1
|
Ghosh G, Das D, Nandi A, De S, Gangappa SN, Prasad M. Ecdysone regulates phagocytic cell fate of epithelial cells in developing Drosophila eggs. J Cell Biol 2025; 224:e202411073. [PMID: 40434296 PMCID: PMC12118371 DOI: 10.1083/jcb.202411073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/01/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Acquisition of nonprofessional phagocytic cell fate plays an important role in sculpting functional metazoan organs and maintaining overall tissue homeostasis. Though physiologically highly relevant, how the normal epithelial cells acquire phagocytic fate is still mostly unclear. We have employed the Drosophila ovary model to demonstrate that the classical ecdysone signaling in the somatic epithelial follicle cells (AFCs) aids the removal of germline nurse cells (NCs) in late oogenesis. Our live-cell imaging data reveal a novel phenomenon wherein collective behavior of 4-5 AFCs is required for clearing a single NC. By employing classical genetics, molecular biology, and yeast one-hybrid assay, we demonstrate that ecdysone modulates the phagocytic disposition of AFCs at two levels. It regulates the epithelial-mesenchymal transition of the AFCs through Serpent and modulates the phagocytic behavior of the AFCs through Croquemort and Draper. Our data provide unprecedented novel molecular insights into how ecdysone signaling reprograms AFCs toward a phagocytic fate.
Collapse
Affiliation(s)
- Gaurab Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Devyan Das
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Abhrajyoti Nandi
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Souvik De
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Sreeramaiah N. Gangappa
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Mohit Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| |
Collapse
|
2
|
Randolph LN, Castiglioni C, Tavian M, Sturgeon CM, Ditadi A. Bloodhounds chasing the origin of blood cells. Trends Cell Biol 2025:S0962-8924(25)00067-4. [PMID: 40221343 DOI: 10.1016/j.tcb.2025.03.003] [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: 10/24/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
The generation of blood cells during embryonic development involves a process resembling lineage reprogramming, where specialized cells within the vasculature become blood forming, or hemogenic. These hemogenic cells undergo rapid transcriptional and morphological changes as they appear to switch from an endothelial to blood identity. What controls this process and the exact nature of the hemogenic cells remains debated, with evidence supporting several hypotheses. In this opinion, we synthesize current knowledge and propose a model reconciling conflicting observations, integrating evolutionary and mechanistic insights into blood cell emergence.
Collapse
Affiliation(s)
- Lauren N Randolph
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Castiglioni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Manuela Tavian
- University of Strasbourg, INSERM UMR-S1109, FMTS, Strasbourg, France
| | - Christopher M Sturgeon
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Ditadi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
3
|
Lee JI, Park S, Park H, Lee Y, Park J, Lee D, Kim MJ, Choe KM. The matrix glycoprotein Papilin maintains the haematopoietic progenitor pool in Drosophila lymph glands. Development 2025; 152:dev204367. [PMID: 40094323 PMCID: PMC12045604 DOI: 10.1242/dev.204367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025]
Abstract
Differentiation of prohaemocytes, the precursors of Drosophila blood cells (haemocytes), and the release of haemocytes from the lymph gland, a major larval haematopoietic organ, are vital responses to wasp infestation or tissue degeneration. Although cells and extracellular matrix (ECM) in the lymph gland are known to play a crucial role in haemocyte differentiation, the underlying mechanisms remain unclear. Here, we show that the matrix glycoprotein Papilin (Ppn) is essential for maintaining the prohaemocyte population in lymph glands. In Ppn-depleted larvae, haemocyte differentiation increased with a reduction in the prohaemocyte-containing medullary zone, and lymph gland lobes dispersed prematurely. Ppn was synthesised by plasmatocytes, forming lamellae mainly in the medullary zone. Microbial infection or wasp infestation disrupted the Ppn meshwork within lymph glands. Ppn colocalised with collagen, laminin, nidogen and perlecan. Ppn depletion disrupted the ECM structure, including perlecan organisation. Phenotypes caused by Ppn depletion were partially rescued by perlecan overexpression or inactivation of the epidermal growth factor receptor pathway. Thus, Ppn is crucial for maintaining lymph gland architecture and regulating haemocyte differentiation, highlighting an intricate interaction between the ECM and signalling pathways in haematopoiesis.
Collapse
Affiliation(s)
- Jae-In Lee
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Sumin Park
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Hyunji Park
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Youngbin Lee
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - JinYoung Park
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Donghoon Lee
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Moon Jong Kim
- Department of Life Science, Gachon University, Seongnam 13120, South Korea
| | - Kwang-Min Choe
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| |
Collapse
|
4
|
Marcetteau J, Duarte P, Leitão AB, Sucena É. Transdifferentiation of plasmatocytes to crystal cells in the lymph gland of Drosophila melanogaster. EMBO Rep 2025; 26:2077-2097. [PMID: 40075235 PMCID: PMC12019564 DOI: 10.1038/s44319-025-00366-z] [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: 02/06/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025] Open
Abstract
Under homeostatic conditions, haematopoiesis in Drosophila larvae occurs in the lymph gland and sessile haemocyte clusters to produce two functionally and morphologically different cells: plasmatocytes and crystal cells. It is well-established that in the lymph gland both cell types stem from a binary decision of the medullary prohaemocyte precursors. However, in sessile clusters and dorsal vessel, crystal cells have been shown to originate from the transdifferentiation of plasmatocytes in a Notch/Serrate-dependent manner. We show that transdifferentiation occurs also in the lymph gland. In vivo phagocytosis assays confirm that cortical plasmatocytes are functionally differentiated phagocytic cells. We uncover a double-positive population in the cortical zone that lineage-tracing and long-term live imaging experiments show will differentiate into crystal cells. The reduction of Notch levels within the lymph gland plasmatocyte population reduces crystal cell number. This extension of a transdifferentiation mechanism reinforces the growing role of haematopoietic plasticity in maintaining homeostasis in Drosophila and vertebrate systems. Future work should test the regulation and relative contribution of these two processes under different immunological and/or metabolic conditions.
Collapse
Affiliation(s)
- Julien Marcetteau
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Patrícia Duarte
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | | | - Élio Sucena
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal.
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Edifício C2, Campo Grande, 1749-016, Lisbon, Portugal.
- cE3c: Centre for Ecology, Evolution and Environmental Changes, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal.
| |
Collapse
|
5
|
Hirata M, Nomura T, Inoue YH. Anti-Tumor Effects of Cecropin A and Drosocin Incorporated into Macrophage-like Cells Against Hematopoietic Tumors in Drosophila mxc Mutants. Cells 2025; 14:389. [PMID: 40136638 PMCID: PMC11940895 DOI: 10.3390/cells14060389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Five major antimicrobial peptides (AMPs) in Drosophila are induced in multiple sex combs (mxc) mutant larvae harboring lymph gland (LG) tumors, and they exhibit anti-tumor effects. The effects of other well-known AMPs, Cecropin A and Drosocin, remain unexplored. We investigated the tumor-elimination mechanism of these AMPs. A half-dose reduction in either the Toll or Imd gene reduced the induction of these AMPs and enhanced tumor growth in mxcmbn1 mutant larvae, indicating that their anti-tumor effects depend on the innate immune pathway. Overexpression of these AMPs in the fat body suppressed tumor growth without affecting cell proliferation. Apoptosis was promoted in the mutant but not in normal LGs. Conversely, knockdown of them inhibited apoptosis and enhanced tumor growth; therefore, they inhibit LG tumor growth by inducing apoptosis. The AMPs from the fat body were incorporated into the hemocytes of mutant but not normal larvae. Another AMP, Drosomycin, was taken up via phagocytosis factors. Enhanced phosphatidylserine signals were observed on the tumor surface. Inhibition of the signals exposed on the cell surface enhanced tumor growth. AMPs may target phosphatidylserine in tumors to induce apoptosis and execute their tumor-specific effects. AMPs could be beneficial anti-cancer drugs with minimal side effects for clinical development.
Collapse
Affiliation(s)
- Marina Hirata
- Biomedical Research Center, Kyoto Institute of Technology, Kyoto 606-0962, Japan (T.N.)
- Graduate School of Science and Technology, Kyoto Institute of Technology, Kyoto 606-8585, Japan
| | - Tadashi Nomura
- Biomedical Research Center, Kyoto Institute of Technology, Kyoto 606-0962, Japan (T.N.)
- Graduate School of Science and Technology, Kyoto Institute of Technology, Kyoto 606-8585, Japan
| | - Yoshihiro H. Inoue
- Biomedical Research Center, Kyoto Institute of Technology, Kyoto 606-0962, Japan (T.N.)
| |
Collapse
|
6
|
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
|
7
|
Liu N, Nakano A. A crucial new aspect of cardiac morphogenesis: endocardial hematopoiesis. Front Physiol 2024; 15:1525985. [PMID: 39720314 PMCID: PMC11667111 DOI: 10.3389/fphys.2024.1525985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Affiliation(s)
- Norika Liu
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Atsushi Nakano
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
- David Geffen Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
8
|
Kwon SY, Chan K, Stofanko M, Chan KH, Badenhorst P. Abrupt-mediated control of ninjurins regulates Drosophila sessile haemocyte compartments. Development 2024; 151:dev202977. [PMID: 39545919 DOI: 10.1242/dev.202977] [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: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Macrophage-like cells called haemocytes are key effectors of Drosophila cellular innate immune function. Larval haemocytes exist either in circulation or localize to segmentally repeated sessile haemocyte compartments (SHCs). While numerous functions have been proposed for SHCs, the mechanisms directing haemocytes to them are unclear. Here, we have exploited the developmentally regulated dispersal of SHCs that occurs at pupariation to identify the Abrupt (Ab) transcription factor (TF) and ninjurin cell-adhesion molecules as regulators of haemocyte recruitment to SHCs. We show that larval haemocytes express ninjurins, which are required for targeting haemocytes to SHCs. However, at pupariation, ecdysteroid signalling stimulates Ab expression, which collaborates with TFs, including Blimp-1 and Hr3, to repress ninjurins and disperse haemocytes. We observe that experimental manipulations that antagonize ninjurin function in larval haemocytes cause premature SHC dispersal, while stabilization of ninjurins in haemocytes blocks developmentally regulated SHC remodelling and increases sensitivity to immune challenges. Cumulatively, our data indicate that control of ninjurin activity provides a common target through which diverse developmental, environmental and immune stimuli can be integrated to control haemocyte dispersal and immune function.
Collapse
Affiliation(s)
- So Yeon Kwon
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Kimberly Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Martin Stofanko
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Ka Hei Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Paul Badenhorst
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
9
|
Monticelli S, Giangrande A. Evolutionary Conservation of the Gcm/Glide Cascade: Of Glia and Beyond. BRAIN, BEHAVIOR AND EVOLUTION 2024; 100:58-66. [PMID: 39586239 DOI: 10.1159/000542753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Glia represent a major cell population of the nervous system, and they take part in virtually any process sustaining the development, the functioning, and the pathology of the nervous system. Glial cells diversified significantly during evolution and distinct signals have been adopted to initiate glial development in mammals as compared to flies. In the invertebrate model Drosophila melanogaster, the transcription factor Gcm is necessary and sufficient to generate glial cells. Although Gcm orthologs have been found in protostomes and deuterostomes, they do not act in glial fate commitment as in flies, calling for further investigations of the evolutionarily conserved role of Gcm. SUMMARY Here, we review the impact of the fly Gcm transcription factor in the differentiation of phagocytic competent cells inside and outside the nervous system, glia, and macrophages, respectively. Then, we discuss the evolutionary conservation of Gcm and the neural/nonneural functions of Gcm orthologs. Finally, we present a recent work from Pavlidaki et al. [Cell Rep. 2022;41(3):111506] showing that the Gcm cascade is conserved from fly macrophages to mammalian microglia to counteract acute and chronic inflammation. KEY MESSAGES Gcm has an ancestral role in immunity, and its anti-inflammatory effect is evolutionarily conserved. This opens new avenues to assess Gcm function in other species/animal models, its potential involvement in inflammation-related processes, such as regeneration, and to expand the investigation on glia evolution.
Collapse
Affiliation(s)
- Sara Monticelli
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, Illkirch, France
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, Illkirch, France
| | - Angela Giangrande
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, UMR-S 1258, Illkirch, France
- Université de Strasbourg, IGBMC UMR 7104- UMR-S 1258, Illkirch, France
| |
Collapse
|
10
|
Deichsel S, Frankenreiter L, Fechner J, Gahr BM, Zimmermann M, Mastel H, Preis I, Preiss A, Nagel AC. Inhibition of the Notch signal transducer CSL by Pkc53E-mediated phosphorylation to fend off parasitic immune challenge in Drosophila. eLife 2024; 12:RP89582. [PMID: 39503739 PMCID: PMC11540305 DOI: 10.7554/elife.89582] [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] [Indexed: 11/08/2024] Open
Abstract
Notch signalling activity regulates hematopoiesis in Drosophila and vertebrates alike. Parasitoid wasp infestation of Drosophila larvae, however, requires a timely downregulation of Notch activity to allow the formation of encapsulation-active blood cells. Here, we show that the Drosophila CSL transcription factor Suppressor of Hairless [Su(H)] is phosphorylated at Serine 269 in response to parasitoid wasp infestation. As this phosphorylation interferes with the DNA binding of Su(H), it reversibly precludes its activity. Accordingly, phospho-deficient Su(H)S269A mutants are immune-compromised. A screen for kinases involved in Su(H) phosphorylation identified Pkc53E, required for normal hematopoiesis as well as for parasitoid immune response. Genetic and molecular interactions support the specificity of the Su(H)-Pkc53E relationship. Moreover, phorbol ester treatment inhibits Su(H) activity in vivo and in human cell culture. We conclude that Pkc53E targets Su(H) during parasitic wasp infestation, thereby remodelling the blood cell population required for wasp egg encapsulation.
Collapse
Affiliation(s)
- Sebastian Deichsel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Department of Medical Genetics and Applied Genomics, University of TübingenTübingenGermany
| | - Lisa Frankenreiter
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Johannes Fechner
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Institute of Biomedical Genetics (IBMG), University of StuttgartStuttgartGermany
| | - Bernd M Gahr
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Department of Internal Medicine II, Molecular Cardiology, University of UlmUlmGermany
| | - Mirjam Zimmermann
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Helena Mastel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Irina Preis
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Anette Preiss
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Anja C Nagel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| |
Collapse
|
11
|
Rai P, Bergmann A. Unraveling the intricate link between cell death and neuroinflammation using Drosophila as a model. Front Cell Dev Biol 2024; 12:1479864. [PMID: 39411483 PMCID: PMC11474694 DOI: 10.3389/fcell.2024.1479864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Protein aggregation is a common pathological occurrence in neurodegenerative diseases. This often leads to neuroinflammation, which exacerbates the aggregation and progression of diseases like Parkinson's and Alzheimer's. Here, we focus on immune responses and neurotoxicity in a Parkinson's disease model in Drosophila. Mutations in the SNCA gene that encodes the alpha (α)-Synuclein protein have been linked to familial Parkinson's disease, disrupting autophagy regulation in neuronal cells and promoting the formation of Lewy bodies, a hallmark of Parkinson's pathology. This results in the loss of dopaminergic neurons, manifesting as movement disorders. α-Synuclein aggregation triggers innate immune responses by activating microglial cells, leading to phagocytic activity and the expression of neuroprotective antimicrobial peptides (AMPs). However, sustained AMP expression or chronic inflammation resulting from inadequate microglial phagocytosis can induce neuronal toxicity and apoptosis, leading to severe dopaminergic neuron loss. This review underscores the mechanistic connection between immune response pathways and α-Synuclein-mediated neurodegeneration using Drosophila models. Furthermore, we extensively explore factors influencing neuroinflammation and key immune signaling pathways implicated in neurodegenerative diseases, particularly Parkinson's disease. Given the limited success of traditional treatments, recent research has focused on therapies targeting inflammatory signaling pathways. Some of these approaches have shown promising results in animal models and clinical trials. We provide an overview of current therapeutic strategies showing potential in treating neurodegenerative diseases, offering new avenues for future research and treatment development.
Collapse
|
12
|
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
|
13
|
Doyle EH, Vaughan HJ, Mariani SA. From drosophila to humans: a journey through macrophage development. Exp Hematol 2024; 136:104272. [PMID: 38972565 DOI: 10.1016/j.exphem.2024.104272] [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: 05/04/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
Macrophages are fascinating immune cells involved in a variety of processes in both health and disease. Although they were first discovered and characterized by their functions as professional phagocytes and antigen-presenting cells, it is now clear that macrophages have multiple roles within embryonic development, tissue homeostasis, regulation of inflammation, and host response to pathogens and tissue insults. Interestingly, macrophages, or macrophage-like cells, exist in a variety of organisms, from echinoderms to humans, and are present also in species that lack an adaptive immune system or hematopoietic stem cells (HSCs). In mammals, macrophages can be generated from bone marrow precursors through a monocyte intermediate, but it is now known that they are also generated during earlier hematopoietic waves in the embryo. Seeding a variety of tissues at different times, macrophages contribute to embryonic organogenesis and tissue homeostasis. Interestingly, in species where embryonic macrophages are generated before HSC specification, they seem to be an important component of the HSC generative microenvironment. There are many excellent reviews reporting the current knowledge on the ontogeny and functions of macrophages in adult tissues. Here, we aim to summarize the current knowledge on the development and functions of embryonic macrophages across the most used animal models, with a special focus on developmental hematopoiesis.
Collapse
Affiliation(s)
- Eva H Doyle
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hollie J Vaughan
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Samanta A Mariani
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
14
|
Shin M, Chang E, Lee D, Kim N, Cho B, Cha N, Koranteng F, Song JJ, Shim J. Drosophila immune cells transport oxygen through PPO2 protein phase transition. Nature 2024; 631:350-359. [PMID: 38926577 PMCID: PMC11236712 DOI: 10.1038/s41586-024-07583-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/17/2024] [Indexed: 06/28/2024]
Abstract
Insect respiration has long been thought to be solely dependent on an elaborate tracheal system without assistance from the circulatory system or immune cells1,2. Here we describe that Drosophila crystal cells-myeloid-like immune cells called haemocytes-control respiration by oxygenating Prophenoloxidase 2 (PPO2) proteins. Crystal cells direct the movement of haemocytes between the trachea of the larval body wall and the circulation to collect oxygen. Aided by copper and a neutral pH, oxygen is trapped in the crystalline structures of PPO2 in crystal cells. Conversely, PPO2 crystals can be dissolved when carbonic anhydrase lowers the intracellular pH and then reassembled into crystals in cellulo by adhering to the trachea. Physiologically, larvae lacking crystal cells or PPO2, or those expressing a copper-binding mutant of PPO2, display hypoxic responses under normoxic conditions and are susceptible to hypoxia. These hypoxic phenotypes can be rescued by hyperoxia, expression of arthropod haemocyanin or prevention of larval burrowing activity to expose their respiratory organs. Thus, we propose that insect immune cells collaborate with the tracheal system to reserve and transport oxygen through the phase transition of PPO2 crystals, facilitating internal oxygen homeostasis in a process that is comparable to vertebrate respiration.
Collapse
Affiliation(s)
- Mingyu Shin
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Eunji Chang
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Daewon Lee
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Nayun Kim
- Department of Biological Sciences, KI for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Bumsik Cho
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Nuri Cha
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Ferdinand Koranteng
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Ji-Joon Song
- Department of Biological Sciences, KI for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jiwon Shim
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Natural Science, Hanyang University, Seoul, Republic of Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea.
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Edwards AJ, Beltz BS. Longitudinal tracking of hemocyte populations in vivo indicates lineage relationships and supports neural progenitor identity in adult neurogenesis. Neural Dev 2024; 19:7. [PMID: 38902780 PMCID: PMC11191286 DOI: 10.1186/s13064-024-00185-3] [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: 03/29/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Adult neurogenesis, which takes place in both vertebrate and invertebrate species, is the process by which new neurons are born and integrated into existing functional neural circuits, long after embryonic development. Most studies in mammals suggest that self-renewing stem cells are the source of the new neurons, although the extent of self-renewal is a matter of debate. In contrast, research in the crayfish Procambarus clarkii has demonstrated that the neural progenitors producing adult-born neurons are capable of both self-renewing and consuming (non-self-renewing) divisions. However, self-renewing divisions are relatively rare, and therefore the production of adult-born neurons depends heavily on progenitors that are not replenishing themselves. Because the small pool of neural progenitors in the neurogenic niche is never exhausted throughout the long lives of these animals, we hypothesized that there must also be an extrinsic source of these cells. It was subsequently demonstrated that the neural progenitors originate in hemocytes (blood cells) produced by the immune system that travel in the circulation before ultimately integrating into niches where the neural lineage begins. The current study examines the developmental lineage of the three hemocyte types - hyaline (HC), semigranular (SGC) and granular (GC) cells - with the goal of understanding the origins of the progenitor cells that produce adult-born neurons. Longstanding qualitative metrics for hemocyte classification were validated quantitatively. Then, in a longitudinal study, proliferation markers were used to label the hemocytes in vivo, followed by sampling the circulating hemocyte population over the course of two months. Hemolymph samples were taken at intervals to track the frequencies of the different hemocyte types. These data reveal sequential peaks in the relative frequencies of HCs, SGCs and GCs, which were identified using qualitative and quantitative measures. These findings suggest that the three hemocyte types comprise a single cellular lineage that occurs in the circulation, with each type as a sequential progressive stage in hemocyte maturation beginning with HCs and ending with GCs. When combined with previously published data, this timeline provides additional evidence that HCs serve as the primary neural progenitor during adult neurogenesis in P. clarkii.
Collapse
Affiliation(s)
- Alex J Edwards
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Barbara S Beltz
- Neuroscience Department, Wellesley College, Wellesley, MA, 02481, USA.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Deichsel S, Gahr BM, Mastel H, Preiss A, Nagel AC. Numerous Serine/Threonine Kinases Affect Blood Cell Homeostasis in Drosophila melanogaster. Cells 2024; 13:576. [PMID: 38607015 PMCID: PMC11011202 DOI: 10.3390/cells13070576] [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: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Blood cells in Drosophila serve primarily innate immune responses. Various stressors influence blood cell homeostasis regarding both numbers and the proportion of blood cell types. The principle molecular mechanisms governing hematopoiesis are conserved amongst species and involve major signaling pathways like Notch, Toll, JNK, JAK/Stat or RTK. Albeit signaling pathways generally rely on the activity of protein kinases, their specific contribution to hematopoiesis remains understudied. Here, we assess the role of Serine/Threonine kinases with the potential to phosphorylate the transcription factor Su(H) in crystal cell homeostasis. Su(H) is central to Notch signal transduction, and its inhibition by phosphorylation impedes crystal cell formation. Overall, nearly twenty percent of all Drosophila Serine/Threonine kinases were studied in two assays, global and hemocyte-specific overexpression and downregulation, respectively. Unexpectedly, the majority of kinases influenced crystal cell numbers, albeit only a few were related to hematopoiesis so far. Four kinases appeared essential for crystal cell formation, whereas most kinases restrained crystal cell development. This group comprises all kinase classes, indicative of the complex regulatory network underlying blood cell homeostasis. The rather indiscriminative response we observed opens the possibility that blood cells measure their overall phospho-status as a proxy for stress-signals, and activate an adaptive immune response accordingly.
Collapse
Affiliation(s)
- Sebastian Deichsel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Bernd M. Gahr
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Helena Mastel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Anette Preiss
- Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Anja C. Nagel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
19
|
Drewell RA, Klonaros D, Dresch JM. Transcription factor expression landscape in Drosophila embryonic cell lines. BMC Genomics 2024; 25:307. [PMID: 38521929 PMCID: PMC10960990 DOI: 10.1186/s12864-024-10241-1] [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: 01/26/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Transcription factor (TF) proteins are a key component of the gene regulatory networks that control cellular fates and function. TFs bind DNA regulatory elements in a sequence-specific manner and modulate target gene expression through combinatorial interactions with each other, cofactors, and chromatin-modifying proteins. Large-scale studies over the last two decades have helped shed light on the complex network of TFs that regulate development in Drosophila melanogaster. RESULTS Here, we present a detailed characterization of expression of all known and predicted Drosophila TFs in two well-established embryonic cell lines, Kc167 and S2 cells. Using deep coverage RNA sequencing approaches we investigate the transcriptional profile of all 707 TF coding genes in both cell types. Only 103 TFs have no detectable expression in either cell line and 493 TFs have a read count of 5 or greater in at least one of the cell lines. The 493 TFs belong to 54 different DNA-binding domain families, with significant enrichment of those in the zf-C2H2 family. We identified 123 differentially expressed genes, with 57 expressed at significantly higher levels in Kc167 cells than S2 cells, and 66 expressed at significantly lower levels in Kc167 cells than S2 cells. Network mapping reveals that many of these TFs are crucial components of regulatory networks involved in cell proliferation, cell-cell signaling pathways, and eye development. CONCLUSIONS We produced a reference TF coding gene expression dataset in the extensively studied Drosophila Kc167 and S2 embryonic cell lines, and gained insight into the TF regulatory networks that control the activity of these cells.
Collapse
Affiliation(s)
- Robert A Drewell
- Biology Department, Clark University, 950 Main Street, Worcester, MA, 01610, USA.
| | - Daniel Klonaros
- Biology Department, Clark University, 950 Main Street, Worcester, MA, 01610, USA
| | - Jacqueline M Dresch
- Biology Department, Clark University, 950 Main Street, Worcester, MA, 01610, USA
| |
Collapse
|
20
|
de Almeida BP, Schaub C, Pagani M, Secchia S, Furlong EEM, Stark A. Targeted design of synthetic enhancers for selected tissues in the Drosophila embryo. Nature 2024; 626:207-211. [PMID: 38086418 PMCID: PMC10830412 DOI: 10.1038/s41586-023-06905-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
Enhancers control gene expression and have crucial roles in development and homeostasis1-3. However, the targeted de novo design of enhancers with tissue-specific activities has remained challenging. Here we combine deep learning and transfer learning to design tissue-specific enhancers for five tissues in the Drosophila melanogaster embryo: the central nervous system, epidermis, gut, muscle and brain. We first train convolutional neural networks using genome-wide single-cell assay for transposase-accessible chromatin with sequencing (ATAC-seq) datasets and then fine-tune the convolutional neural networks with smaller-scale data from in vivo enhancer activity assays, yielding models with 13% to 76% positive predictive value according to cross-validation. We designed and experimentally assessed 40 synthetic enhancers (8 per tissue) in vivo, of which 31 (78%) were active and 27 (68%) functioned in the target tissue (100% for central nervous system and muscle). The strategy of combining genome-wide and small-scale functional datasets by transfer learning is generally applicable and should enable the design of tissue-, cell type- and cell state-specific enhancers in any system.
Collapse
Affiliation(s)
- Bernardo P de Almeida
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
- InstaDeep, Paris, France
| | - Christoph Schaub
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Michaela Pagani
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Stefano Secchia
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Eileen E M Furlong
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Alexander Stark
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria.
- Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria.
| |
Collapse
|
21
|
Brooks EC, Zeidler MP, Ong ACM, Evans IR. Macrophage subpopulation identity in Drosophila is modulated by apoptotic cell clearance and related signalling pathways. Front Immunol 2024; 14:1310117. [PMID: 38283366 PMCID: PMC10811221 DOI: 10.3389/fimmu.2023.1310117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
In Drosophila blood, plasmatocytes of the haemocyte lineage represent the functional equivalent of vertebrate macrophages and have become an established in vivo model with which to study macrophage function and behaviour. However, the use of plasmatocytes as a macrophage model has been limited by a historical perspective that plasmatocytes represent a homogenous population of cells, in contrast to the high levels of heterogeneity of vertebrate macrophages. Recently, a number of groups have reported transcriptomic approaches which suggest the existence of plasmatocyte heterogeneity, while we identified enhancer elements that identify subpopulations of plasmatocytes which exhibit potentially pro-inflammatory behaviours, suggesting conservation of plasmatocyte heterogeneity in Drosophila. These plasmatocyte subpopulations exhibit enhanced responses to wounds and decreased rates of efferocytosis when compared to the overall plasmatocyte population. Interestingly, increasing the phagocytic requirement placed upon plasmatocytes is sufficient to decrease the size of these plasmatocyte subpopulations in the embryo. However, the mechanistic basis for this response was unclear. Here, we examine how plasmatocyte subpopulations are modulated by apoptotic cell clearance (efferocytosis) demands and associated signalling pathways. We show that loss of the phosphatidylserine receptor Simu prevents an increased phagocytic burden from modulating specific subpopulation cells, while blocking other apoptotic cell receptors revealed no such rescue. This suggests that Simu-dependent efferocytosis is specifically involved in determining fate of particular subpopulations. Supportive of our original finding, mutations in amo (the Drosophila homolog of PKD2), a calcium-permeable channel which operates downstream of Simu, phenocopy simu mutants. Furthermore, we show that Amo is involved in the acidification of the apoptotic cell-containing phagosomes, suggesting that this reduction in pH may be associated with macrophage reprogramming. Additionally, our results also identify Ecdysone receptor signalling, a pathway related to control of cell death during developmental transitions, as a controller of plasmatocyte subpopulation identity. Overall, these results identify fundamental pathways involved in the specification of plasmatocyte subpopulations and so further validate Drosophila plasmatocytes as a heterogeneous population of macrophage-like cells within this important developmental and immune model.
Collapse
Affiliation(s)
- Elliot C. Brooks
- School of Medicine and Population Health and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Martin P. Zeidler
- School of Biosciences and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Albert C. M. Ong
- School of Medicine and Population Health and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Iwan R. Evans
- School of Medicine and Population Health and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
22
|
Zhang K, Man X, Hu X, Tan P, Su J, Abbas MN, Cui H. GATA binding protein 6 regulates apoptosis in silkworms through interaction with poly (ADP-ribose) polymerase. Int J Biol Macromol 2024; 256:128515. [PMID: 38040165 DOI: 10.1016/j.ijbiomac.2023.128515] [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: 10/08/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
The GATA family of genes plays various roles in crucial biological processes, such as development, cell differentiation, and disease progression. However, the roles of GATA in insects have not been thoroughly explored. In this study, a genome-wide characterization of the GATA gene family in the silkworm, Bombyx mori, was conducted, revealing lineage-specific expression profiles. Notably, GATA6 is ubiquitously expressed across various developmental stages and tissues, with predominant expression in the midgut, ovaries, and Malpighian tubules. Overexpression of GATA6 inhibits cell growth and promotes apoptosis, whereas, in contrast, knockdown of PARP mitigates the apoptotic effects driven by GATA6 overexpression. Co-immunoprecipitation (co-IP) has demonstrated that GATA6 can interact with Poly (ADP-ribose) polymerase (PARP), suggesting that GATA6 may induce cell apoptosis by activating the enzyme's activity. These findings reveal a dynamic and regulatory relationship between GATA6 and PARP, suggesting a potential role for GATA6 as a key regulator in apoptosis through its interaction with PARP. This research deepens the understanding of the diverse roles of the GATA family in insects, shedding light on new avenues for studies in sericulture and pest management.
Collapse
Affiliation(s)
- Kui Zhang
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China.
| | - Xu Man
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Peng Tan
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Jingjing Su
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China.
| |
Collapse
|
23
|
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
|
24
|
Abstract
The Nucleosome Remodeling and Deacetylating Complex (NuRD) is ubiquitously expressed in all metazoans. It combines nucleosome remodeling and histone deacetylating activities to generate inaccessible chromatin structures and to repress gene transcription. NuRD is involved in the generation and maintenance of a wide variety of lineage-specific gene expression programs during differentiation and in differentiated cells. A close cooperation with a large number of lineage-specific transcription factors is key to allow NuRD to function in many distinct differentiation contexts. The molecular nature of this interplay between transcription factors and NuRD is complex and not well understood. This review uses hematopoiesis as a paradigm to highlight recent advances in our understanding of how transcription factors and NuRD cooperate at the molecular level during differentiation. A comparison of vertebrate and invertebrate systems serves to identify the conserved and fundamental concepts guiding functional interactions between transcription factors and NuRD. We also discuss how the transcription factor-NuRD axis constitutes a potential therapeutic target for the treatment of hemoglobinopathies.
Collapse
Affiliation(s)
- Jonathan Lenz
- Institute for Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University Marburg, Marburg, Germany
| | - Alexander Brehm
- Institute for Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
25
|
Klonaros D, Dresch JM, Drewell RA. Transcriptome profile in Drosophila Kc and S2 embryonic cell lines. G3 (BETHESDA, MD.) 2023; 13:jkad054. [PMID: 36869676 PMCID: PMC10151398 DOI: 10.1093/g3journal/jkad054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/04/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023]
Abstract
Drosophila melanogaster cell lines are an important resource for a range of studies spanning genomics, molecular genetics, and cell biology. Amongst these valuable lines are Kc167 (Kc) and Schneider 2 (S2) cells, which were originally isolated in the late 1960s from embryonic sources and have been used extensively to investigate a broad spectrum of biological activities including cell-cell signaling and immune system function. Whole-genome tiling microarray analysis of total RNA from these two cell types was performed as part of the modENCODE project over a decade ago and revealed that they share a number of gene expression features. Here, we expand on these earlier studies by using deep-coverage RNA-sequencing approaches to investigate the transcriptional profile in Kc and S2 cells in detail. Comparison of the transcriptomes reveals that ∼75% of the 13,919 annotated genes are expressed at a detectable level in at least one of the cell lines, with the majority of these genes expressed at high levels in both cell lines. Despite the overall similarity of the transcriptional landscape in the two cell types, 2,588 differentially expressed genes are identified. Many of the genes with the largest fold change are known only by their "CG" designations, indicating that the molecular control of Kc and S2 cell identity may be regulated in part by a cohort of relatively uncharacterized genes. Our data also indicate that both cell lines have distinct hemocyte-like identities, but share active signaling pathways and express a number of genes in the network responsible for dorsal-ventral patterning of the early embryo.
Collapse
Affiliation(s)
- Daniel Klonaros
- Biology Department, Clark University, 950 Main Street, Worcester, MA 01610, USA
| | - Jacqueline M Dresch
- Biology Department, Clark University, 950 Main Street, Worcester, MA 01610, USA
| | - Robert A Drewell
- Biology Department, Clark University, 950 Main Street, Worcester, MA 01610, USA
| |
Collapse
|
26
|
Adegoke A, Ribeiro JMC, Brown S, Smith RC, Karim S. Rickettsia parkeri hijacks tick hemocytes to manipulate cellular and humoral transcriptional responses. Front Immunol 2023; 14:1094326. [PMID: 36845157 PMCID: PMC9950277 DOI: 10.3389/fimmu.2023.1094326] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/16/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Blood-feeding arthropods rely on robust cellular and humoral immunity to control pathogen invasion and replication. Tick hemocytes produce factors that can facilitate or suppress microbial infection and pathogenesis. Despite the importance of hemocytes in regulating microbial infection, understanding of their basic biology and molecular mechanisms remains limited. Methods Here we combined histomorphology and functional analysis to identify five distinct phagocytic and non-phagocytic hemocyte populations circulating within the Gulf Coast tick Amblyomma maculatum. Results and discussion Depletion of phagocytic hemocytes using clodronate liposomes revealed their function in eliminating bacterial infection. We provide the first direct evidence that an intracellular tick-borne pathogen, Rickettsia parkeri, infects phagocytic hemocytes in Am. maculatum to modify tick cellular immune responses. A hemocyte-specific RNA-seq dataset generated from hemocytes isolated from uninfected and R. parkeri-infected partially blood-fed ticks generated ~40,000 differentially regulated transcripts, >11,000 of which were immune genes. Silencing two differentially regulated phagocytic immune marker genes (nimrod B2 and eater-two Drosophila homologs), significantly reduced hemocyte phagocytosis. Conclusion Together, these findings represent a significant step forward in understanding how hemocytes regulate microbial homeostasis and vector competence.
Collapse
Affiliation(s)
- Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Sidney Brown
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
27
|
Luo W, Liu S, Zhang F, Zhao L, Su Y. Metabolic strategy of macrophages under homeostasis or immune stress in Drosophila. MARINE LIFE SCIENCE & TECHNOLOGY 2022; 4:291-302. [PMID: 37073169 PMCID: PMC10077226 DOI: 10.1007/s42995-022-00134-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/06/2022] [Indexed: 05/03/2023]
Abstract
Macrophages are well known for their phagocytic functions in innate immunity across species. In mammals, they rapidly consume a large amount of energy by shifting their metabolism from mitochondrial oxidative phosphorylation toward aerobic glycolysis, to perform the effective bactericidal function upon infection. Meanwhile, they strive for sufficient energy resources by restricting systemic metabolism. In contrast, under nutrient deprivation, the macrophage population is down-regulated to save energy for survival. Drosophila melanogaster possesses a highly conserved and comparatively simple innate immune system. Intriguingly, recent studies have shown that Drosophila plasmatocytes, the macrophage-like blood cells, adopt comparable metabolic remodeling and signaling pathways to achieve energy reassignment when challenged by pathogens, indicating the conservation of such metabolic strategies between insects and mammals. Here, focusing on Drosophila macrophages (plasmatocytes), we review recent advances regarding their comprehensive roles in local or systemic metabolism under homeostasis or stress, emphasizing macrophages as critical players in the crosstalk between the immune system and organic metabolism from a Drosophila perspective.
Collapse
Affiliation(s)
- Wang Luo
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| | - Sumin Liu
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| | - Fang Zhang
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| | - Long Zhao
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- Fisheries College, Ocean University of China, Qingdao, 266003 China
- Key Laboratory of Mariculture (OUC), Ministry of Education, Qingdao, 266003 China
| | - Ying Su
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003 China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003 China
| |
Collapse
|
28
|
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
|
29
|
Bakopoulos D, Whisstock JC, Warr CG, Johnson TK. Macrophage self‐renewal is regulated by transient expression of
PDGF‐ and VEGF‐related factor 2. FEBS J 2022; 289:3735-3751. [DOI: 10.1111/febs.16364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/24/2021] [Accepted: 01/19/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Daniel Bakopoulos
- School of Biological Sciences Monash University Clayton Vic. Australia
| | - James C. Whisstock
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University Clayton Vic. Australia
- Department of Biochemistry and Molecular Biology Monash University Clayton Vic. Australia
| | - Coral G. Warr
- School of Biological Sciences Monash University Clayton Vic. Australia
- School of Molecular Sciences La Trobe University Bundoora Vic. Australia
| | - Travis K. Johnson
- School of Biological Sciences Monash University Clayton Vic. Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University Clayton Vic. Australia
| |
Collapse
|
30
|
Kapoor A, Padmavathi A, Madhwal S, Mukherjee T. Dual control of dopamine in Drosophila myeloid-like progenitor cell proliferation and regulation of lymph gland growth. EMBO Rep 2022; 23:e52951. [PMID: 35476897 PMCID: PMC9171693 DOI: 10.15252/embr.202152951] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 11/09/2022] Open
Abstract
In Drosophila, definitive haematopoiesis takes place in a specialized organ termed "lymph gland". It harbours multi-potent stem-like blood progenitor cells whose development controls overall growth of this haematopoietic tissue and formation of mature blood cells. With respect to its development, neurotransmitters have emerged as potent regulators of blood-progenitor cell development and function. In this study, we extend our understanding of neurotransmitters and show that progenitors are self-sufficient with regard to synthesizing dopamine, a well-established neurotransmitter. These cells also have modules for dopamine sensing through the receptor and transporter. We found that modulating expression of these components in progenitor cells affected lymph gland growth, which suggested growth-promoting function of dopamine in blood-progenitor cells. Cell-cycle analysis of developing lymph glands revealed an unexpected requirement for intracellular dopamine in moderating the progression of early progenitor cells from S to G2 phase of the cell cycle, while activation of dopamine receptor signalling later in development regulated their progression from G2 and entry into mitosis. The dual capacity in which dopamine operated, first intracellularly to coordinate S/G2 transition and later extracellularly in G2/M transition, was critical for the growth of the lymph gland. Overall, the data presented highlight a novel non-canonical use of dopamine in the myeloid system that reveals an uncharacterized function of intracellular dopamine in cell-cycle phasing with outcomes on haematopoietic growth and immunity as well.
Collapse
Affiliation(s)
- Ankita Kapoor
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Achalla Padmavathi
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India
| | - Sukanya Madhwal
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India.,Manipal Academy of Higher Education, Manipal, India
| | - Tina Mukherjee
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India
| |
Collapse
|
31
|
Zhao H, Chen Z, Li H, Zhao Y, Wang Q, Li W. Suppressed COP9 signalosome 5 promotes hemocyte proliferation through Cyclin E in the early G1 phase to defend against bacterial infection in crab. FASEB J 2022; 36:e22321. [DOI: 10.1096/fj.202101710rrrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Hui Zhao
- Laboratory of Invertebrate Immunological Defense School of Life Sciences East China Normal University Shanghai China
| | - Zhe Chen
- Laboratory of Invertebrate Immunological Defense School of Life Sciences East China Normal University Shanghai China
| | - Hao Li
- Laboratory of Invertebrate Immunological Defense School of Life Sciences East China Normal University Shanghai China
| | - Yue‐Hong Zhao
- Laboratory of Invertebrate Immunological Defense School of Life Sciences East China Normal University Shanghai China
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense School of Life Sciences East China Normal University Shanghai China
| | - Wei‐Wei Li
- Laboratory of Invertebrate Immunological Defense School of Life Sciences East China Normal University Shanghai China
| |
Collapse
|
32
|
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
|
33
|
Moussalem D, Augé B, Di Stefano L, Osman D, Gobert V, Haenlin M. Two Isoforms of serpent Containing Either One or Two GATA Zinc Fingers Provide Functional Diversity During Drosophila Development. Front Cell Dev Biol 2022; 9:795680. [PMID: 35178397 PMCID: PMC8844375 DOI: 10.3389/fcell.2021.795680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
GATA transcription factors play crucial roles in various developmental processes in organisms ranging from flies to humans. In mammals, GATA factors are characterized by the presence of two highly conserved domains, the N-terminal (N-ZnF) and the C-terminal (C-ZnF) zinc fingers. The Drosophila GATA factor Serpent (Srp) is produced in different isoforms that contains either both N-ZnF and C-ZnF (SrpNC) or only the C-ZnF (SrpC). Here, we investigated the functional roles ensured by each of these isoforms during Drosophila development. Using the CRISPR/Cas9 technique, we generated new mutant fly lines deleted for one (ΔsrpNC) or the other (ΔsrpC) encoded isoform, and a third one with a single point mutation in the N-ZnF that alters its interaction with its cofactor, the Drosophila FOG homolog U-shaped (Ush). Analysis of these mutants revealed that the Srp zinc fingers are differentially required for Srp to fulfill its functions. While SrpC is essential for embryo to adult viability, SrpNC, which is the closest conserved isoform to that of vertebrates, is not. However, to ensure its specific functions in larval hematopoiesis and fertility, Srp requires the presence of both N- and C-ZnF (SrpNC) and interaction with its cofactor Ush. Our results also reveal that in vivo the presence of N-ZnF restricts rather than extends the ability of GATA factors to regulate the repertoire of C-ZnF bound target genes.
Collapse
Affiliation(s)
- Douaa Moussalem
- Molecular, Cellular and Developmental Biology Department (MCD), Center for Integrative Biology (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| | - Benoit Augé
- Molecular, Cellular and Developmental Biology Department (MCD), Center for Integrative Biology (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| | - Luisa Di Stefano
- Molecular, Cellular and Developmental Biology Department (MCD), Center for Integrative Biology (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| | - Dani Osman
- Faculty of Sciences III, Lebanese University, Tripoli, Lebanon.,Azm Center for Research in Biotechnology and Its Applications, LBA3B, EDST, Lebanese University, Tripoli, Lebanon
| | - Vanessa Gobert
- Molecular, Cellular and Developmental Biology Department (MCD), Center for Integrative Biology (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| | - Marc Haenlin
- Molecular, Cellular and Developmental Biology Department (MCD), Center for Integrative Biology (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
34
|
Xin Q, Chen Z, Wei W, Wu Y. Animal models of acute lymphoblastic leukemia: Recapitulating the human disease to evaluate drug efficacy and discover therapeutic targets. Biochem Pharmacol 2022; 198:114970. [PMID: 35183530 DOI: 10.1016/j.bcp.2022.114970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/02/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant hematologic tumor with highly aggressive characteristics, which is prone to relapse, has a poor prognosis and few clinically effective drugs. It is meaningful to gain a better understanding of its pathogenesis in order to discover and evaluate potential therapeutic drugs and new treatment targets. The goal of developing novel targeted drugs and treatment methods is to increase complete remission, reduce toxicity and morbidity, and that is also the most important prerequisite for modern leukemia treatment. However, the process of new drugs from research and development to clinical application is long and difficult. Many promising drugs were rejected by the USFoodandDrugAdministration(FDA) due to serious adverse drug reactions (ADR) in clinical phase I trials. Animal models provide us with an excellent tool to understand the complex pathological mechanisms of human diseases, to evaluate the potential of new targeted drugs and therapeutic approaches to treat ALL in vivo and, more importantly, to assess the potential ADR they may have on healthy organs. In this article we review ALL animal models' progression, their roles in revealing the pathogenesis of ALL and drug development. Additionally, we mainly focus on the mouse models, especially xenotransplantation and transgenic models that more closely reproduce the human phenotype. In conclusion, we summarize the advantages and limitations of each model, thereby facilitating further understanding the etiology of ALL, and eventually contributing to the effective management of the disease.
Collapse
Affiliation(s)
- Qianling Xin
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei 230032, China
| | - Zhaoying Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| | - Yujing Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Provincial Institute of Translational Medicine, Hefei 230032, China.
| |
Collapse
|
35
|
Ultrastructural characterization of hemocytes in the oriental cockroach Blatta orientalis (Blattodea: Blattidae). ZOOMORPHOLOGY 2022. [DOI: 10.1007/s00435-021-00550-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
36
|
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
|
37
|
Wang L, Liu F, Zhang G, Su H, Sun J. A novel Ush transcription factor involving in hematopoiesis of Eriocheir sinensis. Comp Biochem Physiol B Biochem Mol Biol 2021; 259:110703. [PMID: 34915123 DOI: 10.1016/j.cbpb.2021.110703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/04/2021] [Accepted: 12/08/2021] [Indexed: 11/16/2022]
Abstract
The FOG transcriptional factor is a co-regulator that recognizes and binds to the GATA N-terminal zinc-finger domain and participates in hemocyte production and differentiation. In this study, an FOG-like gene, Ush, was characterized from Eriocheir sinensis, which consists of an 897 bp full-length open reading frame, encoding a polypeptide of 298 amino acids with four ZnF_C2H2 domains. The EsUsh mRNA transcripts were mainly expressed in the hematopoietic tissue (HPT) and hemocytes, and were significantly higher in hyalinocytes than semi-granulocytes and granulocytes, which were separated by Percoll gradient centrifugation. The transcription levels of EsUsh were found to be significantly upregulated in HPT, but downregulated in hemocytes after exsanguination. By using flow cytometry to determine the percentage of hemocyte sub-population after exsanguination, the percentage of hyalinocytes was found to significantly downregulated, while the percentage of granulocytes was significantly upregulated. Silencing EsUsh by dsRNA interference significantly decreased the percentage of hyalinocytes and small granulocytes, and increased the percentage of medium granulocytes and large granulocytes. Such findings suggest that EsUsh might be involved in hemocyte production and differentiation, especially in promoting hyalinocyte formation and limiting granulocyte generation and differentiation.
Collapse
Affiliation(s)
- Liyan Wang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China,.
| | - Fang Liu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Guangcheng Zhang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Hui Su
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Science, Tianjin Normal University, Tianjin 300387, China,.
| |
Collapse
|
38
|
Carrau T, Thümecke S, Silva LMR, Perez-Bravo D, Gärtner U, Taubert A, Hermosilla C, Vilcinskas A, Lee KZ. The Cellular Innate Immune Response of the Invasive Pest Insect Drosophila suzukii against Pseudomonas entomophila Involves the Release of Extracellular Traps. Cells 2021; 10:cells10123320. [PMID: 34943828 PMCID: PMC8699444 DOI: 10.3390/cells10123320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 01/02/2023] Open
Abstract
Drosophila suzukii is a neobiotic invasive pest that causes extensive damage to fruit crops worldwide. The biological control of this species has been unsuccessful thus far, in part because of its robust cellular innate immune system, including the activity of professional phagocytes known as hemocytes and plasmatocytes. The in vitro cultivation of primary hemocytes isolated from D. suzukii third-instar larvae is a valuable tool for the investigation of hemocyte-derived effector mechanisms against pathogens such as wasp parasitoid larvae, bacteria, fungi and viruses. Here, we describe the morphological characteristics of D. suzukii hemocytes and evaluate early innate immune responses, including extracellular traps released against the entomopathogen Pseudomonas entomophila and lipopolysaccharides. We show for the first time that D. suzukii plasmatocytes cast extracellular traps to combat P. entomophila, along with other cell-mediated reactions, such as phagocytosis and the formation of filopodia.
Collapse
Affiliation(s)
- Tessa Carrau
- Department Pests and Vector Insect Control, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, D-35394 Giessen, Germany; (T.C.); (A.V.)
| | - Susanne Thümecke
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich Buff Ring 26-32, D-35392 Giessen, Germany;
| | - Liliana M. R. Silva
- Institute of Parasitology, Justus Liebig University, Schubert Strasse 81, D-35392 Giessen, Germany; (A.T.); (C.H.)
- Correspondence: (L.M.R.S.); (K.-Z.L.)
| | - David Perez-Bravo
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Aulweg 123, D-35394 Giessen, Germany;
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Justus Liebig University, Aulweg 123, D-35392 Giessen, Germany;
| | - Anja Taubert
- Institute of Parasitology, Justus Liebig University, Schubert Strasse 81, D-35392 Giessen, Germany; (A.T.); (C.H.)
| | - Carlos Hermosilla
- Institute of Parasitology, Justus Liebig University, Schubert Strasse 81, D-35392 Giessen, Germany; (A.T.); (C.H.)
| | - Andreas Vilcinskas
- Department Pests and Vector Insect Control, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, D-35394 Giessen, Germany; (T.C.); (A.V.)
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich Buff Ring 26-32, D-35392 Giessen, Germany;
| | - Kwang-Zin Lee
- Department Pests and Vector Insect Control, Fraunhofer Institute for Molecular Biology and Applied Ecology, Ohlebergsweg 12, D-35394 Giessen, Germany; (T.C.); (A.V.)
- Correspondence: (L.M.R.S.); (K.-Z.L.)
| |
Collapse
|
39
|
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
|
40
|
Sun W, Song X, Dong M, Liu Z, Song Y, Wang L, Song L. DNA binding protein CgIkaros-like regulates the proliferation of agranulocytes and granulocytes in oyster (Crassostrea gigas). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 124:104201. [PMID: 34252475 DOI: 10.1016/j.dci.2021.104201] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
DNA-binding protein Ikaros is a major determinant of haematopoietic lineage, especially in the development, differentiation and proliferation of lymphocytes. In the present study, a Ikaros homologue (designed as CgIkaros-like) was identified and characterized as a vital determinant in the proliferation of haemocytes during haematopoiesis of Pacific oyster Crassostrea gigas. The complete coding sequence of CgIkaros-like was of 1329 bp encoding a predicted polypeptide of 442 amino acids with four ZnF regions, locating at the C-terminus and N-terminus respectively. The highest expression level of CgIkaros-like mRNA was found in gills, followed by haemocytes and gonad. The mRNA transcripts of CgIkaros-like could be detected in all the haemocytes with higher abundance in semi-granulocytes and agranulocytes. CgIkaros-like protein was localized in both of cytoplasm and nucleus with higher abundance in nucleus of oyster haemocytes. The mRNA and protein expression levels of agranulocyte marker CgCD9, granulocyte marker CgAATase, cell cycle related gene CgCDK2, Notch receptor CgNotch and Notch target gene CgHes1 all increased significantly (p < 0.05) after CgIkaros-like was interfered by siRNAs, which were about 27.33-, 2.63-, 24.34-, 4.45- and 6.08-fold of that in the siRNA-NC control group, respectively. While the transcripts of CgGATA3 and CgRunx did not change significantly after CgIkaros-like was interfered. These results demonstrated that CgIkaros-like functioned as a transcription factor combined with Notch pathway to mediate CgCDK2 and regulate the proliferation of oyster haemocytes.
Collapse
Affiliation(s)
- Wending Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaorui Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Miren Dong
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Zhuyun Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Ying Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, 519000, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
41
|
Li H, Jin XK, Zhou KM, Zhao H, Zhao YH, Wang Q, Li WW. Down Syndrome Cell Adhesion Molecule Triggers Membrane-to-Nucleus Signaling-Regulated Hemocyte Proliferation against Bacterial Infection in Invertebrates. THE JOURNAL OF IMMUNOLOGY 2021; 207:2265-2277. [PMID: 34580107 DOI: 10.4049/jimmunol.2100575] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/27/2021] [Indexed: 11/19/2022]
Abstract
Down syndrome cell adhesion molecule (Dscam) generates tens of thousands of isoforms by alternative splicing, thereby providing crucial functions during immune responses. In this study, a novel Dscam signaling pathway was investigated in crab, which remains poorly characterized in invertebrates. Bacterial infection induced the cytoplasmic cleavage of Dscam intracellular domains (ICDs) by γ-secretase, and then the released ICDs carrying specific alternatively spliced exons could directly interact with IPO5 to facilitate nuclear translocation. Nuclear imported ICDs thus promoted hemocyte proliferation and protect the host from bacterial infection. Protein-interaction studies revealed that the ectodomain of Dscam bound to a disintegrin and metalloprotease domain 10 (ADAM10) rather than ADAM17. Inhibition or overexpression of ADAM10 impaired or accelerated Dscam shedding activity post-bacterial stimulation, respectively. Moreover, the shedding signal then mediated Dscam with an intact cytoplasmic domain to promote the cleavage of ICDs by γ-secretase. Furthermore, the transcription of ADAM10 was regulated by Dscam-induced canonical signaling, but not nuclear imported ICDs, to serve as a feedback regulation between two different Dscam pathways. Thus, membrane-to-nuclear signaling of Dscam regulated hemocyte proliferation in response to bacterial infection.
Collapse
Affiliation(s)
- Hao Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China; and
| | - Xing-Kun Jin
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, China
| | - Kai-Min Zhou
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China; and
| | - Hui Zhao
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China; and
| | - Yue-Hong Zhao
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China; and
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China; and
| | - Wei-Wei Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China; and
| |
Collapse
|
42
|
Gong S, Zhang Y, Tian A, Deng W. Tumor models in various Drosophila tissues. WIREs Mech Dis 2021; 13:e1525. [PMID: 34730289 PMCID: PMC8566734 DOI: 10.1002/wsbm.1525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 01/07/2023]
Abstract
The development of cancer is a complex multistage process. Over the past few decades, the model organism Drosophila melanogaster has been crucial in identifying cancer-related genes and pathways and elucidating mechanisms underlying growth regulation in development. Investigations using Drosophila has yielded new insights into the molecular mechanisms involved in tumor initiation and progression. In this review, we describe various tumor models that have been developed in recent years using different Drosophila tissues, such as the imaginal tissue, the neural tissue, the gut, the ovary, and hematopoietic cells. We discuss underlying genetic alterations, cancer-like characteristics, as well as similarities and key differences among these models. We also discuss how disruptions in stem cell division and differentiation result in tumor formation in diverse tissues, and highlight new concepts developed using the fly model to understand context-dependent tumorigenesis. We further discuss the progress made in Drosophila to explore tumor-host interactions that involve the innate immune response to tumor growth and the cachexia wasting phenotype. This article is categorized under: Cancer > Genetics/Genomics/Epigenetics Cancer > Stem Cells and Development Cancer > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Shangyu Gong
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yichi Zhang
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Aiguo Tian
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Wu‐Min Deng
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
43
|
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: 88] [Impact Index Per Article: 22.0] [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
|
44
|
Winkler B, Funke D, Benmimoun B, Spéder P, Rey S, Logan MA, Klämbt C. Brain inflammation triggers macrophage invasion across the blood-brain barrier in Drosophila during pupal stages. SCIENCE ADVANCES 2021; 7:eabh0050. [PMID: 34705495 PMCID: PMC8550232 DOI: 10.1126/sciadv.abh0050] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The nervous system is shielded from circulating immune cells by the blood-brain barrier (BBB). During infections and autoimmune diseases, macrophages can enter the brain where they participate in pathogen elimination but can also cause tissue damage. Here, we establish a Drosophila model to study macrophage invasion into the inflamed brain. We show that the immune deficiency (Imd) pathway, but not the Toll pathway, is responsible for attraction and invasion of hemolymph-borne macrophages across the BBB during pupal stages. Macrophage recruitment is mediated by glial, but not neuronal, induction of the Imd pathway through expression of Pvf2. Within the brain, macrophages can phagocytose synaptic material and reduce locomotor abilities and longevity. Similarly, we show that central nervous system infection by group B Streptococcus elicits macrophage recruitment in an Imd-dependent manner. This suggests that evolutionarily conserved inflammatory responses require a delicate balance between beneficial and detrimental activities.
Collapse
Affiliation(s)
- Bente Winkler
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Dominik Funke
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Billel Benmimoun
- Brain Plasticity in response to the Environment, Institut Pasteur, UMR3738 CNRS, 75015 Paris, France
| | - Pauline Spéder
- Brain Plasticity in response to the Environment, Institut Pasteur, UMR3738 CNRS, 75015 Paris, France
| | - Simone Rey
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Mary A. Logan
- Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR 97239, USA
| | - Christian Klämbt
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
- Corresponding author.
| |
Collapse
|
45
|
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: 25] [Impact Index Per Article: 6.3] [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
|
46
|
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
|
47
|
Zhao YH, Li H, Zhao H, Sun WK, Wang Q, Li WW. An ancient interleukin-16-like molecule regulates hemocyte proliferation via integrin β1 in invertebrates. J Biol Chem 2021; 297:100943. [PMID: 34245782 PMCID: PMC8326423 DOI: 10.1016/j.jbc.2021.100943] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/10/2021] [Accepted: 07/06/2021] [Indexed: 12/27/2022] Open
Abstract
Interleukins (ILs) are cytokines with crucial functions in innate and adaptive immunity. IL genes are only found in vertebrates, except for IL-16, which has been cloned in some arthropod species. However, the function of this gene in invertebrates is unknown. In the present study, an IL-16-like gene (EsIL-16) was identified from the Chinese mitten crab Eriocheir sinensis. EsIL-16 was predicted to encode a precursor (proEsIL-16) that shares similarities with pro-IL-16 proteins from insects and vertebrates. We show that caspase-3 processes proEsIL-16 into an approximately 144-kDa N-terminal prodomain with nuclear import activity and an approximately 34-kDa mature peptide that might be secreted into the extracellular region. EsIL-16 mRNA could be detected in all analyzed tissues and was significantly upregulated after immune challenge both in vitro and in vivo. T7 phage display library screening suggested potential binding activity between EsIL-16 and integrin, which was confirmed by coimmunoprecipitation assay. Interestingly, EsIL-16 promoted cell proliferation via integrin β1 in primary cultured crab hemocytes and Drosophila S2 cells. Furthermore, the interaction between EsIL-16 and integrin β1 was necessary to efficiently protect the host from bacterial infection. To our knowledge, this study revealed integrin β1 as a receptor for IL-16 and the function of this interaction in hemocyte proliferation in invertebrates for the first time. These results provide new insights into the regulation of innate immune responses in invertebrates and shed the light on the evolution of ILs within the animal kingdom.
Collapse
Affiliation(s)
- Yue-Hong Zhao
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Hao Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Hui Zhao
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Wei-Kang Sun
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Wei-Wei Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
48
|
Kaech H, Dennis AB, Vorburger C. Triple RNA-Seq characterizes aphid gene expression in response to infection with unequally virulent strains of the endosymbiont Hamiltonella defensa. BMC Genomics 2021; 22:449. [PMID: 34134631 PMCID: PMC8207614 DOI: 10.1186/s12864-021-07742-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/20/2021] [Indexed: 11/10/2022] Open
Abstract
Background Secondary endosymbionts of aphids provide benefits to their hosts, but also impose costs such as reduced lifespan and reproductive output. The aphid Aphis fabae is host to different strains of the secondary endosymbiont Hamiltonella defensa, which encode different putative toxins. These strains have very different phenotypes: They reach different densities in the host, and the costs and benefits (protection against parasitoid wasps) they confer to the host vary strongly. Results We used RNA-Seq to generate hypotheses on why four of these strains inflict such different costs to A. fabae. We found different H. defensa strains to cause strain-specific changes in aphid gene expression, but little effect of H. defensa on gene expression of the primary endosymbiont, Buchnera aphidicola. The highly costly and over-replicating H. defensa strain H85 was associated with strongly reduced aphid expression of hemocytin, a marker of hemocytes in Drosophila. The closely related strain H15 was associated with downregulation of ubiquitin-related modifier 1, which is related to nutrient-sensing and oxidative stress in other organisms. Strain H402 was associated with strong differential regulation of a set of hypothetical proteins, the majority of which were only differentially regulated in presence of H402. Conclusions Overall, our results suggest that costs of different strains of H. defensa are likely caused by different mechanisms, and that these costs are imposed by interacting with the host rather than the host’s obligatory endosymbiont B. aphidicola. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07742-8.
Collapse
Affiliation(s)
- Heidi Kaech
- Aquatic Ecology, Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland. .,D-USYS, Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland.
| | - Alice B Dennis
- Institute of Biochemistry and Biology, University Potsdam, Potsdam, Germany
| | - Christoph Vorburger
- Aquatic Ecology, Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland.,D-USYS, Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
49
|
Coates JA, Brooks E, Brittle AL, Armitage EL, Zeidler MP, Evans IR. Identification of functionally distinct macrophage subpopulations in Drosophila. eLife 2021; 10:e58686. [PMID: 33885361 PMCID: PMC8062135 DOI: 10.7554/elife.58686] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
Vertebrate macrophages are a highly heterogeneous cell population, but while Drosophila blood is dominated by a macrophage-like lineage (plasmatocytes), until very recently these cells were considered to represent a homogeneous population. Here, we present our identification of enhancer elements labelling plasmatocyte subpopulations, which vary in abundance across development. These subpopulations exhibit functional differences compared to the overall population, including more potent injury responses and differential localisation and dynamics in pupae and adults. Our enhancer analysis identified candidate genes regulating plasmatocyte behaviour: pan-plasmatocyte expression of one such gene (Calnexin14D) improves wound responses, causing the overall population to resemble more closely the subpopulation marked by the Calnexin14D-associated enhancer. Finally, we show that exposure to increased levels of apoptotic cell death modulates subpopulation cell numbers. Taken together this demonstrates macrophage heterogeneity in Drosophila, identifies mechanisms involved in subpopulation specification and function and facilitates the use of Drosophila to study macrophage heterogeneity in vivo.
Collapse
Affiliation(s)
- Jonathon Alexis Coates
- Department of Biomedical Science and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Elliot Brooks
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Amy Louise Brittle
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Emma Louise Armitage
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Martin Peter Zeidler
- Department of Biomedical Science and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Iwan Robert Evans
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
50
|
Frankenreiter L, Gahr BM, Schmid H, Zimmermann M, Deichsel S, Hoffmeister P, Turkiewicz A, Borggrefe T, Oswald F, Nagel AC. Phospho-Site Mutations in Transcription Factor Suppressor of Hairless Impact Notch Signaling Activity During Hematopoiesis in Drosophila. Front Cell Dev Biol 2021; 9:658820. [PMID: 33937259 PMCID: PMC8079769 DOI: 10.3389/fcell.2021.658820] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
The highly conserved Notch signaling pathway controls a multitude of developmental processes including hematopoiesis. Here, we provide evidence for a novel mechanism of tissue-specific Notch regulation involving phosphorylation of CSL transcription factors within the DNA-binding domain. Earlier we found that a phospho-mimetic mutation of the Drosophila CSL ortholog Suppressor of Hairless [Su(H)] at Ser269 impedes DNA-binding. By genome-engineering, we now introduced phospho-specific Su(H) mutants at the endogenous Su(H) locus, encoding either a phospho-deficient [Su(H) S269A ] or a phospho-mimetic [Su(H) S269D ] isoform. Su(H) S269D mutants were defective of Notch activity in all analyzed tissues, consistent with impaired DNA-binding. In contrast, the phospho-deficient Su(H) S269A mutant did not generally augment Notch activity, but rather specifically in several aspects of blood cell development. Unexpectedly, this process was independent of the corepressor Hairless acting otherwise as a general Notch antagonist in Drosophila. This finding is in agreement with a novel mode of Notch regulation by posttranslational modification of Su(H) in the context of hematopoiesis. Importantly, our studies of the mammalian CSL ortholog (RBPJ/CBF1) emphasize a potential conservation of this regulatory mechanism: phospho-mimetic RBPJ S221D was dysfunctional in both the fly as well as two human cell culture models, whereas phospho-deficient RBPJ S221A rather gained activity during fly hematopoiesis. Thus, dynamic phosphorylation of CSL-proteins within the DNA-binding domain provides a novel means to fine-tune Notch signal transduction in a context-dependent manner.
Collapse
Affiliation(s)
- Lisa Frankenreiter
- Department of General Genetics (190g), Institute of Biology (190), University of Hohenheim, Stuttgart, Germany
| | - Bernd M Gahr
- Department of General Genetics (190g), Institute of Biology (190), University of Hohenheim, Stuttgart, Germany
| | - Hannes Schmid
- Department of General Genetics (190g), Institute of Biology (190), University of Hohenheim, Stuttgart, Germany
| | - Mirjam Zimmermann
- Department of General Genetics (190g), Institute of Biology (190), University of Hohenheim, Stuttgart, Germany
| | - Sebastian Deichsel
- Department of General Genetics (190g), Institute of Biology (190), University of Hohenheim, Stuttgart, Germany
| | - Philipp Hoffmeister
- Department of Internal Medicine 1, Center for Internal Medicine, University Medical Center Ulm, Ulm, Germany
| | | | - Tilman Borggrefe
- Institute of Biochemistry, Justus-Liebig University of Giessen, Giessen, Germany
| | - Franz Oswald
- Department of Internal Medicine 1, Center for Internal Medicine, University Medical Center Ulm, Ulm, Germany
| | - Anja C Nagel
- Department of General Genetics (190g), Institute of Biology (190), University of Hohenheim, Stuttgart, Germany
| |
Collapse
|