian hematopoiesis.
Collapse
Affiliation(s)
- Juliet R Girard
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Lauren M Goins
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Dung M Vuu
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Mark S Sharpley
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Carrie M Spratford
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Shreya R Mantri
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Utpal Banerjee
- Department of Molecular, Cell and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
- Department of Biological Chemistry, University of California, Los AngelesLos AngelesUnited States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
22
|
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
|
23
|
Kanwal A, Joshi PV, Mandal S, Mandal L. Ubx-Collier signaling cascade maintains blood progenitors in the posterior lobes of the Drosophila larval lymph gland. PLoS Genet 2021; 17:e1009709. [PMID: 34370733 PMCID: PMC8376192 DOI: 10.1371/journal.pgen.1009709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/19/2021] [Accepted: 07/12/2021] [Indexed: 12/02/2022] Open
Abstract
Drosophila larval hematopoiesis occurs in a specialized multi-lobed organ called the lymph gland. Extensive characterization of the organ has provided mechanistic insights into events related to developmental hematopoiesis. Spanning from the thoracic to the abdominal segment of the larvae, this organ comprises a pair of primary, secondary, and tertiary lobes. Much of our understanding arises from the studies on the primary lobe, while the secondary and tertiary lobes have remained mostly unexplored. Previous studies have inferred that these lobes are composed of progenitors that differentiate during pupation; however, the mechanistic basis of this extended progenitor state remains unclear. This study shows that posterior lobe progenitors are maintained by a local signaling center defined by Ubx and Collier in the tertiary lobe. This Ubx zone in the tertiary lobe shares several markers with the niche of the primary lobe. Ubx domain regulates the homeostasis of the posterior lobe progenitors in normal development and an immune-challenged scenario. Our study establishes the lymph gland as a model to tease out how the progenitors interface with the dual niches within an organ during development and disorders.
Collapse
Affiliation(s)
- Aditya Kanwal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| | - Pranav Vijay Joshi
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| | - Lolitika Mandal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| |
Collapse
|
24
|
Wan B, Belghazi M, Lemauf S, Poirié M, Gatti JL. Proteomics of purified lamellocytes from Drosophila melanogaster HopT um-l identifies new membrane proteins and networks involved in their functions. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 134:103584. [PMID: 34033897 DOI: 10.1016/j.ibmb.2021.103584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
In healthy Drosophila melanogaster larvae, plasmatocytes and crystal cells account for 95% and 5% of the hemocytes, respectively. A third type of hemocytes, lamellocytes, are rare, but their number increases after oviposition by parasitoid wasps. The lamellocytes form successive layers around the parasitoid egg, leading to its encapsulation and melanization, and finally the death of this intruder. However, the total number of lamellocytes per larva remains quite low even after parasitoid infestation, making direct biochemical studies difficult. Here, we used the HopTum-l mutant strain that constitutively produces large numbers of lamellocytes to set up a purification method and analyzed their major proteins by 2D gel electrophoresis and their plasma membrane surface proteins by 1D SDS-PAGE after affinity purification. Mass spectrometry identified 430 proteins from 2D spots and 344 affinity-purified proteins from 1D bands, for a total of 639 unique proteins. Known lamellocyte markers such as PPO3 and the myospheroid integrin were among the components identified with specific chaperone proteins. Affinity purification detected other integrins, as well as a wide range of integrin-associated proteins involved in the formation and function of cell-cell junctions. Overall, the newly identified proteins indicate that these cells are highly adapted to the encapsulation process (recognition, motility, adhesion, signaling), but may also have several other physiological functions (such as secretion and internalization of vesicles) under different signaling pathways. These results provide the basis for further in vivo and in vitro studies of lamellocytes, including the development of new markers to identify coexisting populations and their respective origins and functions in Drosophila immunity.
Collapse
Affiliation(s)
- Bin Wan
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Maya Belghazi
- Institute of NeuroPhysiopathology (INP), UMR7051, CNRS, Aix-Marseille Université, Marseille, 13015, France
| | - Séverine Lemauf
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Marylène Poirié
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France
| | - Jean-Luc Gatti
- Université Côte d'Azur, INRAE, CNRS, Institute Sophia-Agrobiotech, Sophia Antipolis, France.
| |
Collapse
|
25
|
A parasitoid wasp of Drosophila employs preemptive and reactive strategies to deplete its host's blood cells. PLoS Pathog 2021; 17:e1009615. [PMID: 34048506 PMCID: PMC8191917 DOI: 10.1371/journal.ppat.1009615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/10/2021] [Accepted: 05/05/2021] [Indexed: 11/19/2022] Open
Abstract
The wasps Leptopilina heterotoma parasitize and ingest their Drosophila hosts. They produce extracellular vesicles (EVs) in the venom that are packed with proteins, some of which perform immune suppressive functions. EV interactions with blood cells of host larvae are linked to hematopoietic depletion, immune suppression, and parasite success. But how EVs disperse within the host, enter and kill hematopoietic cells is not well understood. Using an antibody marker for L. heterotoma EVs, we show that these parasite-derived structures are readily distributed within the hosts’ hemolymphatic system. EVs converge around the tightly clustered cells of the posterior signaling center (PSC) of the larval lymph gland, a small hematopoietic organ in Drosophila. The PSC serves as a source of developmental signals in naïve animals. In wasp-infected animals, the PSC directs the differentiation of lymph gland progenitors into lamellocytes. These lamellocytes are needed to encapsulate the wasp egg and block parasite development. We found that L. heterotoma infection disassembles the PSC and PSC cells disperse into the disintegrating lymph gland lobes. Genetically manipulated PSC-less lymph glands remain non-responsive and largely intact in the face of L. heterotoma infection. We also show that the larval lymph gland progenitors use the endocytic machinery to internalize EVs. Once inside, L. heterotoma EVs damage the Rab7- and LAMP-positive late endocytic and phagolysosomal compartments. Rab5 maintains hematopoietic and immune quiescence as Rab5 knockdown results in hematopoietic over-proliferation and ectopic lamellocyte differentiation. Thus, both aspects of anti-parasite immunity, i.e., (a) phagocytosis of the wasp’s immune-suppressive EVs, and (b) progenitor differentiation for wasp egg encapsulation reside in the lymph gland. These results help explain why the lymph gland is specifically and precisely targeted for destruction. The parasite’s simultaneous and multipronged approach to block cellular immunity not only eliminates blood cells, but also tactically blocks the genetic programming needed for supplementary hematopoietic differentiation necessary for host success. In addition to its known functions in hematopoiesis, our results highlight a previously unrecognized phagocytic role of the lymph gland in cellular immunity. EV-mediated virulence strategies described for L. heterotoma are likely to be shared by other parasitoid wasps; their understanding can improve the design and development of novel therapeutics and biopesticides as well as help protect biodiversity. Parasitoid wasps serve as biological control agents of agricultural insect pests and are worthy of study. Many parasitic wasps develop inside their hosts to emerge as free-living adults. To overcome the resistance of their hosts, parasitic wasps use varied and ingenious strategies such as mimicry, evasion, bioactive venom, virus-like particles, viruses, and extracellular vesicles (EVs). We describe the effects of a unique class of EVs containing virulence proteins and produced in the venom of wasps that parasitize fruit flies of Drosophila species. EVs from Leptopilina heterotoma are widely distributed throughout the Drosophila hosts’ circulatory system after infection. They enter and kill macrophages by destroying the very same subcellular machinery that facilitates their uptake. An important protein in this process, Rab5, is needed to maintain the identity of the macrophage; when Rab5 function is reduced, macrophages turn into a different cell type called lamellocytes. Activities in the EVs can eliminate lamellocytes as well. EVs also interfere with the hosts’ genetic program that promotes lamellocyte differentiation needed to block parasite development. Thus, wasps combine specific preemptive and reactive strategies to deplete their hosts of the very cells that would otherwise sequester and kill them. These findings have applied value in agricultural pest control and medical therapeutics.
Collapse
|
26
|
Destalminil-Letourneau M, Morin-Poulard I, Tian Y, Vanzo N, Crozatier M. The vascular niche controls Drosophila hematopoiesis via fibroblast growth factor signaling. eLife 2021; 10:64672. [PMID: 33395389 PMCID: PMC7781598 DOI: 10.7554/elife.64672] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
In adult mammals, hematopoiesis, the production of blood cells from hematopoietic stem and progenitor cells (HSPCs), is tightly regulated by extrinsic signals from the microenvironment called 'niche'. Bone marrow HSPCs are heterogeneous and controlled by both endosteal and vascular niches. The Drosophila hematopoietic lymph gland is located along the cardiac tube which corresponds to the vascular system. In the lymph gland, the niche called Posterior Signaling Center controls only a subset of the heterogeneous hematopoietic progenitor population indicating that additional signals are necessary. Here we report that the vascular system acts as a second niche to control lymph gland homeostasis. The FGF ligand Branchless produced by vascular cells activates the FGF pathway in hematopoietic progenitors. By regulating intracellular calcium levels, FGF signaling maintains progenitor pools and prevents blood cell differentiation. This study reveals that two niches contribute to the control ofDrosophila blood cell homeostasis through their differential regulation of progenitors.
Collapse
Affiliation(s)
- Manon Destalminil-Letourneau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Ismaël Morin-Poulard
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yushun Tian
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Vanzo
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Michele Crozatier
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
27
|
Cho B, Yoon SH, Lee D, Koranteng F, Tattikota SG, Cha N, Shin M, Do H, Hu Y, Oh SY, Lee D, Vipin Menon A, Moon SJ, Perrimon N, Nam JW, Shim J. Single-cell transcriptome maps of myeloid blood cell lineages in Drosophila. Nat Commun 2020; 11:4483. [PMID: 32900993 PMCID: PMC7479620 DOI: 10.1038/s41467-020-18135-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/06/2020] [Indexed: 11/22/2022] Open
Abstract
The Drosophila lymph gland, the larval hematopoietic organ comprised of prohemocytes and mature hemocytes, has been a valuable model for understanding mechanisms underlying hematopoiesis and immunity. Three types of mature hemocytes have been characterized in the lymph gland: plasmatocytes, lamellocytes, and crystal cells, which are analogous to vertebrate myeloid cells, yet molecular underpinnings of the lymph gland hemocytes have been less investigated. Here, we use single-cell RNA sequencing to comprehensively analyze heterogeneity of developing hemocytes in the lymph gland, and discover previously undescribed hemocyte types including adipohemocytes, stem-like prohemocytes, and intermediate prohemocytes. Additionally, we identify the developmental trajectory of hemocytes during normal development as well as the emergence of the lamellocyte lineage following active cellular immunity caused by wasp infestation. Finally, we establish similarities and differences between embryonically derived- and larval lymph gland hemocytes. Altogether, our study provides detailed insights into the hemocyte development and cellular immune responses at single-cell resolution. How the Drosophila lymph gland hemocytes develop and are regulated at a single-cell level is unclear. Here, the authors use single-cell RNA sequencing to show heterogeneity of developing hemocytes in the lymph gland and how they react to wasp infestation, and compare hemocytes from two independent origins.
Collapse
Affiliation(s)
- Bumsik Cho
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Sang-Ho Yoon
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Daewon Lee
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Ferdinand Koranteng
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | | | - Nuri Cha
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Mingyu Shin
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Hobin Do
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Sue Young Oh
- Department of Oral Biology, Yonsei University, College of Dentistry, Seoul, 03722, Republic of Korea
| | - Daehan Lee
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - A Vipin Menon
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea
| | - Seok Jun Moon
- Department of Oral Biology, Yonsei University, College of Dentistry, Seoul, 03722, Republic of Korea
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA.,Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Jin-Wu Nam
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea. .,Research Institute for Natural Sciences, Hanyang University, Seoul, 04736, Republic of Korea. .,Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04736, Republic of Korea.
| | - Jiwon Shim
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, 04736, Republic of Korea. .,Research Institute for Natural Sciences, Hanyang University, Seoul, 04736, Republic of Korea. .,Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 04736, Republic of Korea.
| |
Collapse
|
28
|
Lan W, Liu S, Zhao L, Su Y. Regulation of Drosophila Hematopoiesis in Lymph Gland: From a Developmental Signaling Point of View. Int J Mol Sci 2020; 21:ijms21155246. [PMID: 32722007 PMCID: PMC7432643 DOI: 10.3390/ijms21155246] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The Drosophila hematopoietic system is becoming increasingly attractive for its simple blood cell lineage and its developmental and functional parallels with the vertebrate system. As the dedicated organ for Drosophila larval hematopoiesis, the lymph gland harbors both multipotent stem-like progenitor cells and differentiated blood cells. The balance between progenitor maintenance and differentiation in the lymph gland must be precisely and tightly controlled. Multiple developmental signaling pathways, such as Notch, Hedgehog, and Wnt/Wingless, have been demonstrated to regulate the hematopoietic processes in the lymph gland. Focusing on blood cell maintenance and differentiation, this article summarizes the functions of several classic developmental signaling pathways for lymph gland growth and patterning, highlighting the important roles of developmental signaling during lymph gland development as well as Drosophila larval hematopoiesis.
Collapse
Affiliation(s)
- Wenwen Lan
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Sumin Liu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- Fisheries College, Ocean University of China, Qingdao 266003, China
- Correspondence: (L.Z.); (Y.S.)
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Correspondence: (L.Z.); (Y.S.)
| |
Collapse
|
29
|
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: 5] [Impact Index Per Article: 1.0] [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
|
30
|
Bataillé L, Colombié N, Pelletier A, Paululat A, Lebreton G, Carrier Y, Frendo JL, Vincent A. Alary muscles and thoracic alary-related muscles are atypical striated muscles involved in maintaining the position of internal organs. Development 2020; 147:dev.185645. [PMID: 32188630 DOI: 10.1242/dev.185645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Alary muscles (AMs) have been described as a component of the cardiac system in various arthropods. Lineage-related thoracic muscles (TARMs), linking the exoskeleton to specific gut regions, have recently been discovered in Drosophila Asymmetrical attachments of AMs and TARMs, to the exoskeleton on one side and internal organs on the other, suggested an architectural function in moving larvae. Here, we analysed the shape and sarcomeric organisation of AMs and TARMs, and imaged their atypical deformability in crawling larvae. We then selectively eliminated AMs and TARMs by targeted apoptosis. Elimination of AMs revealed that AMs are required for suspending the heart in proper intra-haemocelic position and for opening of the heart lumen, and that AMs constrain the curvature of the respiratory tracheal system during crawling; TARMs are required for proper positioning of visceral organs and efficient food transit. AM/TARM cardiac versus visceral attachment depends on Hox control, with visceral attachment being the ground state. TARMs and AMs are the first example of multinucleate striated muscles connecting the skeleton to the cardiac and visceral systems in bilaterians, with multiple physiological functions.
Collapse
Affiliation(s)
- Laetitia Bataillé
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Nathalie Colombié
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Aurore Pelletier
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Achim Paululat
- University of Osnabrück, Department of Biology/Chemistry, Zoology and Developmental Biology, Barbarastraße 11, 49076 Osnabrück, Germany
| | - Gaëlle Lebreton
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Yannick Carrier
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Jean-Louis Frendo
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Alain Vincent
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| |
Collapse
|
31
|
Blanco-Obregon D, Katz MJ, Durrieu L, Gándara L, Wappner P. Context-specific functions of Notch in Drosophila blood cell progenitors. Dev Biol 2020; 462:101-115. [PMID: 32243888 DOI: 10.1016/j.ydbio.2020.03.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 01/10/2023]
Abstract
Drosophila Larval hematopoiesis takes place at the lymph gland, where myeloid-like progenitors differentiate into Plasmatocytes and Crystal Cells, under regulation of conserved signaling pathways. It has been established that the Notch pathway plays a specific role in Crystal Cell differentiation and maintenance. In mammalian hematopoiesis, the Notch pathway has been proposed to fulfill broader functions, including Hematopoietic Stem Cell maintenance and cell fate decision in progenitors. In this work we describe different roles that Notch plays in the lymph gland. We show that Notch, activated by its ligand Serrate, expressed at the Posterior Signaling Center, is required to restrain Core Progenitor differentiation. We define a novel population of blood cell progenitors that we name Distal Progenitors, where Notch, activated by Serrate expressed in Lineage Specifying Cells at the Medullary Zone/Cortical Zone boundary, regulates a binary decision between Plasmatocyte and Crystal Cell fates. Thus, Notch plays context-specific functions in different blood cell progenitor populations of the Drosophila lymph gland.
Collapse
Affiliation(s)
- D Blanco-Obregon
- Instituto Leloir, CONICET, Patricias Argentinas 435, Buenos Aires, 1405, Argentina
| | - M J Katz
- Instituto Leloir, CONICET, Patricias Argentinas 435, Buenos Aires, 1405, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - L Durrieu
- Instituto Leloir, CONICET, Patricias Argentinas 435, Buenos Aires, 1405, Argentina; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales-Universidad de Buenos Aires, Buenos Aires, 1428, Argentina
| | - L Gándara
- Instituto Leloir, CONICET, Patricias Argentinas 435, Buenos Aires, 1405, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - P Wappner
- Instituto Leloir, CONICET, Patricias Argentinas 435, Buenos Aires, 1405, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales-Universidad de Buenos Aires, Buenos Aires, 1428, Argentina.
| |
Collapse
|
32
|
Cell Adhesion-Mediated Actomyosin Assembly Regulates the Activity of Cubitus Interruptus for Hematopoietic Progenitor Maintenance in Drosophila. Genetics 2019; 212:1279-1300. [PMID: 31138608 PMCID: PMC6707476 DOI: 10.1534/genetics.119.302209] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
The actomyosin network is involved in crucial cellular processes including morphogenesis, cell adhesion, apoptosis, proliferation, differentiation, and collective cell migration in Drosophila, Caenorhabditiselegans, and mammals. Here, we demonstrate that Drosophila larval blood stem-like progenitors require actomyosin activity for their maintenance. Genetic loss of the actomyosin network from progenitors caused a decline in their number. Likewise, the progenitor population increased upon sustained actomyosin activation via phosphorylation by Rho-associated kinase. We show that actomyosin positively regulates larval blood progenitors by controlling the maintenance factor Cubitus interruptus (Ci). Overexpression of the maintenance signal via a constitutively activated construct (ci.HA) failed to sustain Ci-155 in the absence of actomyosin components like Zipper (zip) and Squash (sqh), thus favoring protein kinase A (PKA)-independent regulation of Ci activity. Furthermore, we demonstrate that a change in cortical actomyosin assembly mediated by DE-cadherin modulates Ci activity, thereby determining progenitor status. Thus, loss of cell adhesion and downstream actomyosin activity results in desensitization of the progenitors to Hh signaling, leading to their differentiation. Our data reveal how cell adhesion and the actomyosin network cooperate to influence patterning, morphogenesis, and maintenance of the hematopoietic stem-like progenitor pool in the developing Drosophila hematopoietic organ.
Collapse
|
33
|
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
|
34
|
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
|
35
|
Banerjee U, Girard JR, Goins LM, Spratford CM. Drosophila as a Genetic Model for Hematopoiesis. Genetics 2019; 211:367-417. [PMID: 30733377 PMCID: PMC6366919 DOI: 10.1534/genetics.118.300223] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
In this FlyBook chapter, we present a survey of the current literature on the development of the hematopoietic system in Drosophila The Drosophila blood system consists entirely of cells that function in innate immunity, tissue integrity, wound healing, and various forms of stress response, and are therefore functionally similar to myeloid cells in mammals. The primary cell types are specialized for phagocytic, melanization, and encapsulation functions. As in mammalian systems, multiple sites of hematopoiesis are evident in Drosophila and the mechanisms involved in this process employ many of the same molecular strategies that exemplify blood development in humans. Drosophila blood progenitors respond to internal and external stress by coopting developmental pathways that involve both local and systemic signals. An important goal of these Drosophila studies is to develop the tools and mechanisms critical to further our understanding of human hematopoiesis during homeostasis and dysfunction.
Collapse
Affiliation(s)
- Utpal Banerjee
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
- Molecular Biology Institute, University of California, Los Angeles, California 90095
- Department of Biological Chemistry, University of California, Los Angeles, California 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| | - Juliet R Girard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Lauren M Goins
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Carrie M Spratford
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| |
Collapse
|
36
|
Baldeosingh R, Gao H, Wu X, Fossett N. Hedgehog signaling from the Posterior Signaling Center maintains U-shaped expression and a prohemocyte population in Drosophila. Dev Biol 2018; 441:132-145. [PMID: 29966604 PMCID: PMC6064674 DOI: 10.1016/j.ydbio.2018.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022]
Abstract
Hematopoietic progenitor choice between multipotency and differentiation is tightly regulated by intrinsic factors and extrinsic signals from the surrounding microenvironment. The Drosophila melanogaster hematopoietic lymph gland has emerged as a powerful tool to investigate mechanisms that regulate hematopoietic progenitor choice in vivo. The lymph gland contains progenitor cells, which share key characteristics with mammalian hematopoietic progenitors such as quiescence, multipotency and niche-dependence. The lymph gland is zonally arranged, with progenitors located in medullary zone, differentiating cells in the cortical zone, and the stem cell niche or Posterior Signaling Center (PSC) residing at the base of the medullary zone (MZ). This arrangement facilitates investigations into how signaling from the microenvironment controls progenitor choice. The Drosophila Friend of GATA transcriptional regulator, U-shaped, is a conserved hematopoietic regulator. To identify additional novel intrinsic and extrinsic regulators that interface with U-shaped to control hematopoiesis, we conducted an in vivo screen for factors that genetically interact with u-shaped. Smoothened, a downstream effector of Hedgehog signaling, was one of the factors identified in the screen. Here we report our studies that characterized the relationship between Smoothened and U-shaped. We showed that the PSC and Hedgehog signaling are required for U-shaped expression and that U-shaped is an important intrinsic progenitor regulator. These observations identify a potential link between the progenitor regulatory machinery and extrinsic signals from the PSC. Furthermore, we showed that both Hedgehog signaling and the PSC are required to maintain a subpopulation of progenitors. This led to a delineation of PSC-dependent versus PSC-independent progenitors and provided further evidence that the MZ progenitor population is heterogeneous. Overall, we have identified a connection between a conserved hematopoietic master regulator and a putative stem cell niche, which adds to our understanding of how signals from the microenvironment regulate progenitor multipotency.
Collapse
Affiliation(s)
- Rajkumar Baldeosingh
- Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hongjuan Gao
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaorong Wu
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nancy Fossett
- Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
37
|
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
|
38
|
Tokusumi T, Tokusumi Y, Schulz RA. The mir-7 and bag of marbles genes regulate Hedgehog pathway signaling in blood cell progenitors in Drosophila larval lymph glands. Genesis 2018; 56:e23210. [PMID: 29663653 DOI: 10.1002/dvg.23210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 01/03/2023]
Abstract
Hedgehog (Hh) pathway signaling is crucial for the maintenance of blood cell progenitors in the lymph gland hematopoietic organ present in Drosophila third instar larvae. Previous studies from our lab have likewise shown the importance of the mir-7 and bag of marbles (bam) genes in maintaining the progenitor state. Thus, we sought to investigate a possible interaction between the Hh pathway and mir-7/bam in the prohemocyte population within this hematopoietic tissue. Gain of function mir-7 was able to rescue a blood cell progenitor depletion phenotype caused by Patched (Ptc) inhibition of Hh pathway signaling in these cells. Similarly, expression of a dominant/negative version of Ptc was able to rescue the severe reduction of prohemocytes due to bam loss of function. Furthermore, we demonstrated that Suppressor of fused [Su(fu)], another known inhibitor of Hh signaling, likely serves as a translational repression target of the mir-7 miRNA. Our results suggest the mir-7/bam combination regulates the Hh signaling network through repression of Su(fu) to maintain hemocyte progenitors in the larval lymph gland.
Collapse
Affiliation(s)
- Tsuyoshi Tokusumi
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | - Yumiko Tokusumi
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | - Robert A Schulz
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
39
|
From Drosophila Blood Cells to Human Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1076:195-214. [PMID: 29951821 DOI: 10.1007/978-981-13-0529-0_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The hematopoietic system plays a critical role in establishing the proper response against invading pathogens or in removing cancerous cells. Furthermore, deregulations of the hematopoietic differentiation program are at the origin of numerous diseases including leukemia. Importantly, many aspects of blood cell development have been conserved from human to Drosophila. Hence, Drosophila has emerged as a potent genetic model to study blood cell development and leukemia in vivo. In this chapter, we give a brief overview of the Drosophila hematopoietic system, and we provide a protocol for the dissection and the immunostaining of the larval lymph gland, the most studied hematopoietic organ in Drosophila. We then focus on the various paradigms that have been used in fly to investigate how conserved genes implicated in leukemogenesis control blood cell development. Specific examples of Drosophila models for leukemia are presented, with particular attention to the most translational ones. Finally, we discuss some limitations and potential improvements of Drosophila models for studying blood cell cancer.
Collapse
|
40
|
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
|
41
|
Heavner ME, Ramroop J, Gueguen G, Ramrattan G, Dolios G, Scarpati M, Kwiat J, Bhattacharya S, Wang R, Singh S, Govind S. Novel Organelles with Elements of Bacterial and Eukaryotic Secretion Systems Weaponize Parasites of Drosophila. Curr Biol 2017; 27:2869-2877.e6. [PMID: 28889977 DOI: 10.1016/j.cub.2017.08.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/03/2017] [Accepted: 08/10/2017] [Indexed: 01/16/2023]
Abstract
The evolutionary success of parasitoid wasps, a highly diverse group of insects widely used in biocontrol, depends on a variety of life history strategies in conflict with those of their hosts [1]. Drosophila melanogaster is a natural host of parasitic wasps of the genus Leptopilina. Attack by L. boulardi (Lb), a specialist wasp to flies of the melanogaster group, activates NF-κB-mediated humoral and cellular immunity. Inflammatory blood cells mobilize and encapsulate Lb eggs and embryos [2-5]. L. heterotoma (Lh), a generalist wasp, kills larval blood cells and actively suppresses immune responses. Spiked virus-like particles (VLPs) in wasp venom have clearly been linked to wasps' successful parasitism of Drosophila [6], but the composition of VLPs and their biotic nature have remained mysterious. Our proteomics studies reveal that VLPs lack viral coat proteins but possess a pharmacopoeia of (1) the eukaryotic vesicular transport system, (2) immunity, and (3) previously unknown proteins. These novel proteins distinguish Lh from Lb VLPs; notably, some proteins specific to Lh VLPs possess sequence similarities with bacterial secretion system proteins. Structure-informed analyses of an abundant Lh VLP surface and spike-tip protein, p40, reveal similarities to the needle-tip invasin proteins SipD and IpaD of Gram-negative bacterial type-3 secretion systems that breach immune barriers and deliver virulence factors into mammalian cells. Our studies suggest that Lh VLPs represent a new class of extracellular organelles and share pathways for protein delivery with both eukaryotic microvesicles and bacterial surface secretion systems. Given their mixed prokaryotic and eukaryotic properties, we propose the term mixed-strategy extracellular vesicle (MSEV) to replace VLP.
Collapse
Affiliation(s)
- Mary Ellen Heavner
- Biology, The City College of New York, Convent Avenue, New York, NY 10031, USA; PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Johnny Ramroop
- Biology, The City College of New York, Convent Avenue, New York, NY 10031, USA; PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Gwenaelle Gueguen
- Biology, The City College of New York, Convent Avenue, New York, NY 10031, USA
| | - Girish Ramrattan
- Biological Sciences, Hunter College, Park Avenue, New York, NY 10065, USA
| | - Georgia Dolios
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Madison Avenue, New York, NY 10029, USA
| | - Michael Scarpati
- PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; Biology, Brooklyn College, Bedford Avenue, Brooklyn, NY 11210, USA
| | - Jonathan Kwiat
- Biology, Brooklyn College, Bedford Avenue, Brooklyn, NY 11210, USA
| | - Sharmila Bhattacharya
- Space Biosciences Research Branch, NASA Ames Research Center, Moffett Boulevard, Mountain View, CA 94035, USA
| | - Rong Wang
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Madison Avenue, New York, NY 10029, USA
| | - Shaneen Singh
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; Biology, Brooklyn College, Bedford Avenue, Brooklyn, NY 11210, USA
| | - Shubha Govind
- Biology, The City College of New York, Convent Avenue, New York, NY 10031, USA; PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA; PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA.
| |
Collapse
|
42
|
Khadilkar RJ, Vogl W, Goodwin K, Tanentzapf G. Modulation of occluding junctions alters the hematopoietic niche to trigger immune activation. eLife 2017; 6:28081. [PMID: 28841136 PMCID: PMC5597334 DOI: 10.7554/elife.28081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/24/2017] [Indexed: 12/04/2022] Open
Abstract
Stem cells are regulated by signals from their microenvironment, or niche. During Drosophila hematopoiesis, a niche regulates prohemocytes to control hemocyte production. Immune challenges activate cell-signalling to initiate the cellular and innate immune response. Specifically, certain immune challenges stimulate the niche to produce signals that induce prohemocyte differentiation. However, the mechanisms that promote prohemocyte differentiation subsequent to immune challenges are poorly understood. Here we show that bacterial infection induces the cellular immune response by modulating occluding-junctions at the hematopoietic niche. Occluding-junctions form a permeability barrier that regulates the accessibility of prohemocytes to niche derived signals. The immune response triggered by infection causes barrier breakdown, altering the prohemocyte microenvironment to induce immune cell production. Moreover, genetically induced barrier ablation provides protection against infection by activating the immune response. Our results reveal a novel role for occluding-junctions in regulating niche-hematopoietic progenitor signalling and link this mechanism to immune cell production following infection.
Collapse
Affiliation(s)
- Rohan J Khadilkar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Katharine Goodwin
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
43
|
Zhang CU, Cadigan KM. The matrix protein Tiggrin regulates plasmatocyte maturation in Drosophila larva. Development 2017; 144:2415-2427. [PMID: 28526755 DOI: 10.1242/dev.149641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 05/11/2017] [Indexed: 01/24/2023]
Abstract
The lymph gland (LG) is a major source of hematopoiesis during Drosophila development. In this tissue, prohemocytes differentiate into multiple lineages, including macrophage-like plasmatocytes, which comprise the vast majority of mature hemocytes. Previous studies have uncovered genetic pathways that regulate prohemocyte maintenance and some cell fate choices between hemocyte lineages. However, less is known about how the plasmatocyte pool of the LG is established and matures. Here, we report that Tiggrin, a matrix protein expressed in the LG, is a specific regulator of plasmatocyte maturation. Tiggrin mutants exhibit precocious maturation of plasmatocytes, whereas Tiggrin overexpression blocks this process, resulting in a buildup of intermediate progenitors (IPs) expressing prohemocyte and hemocyte markers. These IPs likely represent a transitory state in prohemocyte to plasmatocyte differentiation. We also found that overexpression of Wee1 kinase, which slows G2/M progression, results in a phenotype similar to Tiggrin overexpression, whereas String/Cdc25 expression phenocopies Tiggrin mutants. Further analysis revealed that Wee1 inhibits plasmatocyte maturation through upregulation of Tiggrin transcription. Our results elucidate connections between the extracellular matrix and cell cycle regulators in the regulation of hematopoiesis.
Collapse
Affiliation(s)
- Chen U Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ken M Cadigan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
44
|
Hao Y, Jin LH. Dual role for Jumu in the control of hematopoietic progenitors in the Drosophila lymph gland. eLife 2017; 6. [PMID: 28350299 PMCID: PMC5391210 DOI: 10.7554/elife.25094] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/20/2017] [Indexed: 12/24/2022] Open
Abstract
The Drosophila lymph gland is a hematopoietic organ in which the maintenance of hematopoietic progenitor cell fate relies on intrinsic factors and extensive interaction with cells within a microenvironment. The posterior signaling center (PSC) is required for maintaining the balance between progenitors and their differentiation into mature hemocytes. Moreover, some factors from the progenitors cell-autonomously control blood cell differentiation. Here, we show that Jumeau (Jumu), a member of the forkhead (Fkh) transcription factor family, controls hemocyte differentiation of lymph gland through multiple regulatory mechanisms. Jumu maintains the proper differentiation of prohemocytes by cell-autonomously regulating the expression of Col in medullary zone and by non-cell-autonomously preventing the generation of expanded PSC cells. Jumu can also cell-autonomously control the proliferation of PSC cells through positive regulation of dMyc expression. We also show that a deficiency of jumu throughout the lymph gland can induce the differentiation of lamellocytes via activating Toll signaling.
Collapse
Affiliation(s)
- Yangguang Hao
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| |
Collapse
|
45
|
Screening and Analysis of Janelia FlyLight Project Enhancer-Gal4 Strains Identifies Multiple Gene Enhancers Active During Hematopoiesis in Normal and Wasp-Challenged Drosophila Larvae. G3-GENES GENOMES GENETICS 2017; 7:437-448. [PMID: 27913635 PMCID: PMC5295592 DOI: 10.1534/g3.116.034439] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A GFP expression screen has been conducted on >1000 Janelia FlyLight Project enhancer-Gal4 lines to identify transcriptional enhancers active in the larval hematopoietic system. A total of 190 enhancers associated with 87 distinct genes showed activity in cells of the third instar larval lymph gland and hemolymph. That is, gene enhancers were active in cells of the lymph gland posterior signaling center (PSC), medullary zone (MZ), and/or cortical zone (CZ), while certain of the transcriptional control regions were active in circulating hemocytes. Phenotypic analyses were undertaken on 81 of these hematopoietic-expressed genes, with nine genes characterized in detail as to gain- and loss-of-function phenotypes in larval hematopoietic tissues and blood cells. These studies demonstrated the functional requirement of the cut gene for proper PSC niche formation, the hairy, Btk29A, and E2F1 genes for blood cell progenitor production in the MZ domain, and the longitudinals lacking, dFOXO, kayak, cap-n-collar, and delilah genes for lamellocyte induction and/or differentiation in response to parasitic wasp challenge and infestation of larvae. Together, these findings contribute substantial information to our knowledge of genes expressed during the larval stage of Drosophila hematopoiesis and newly identify multiple genes required for this developmental process.
Collapse
|
46
|
El Chamy L, Matt N, Reichhart JM. Advances in Myeloid-Like Cell Origins and Functions in the Model Organism Drosophila melanogaster. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0038-2016. [PMID: 28102122 PMCID: PMC11687447 DOI: 10.1128/microbiolspec.mchd-0038-2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Indexed: 12/22/2022] Open
Abstract
Drosophila has long served as a valuable model for deciphering many biological processes, including immune responses. Indeed, the genetic tractability of this organism is particularly suited for large-scale analyses. Studies performed during the last 3 decades have proven that the signaling pathways that regulate the innate immune response are conserved between Drosophila and mammals. This review summarizes the recent advances on Drosophila hematopoiesis and immune cellular responses, with a particular emphasis on phagocytosis.
Collapse
Affiliation(s)
- Laure El Chamy
- Laboratoire de Génétique de la drosophile et virulence microbienne, UR. EGFEM, Faculté des Sciences, Université Saint-Joseph de Beyrouth, B.P. 17-5208 Mar Mikhaël Beyrouth 1104 2020, Liban
| | - Nicolas Matt
- Université de Strasbourg, UPR 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg Cedex 67084, France
| | - Jean-Marc Reichhart
- Université de Strasbourg, UPR 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg Cedex 67084, France
| |
Collapse
|
47
|
Baril C, Gavory G, Bidla G, Knævelsrud H, Sauvageau G, Therrien M. Human NUP98-HOXA9 promotes hyperplastic growth of hematopoietic tissues in Drosophila. Dev Biol 2016; 421:16-26. [PMID: 27838340 DOI: 10.1016/j.ydbio.2016.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 01/02/2023]
Abstract
Acute myeloid leukemia (AML) is a complex malignancy with poor prognosis. Several genetic lesions can lead to the disease. One of these corresponds to the NUP98-HOXA9 (NA9) translocation that fuses sequences encoding the N-terminal part of NUP98 to those encoding the DNA-binding domain of HOXA9. Despite several studies, the mechanism underlying NA9 ability to induce leukemia is still unclear. To bridge this gap, we sought to functionally dissect NA9 activity using Drosophila. For this, we generated transgenic NA9 fly lines and expressed the oncoprotein during larval hematopoiesis. This markedly enhanced cell proliferation and tissue growth, but did not alter cell fate specification. Moreover, reminiscent to NA9 activity in mammals, strong cooperation was observed between NA9 and the MEIS homolog HTH. Genetic characterization of NA9-induced phenotypes suggested interference with PVR (Flt1-4 RTK homolog) signaling, which is similar to functional interactions observed in mammals between Flt3 and HOXA9 in leukemia. Finally, NA9 expression was also found to induce non-cell autonomous effects, raising the possibility that its leukemia-inducing activity also relies on this property. Together, our work suggests that NA9 ability to induce blood cell expansion is evolutionarily conserved. The amenability of NA9 activity to a genetically-tractable system should facilitate unraveling its molecular underpinnings.
Collapse
Affiliation(s)
- Caroline Baril
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| | - Gwenaëlle Gavory
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| | - Gawa Bidla
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| | - Helene Knævelsrud
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7
| | - Guy Sauvageau
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7; Département de médecine, Université de Montréal, Canada
| | - Marc Therrien
- Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3J7; Département de pathologie et de biologie cellulaire, Université de Montréal, Canada.
| |
Collapse
|
48
|
Letourneau M, Lapraz F, Sharma A, Vanzo N, Waltzer L, Crozatier M. Drosophila hematopoiesis under normal conditions and in response to immune stress. FEBS Lett 2016; 590:4034-4051. [PMID: 27455465 DOI: 10.1002/1873-3468.12327] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/07/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
The emergence of hematopoietic progenitors and their differentiation into various highly specialized blood cell types constitute a finely tuned process. Unveiling the genetic cascades that control blood cell progenitor fate and understanding how they are modulated in response to environmental changes are two major challenges in the field of hematopoiesis. In the last 20 years, many studies have established important functional analogies between blood cell development in vertebrates and in the fruit fly, Drosophila melanogaster. Thereby, Drosophila has emerged as a powerful genetic model for studying mechanisms that control hematopoiesis during normal development or in pathological situations. Moreover, recent advances in Drosophila have highlighted how intricate cell communication networks and microenvironmental cues regulate blood cell homeostasis. They have also revealed the striking plasticity of Drosophila mature blood cells and the presence of different sites of hematopoiesis in the larva. This review provides an overview of Drosophila hematopoiesis during development and summarizes our current knowledge on the molecular processes controlling larval hematopoiesis, both under normal conditions and in response to an immune challenge, such as wasp parasitism.
Collapse
Affiliation(s)
- Manon Letourneau
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Francois Lapraz
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Anurag Sharma
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France.,Department of Biomedical Sciences, NU Centre for Science Education & Research, Nitte University, Mangalore-18, India
| | - Nathalie Vanzo
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Lucas Waltzer
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Centre de Biologie Intégrative, Toulouse Cedex 9, France
| |
Collapse
|
49
|
Morin-Poulard I, Sharma A, Louradour I, Vanzo N, Vincent A, Crozatier M. Vascular control of the Drosophila haematopoietic microenvironment by Slit/Robo signalling. Nat Commun 2016; 7:11634. [PMID: 27193394 PMCID: PMC4874035 DOI: 10.1038/ncomms11634] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 04/15/2016] [Indexed: 12/16/2022] Open
Abstract
Self-renewal and differentiation of mammalian haematopoietic stem cells (HSCs) are controlled by a specialized microenvironment called 'the niche'. In the bone marrow, HSCs receive signals from both the endosteal and vascular niches. The posterior signalling centre (PSC) of the larval Drosophila haematopoietic organ, the lymph gland, regulates blood cell differentiation under normal conditions and also plays a key role in controlling haematopoiesis under immune challenge. Here we report that the Drosophila vascular system also contributes to the lymph gland homoeostasis. Vascular cells produce Slit that activates Robo receptors in the PSC. Robo activation controls proliferation and clustering of PSC cells by regulating Myc, and small GTPase and DE-cadherin activity, respectively. These findings reveal that signals from the vascular system contribute to regulating the rate of blood cell differentiation via the regulation of PSC morphology.
Collapse
Affiliation(s)
- Ismaël Morin-Poulard
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Fédération de Recherche de Biologie de Toulouse, 118 route de Narbonne 31062 Toulouse cedex 9, France
| | - Anurag Sharma
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Fédération de Recherche de Biologie de Toulouse, 118 route de Narbonne 31062 Toulouse cedex 9, France
| | - Isabelle Louradour
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Fédération de Recherche de Biologie de Toulouse, 118 route de Narbonne 31062 Toulouse cedex 9, France
| | - Nathalie Vanzo
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Fédération de Recherche de Biologie de Toulouse, 118 route de Narbonne 31062 Toulouse cedex 9, France
| | - Alain Vincent
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Fédération de Recherche de Biologie de Toulouse, 118 route de Narbonne 31062 Toulouse cedex 9, France
| | - Michèle Crozatier
- Centre de Biologie du Développement, UMR 5547 CNRS/Université Toulouse III and Fédération de Recherche de Biologie de Toulouse, 118 route de Narbonne 31062 Toulouse cedex 9, France
| |
Collapse
|
50
|
Gold KS, Brückner K. Macrophages and cellular immunity in Drosophila melanogaster. Semin Immunol 2016; 27:357-68. [PMID: 27117654 DOI: 10.1016/j.smim.2016.03.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/08/2016] [Indexed: 12/16/2022]
Abstract
The invertebrate Drosophila melanogaster has been a powerful model for understanding blood cell development and immunity. Drosophila is a holometabolous insect, which transitions through a series of life stages from embryo, larva and pupa to adulthood. In spite of this, remarkable parallels exist between Drosophila and vertebrate macrophages, both in terms of development and function. More than 90% of Drosophila blood cells (hemocytes) are macrophages (plasmatocytes), making this highly tractable genetic system attractive for studying a variety of questions in macrophage biology. In vertebrates, recent findings revealed that macrophages have two independent origins: self-renewing macrophages, which reside and proliferate in local microenvironments in a variety of tissues, and macrophages of the monocyte lineage, which derive from hematopoietic stem or progenitor cells. Like vertebrates, Drosophila possesses two macrophage lineages with a conserved dual ontogeny. These parallels allow us to take advantage of the Drosophila model when investigating macrophage lineage specification, maintenance and amplification, and the induction of macrophages and their progenitors by local microenvironments and systemic cues. Beyond macrophage development, Drosophila further serves as a paradigm for understanding the mechanisms underlying macrophage function and cellular immunity in infection, tissue homeostasis and cancer, throughout development and adult life.
Collapse
Affiliation(s)
| | - Katja Brückner
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; Department of Cell and Tissue Biology; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|