1
|
Chou J, Ramroop JR, Saravia-Butler AM, Wey B, Lera MP, Torres ML, Heavner ME, Iyer J, Mhatre SD, Bhattacharya S, Govind S. Drosophila parasitoids go to space: Unexpected effects of spaceflight on hosts and their parasitoids. iScience 2024; 27:108759. [PMID: 38261932 PMCID: PMC10797188 DOI: 10.1016/j.isci.2023.108759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/15/2023] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
While fruit flies (Drosophila melanogaster) and humans exhibit immune system dysfunction in space, studies examining their immune systems' interactions with natural parasites in space are lacking. Drosophila parasitoid wasps modify blood cell function to suppress host immunity. In this study, naive and parasitized ground and space flies from a tumor-free control and a blood tumor-bearing mutant strain were examined. Inflammation-related genes were activated in space in both fly strains. Whereas control flies did not develop tumors, tumor burden increased in the space-returned tumor-bearing mutants. Surprisingly, control flies were more sensitive to spaceflight than mutant flies; many of their essential genes were downregulated. Parasitoids appeared more resilient than fly hosts, and spaceflight did not significantly impact wasp survival or the expression of their virulence genes. Previously undocumented mutant wasps with novel wing color and wing shape were isolated post-flight and will be invaluable for host-parasite studies on Earth.
Collapse
Affiliation(s)
- Jennifer Chou
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Johnny R. Ramroop
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Amanda M. Saravia-Butler
- KBR NASA Ames Research Center, Moffett Field, CA 94035, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Brian Wey
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
- PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Matthew P. Lera
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Medaya L. Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Bionetics, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Mary Ellen Heavner
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Janani Iyer
- KBR NASA Ames Research Center, Moffett Field, CA 94035, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Universities Space Research Association, Mountain View, CA 94043, USA
| | - Siddhita D. Mhatre
- KBR NASA Ames Research Center, Moffett Field, CA 94035, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | | - Shubha Govind
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
- PhD Program in Biology, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- PhD Program in Biochemistry, The Graduate Center of the City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
2
|
Kinoshita S, Takarada K, Kinoshita Y, Inoue YH. Drosophila hemocytes recognize lymph gland tumors of mxc mutants and activate the innate immune pathway in a reactive oxygen species-dependent manner. Biol Open 2022; 11:bio059523. [PMID: 36226812 PMCID: PMC9641529 DOI: 10.1242/bio.059523] [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: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 12/29/2022] Open
Abstract
Mechanisms of cancer cell recognition and elimination by the innate immune system remains unclear. The immune signaling pathways are activated in the fat body to suppress the tumor growth in mxcmbn1 hematopoietic tumor mutants in Drosophila by inducing antimicrobial peptides (AMP). Here, we investigated the regulatory mechanism underlying the activation in the mutant. Firstly, we found that reactive oxygen species (ROS) accumulated in the hemocytes due to induction of dual oxidase and one of its activators. This was required for the AMP induction and the tumor growth suppression. Next, more hemocytes transplanted from normal larvae were associated with the mutant tumor than normal lymph glands (LGs). Matrix metalloproteinase 1 and 2 (MMP2) were highly expressed in the tumors. The basement membrane components in the tumors were reduced and ultimately lost inside. Depletion of the MMP2 rather than MMP1 resulted in a significantly reduced AMP expression in the mutant larvae. The hemocytes may recognize the disassembly of basement membrane in the tumors and activate the ROS production. Our findings highlight the mechanism via which macrophage-like hemocytes recognize tumor cells and subsequently convey the information to induce AMPs in the fat body. They contribute to uncover the role of innate immune system against cancer.
Collapse
Affiliation(s)
- Suzuko Kinoshita
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Kazuki Takarada
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Yuriko Kinoshita
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Yoshihiro H. Inoue
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| |
Collapse
|
3
|
Wang L, Qian J, Yang Y, Gu C. Novel insights into the impact of the SUMOylation pathway in hematological malignancies (Review). Int J Oncol 2021; 59:73. [PMID: 34368858 PMCID: PMC8360622 DOI: 10.3892/ijo.2021.5253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/26/2021] [Indexed: 12/17/2022] Open
Abstract
The small ubiquitin-like modifier (SUMO) system serves an important role in the regulation of protein stability and function. SUMOylation sustains the homeostatic equilibrium of protein function in normal tissues and numerous types of tumor. Accumulating evidence has revealed that SUMO enzymes participate in carcinogenesis via a series of complex cellular or extracellular processes. The present review outlines the physiological characteristics of the SUMOylation pathway and provides examples of SUMOylation participation in different cancer types, including in hematological malignancies (leukemia, lymphoma and myeloma). It has been indicated that the SUMO pathway may influence chromosomal instability, cell cycle progression, apoptosis and chemical drug resistance. The present review also discussed the possible relationship between SUMOylation and carcinogenic mechanisms, and evaluated their potential as biomarkers and therapeutic targets in the diagnosis and treatment of hematological malignancies. Developing and investigating inhibitors of SUMO conjugation in the future may offer promising potential as novel therapeutic strategies.
Collapse
Affiliation(s)
- Ling Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, P.R. China
| | - Jinjun Qian
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Ye Yang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, P.R. China
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, P.R. China
| |
Collapse
|
4
|
Tang R, Huang W, Guan J, Liu Q, Beerntsen BT, Ling E. Drosophila H2Av negatively regulates the activity of the IMD pathway via facilitating Relish SUMOylation. PLoS Genet 2021; 17:e1009718. [PMID: 34370736 PMCID: PMC8376203 DOI: 10.1371/journal.pgen.1009718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/19/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Insects depend on the innate immune response for defense against a wide array of pathogens. Central to Drosophila immunity are antimicrobial peptides (AMPs), released into circulation when pathogens trigger either of the two widely studied signal pathways, Toll or IMD. The Toll pathway responds to infection by Gram-positive bacteria and fungi while the IMD pathway is activated by Gram-negative bacteria. During activation of the IMD pathway, the NF-κB-like transcription factor Relish is phosphorylated and then cleaved, which is crucial for IMD-dependent AMP gene induction. Here we show that loss-of-function mutants of the unconventional histone variant H2Av upregulate IMD-dependent AMP gene induction in germ-free Drosophila larvae and adults. After careful dissection of the IMD pathway, we found that Relish has an epistatic relationship with H2Av. In the H2Av mutant larvae, SUMOylation is down-regulated, suggesting a possible role of SUMOylation in the immune phenotype. Eventually we demonstrated that Relish is mostly SUMOylated on amino acid K823. Loss of the potential SUMOylation site leads to significant auto-activation of Relish in vivo. Further work indicated that H2Av regulates Relish SUMOylation after physically interacting with Su(var)2-10, the E3 component of the SUMOylation pathway. Biochemical analysis suggested that SUMOylation of Relish prevents its cleavage and activation. Our findings suggest a new mechanism by which H2Av can negatively regulate, and thus prevent spontaneous activation of IMD-dependent AMP production, through facilitating SUMOylation of the NF-κB like transcription factor Relish. Toll and IMD signaling pathways should be involved in the production of antimicrobial peptides in animals upon infection. Immunity responses are energy consuming. Thus, these two pathways are fine-tuned. Animal H2A variant histones are involved in many physiological functions. In Drosophila, the production of antibacterial peptides is out of control in the mutant of H2A variant (H2Av810). After careful examination, we found that Relish, the transcription factor of the IMD pathway, was activated in this mutant. Eventually we demonstrate that Relish can be SUMOylated with the involvement of H2Av. Loss of the main SUMOylation site in Relish induces it to auto-activate following over-expression. Therefore, H2Av is a negative regulator of the IMD signaling pathway by maintaining the normal level of Relish SUMOylation in Drosophila.
Collapse
Affiliation(s)
- Ruijuan Tang
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Wuren Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Jingmin Guan
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Qiuning Liu
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetland, Yancheng Teachers University, Yancheng, China
| | - Brenda T. Beerntsen
- Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Erjun Ling
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
5
|
Kanwal A, Joshi PV, Mandal S, Mandal L. Ubx-Collier signaling cascade maintains blood progenitors in the posterior lobes of the Drosophila larval lymph gland. PLoS Genet 2021; 17:e1009709. [PMID: 34370733 PMCID: PMC8376192 DOI: 10.1371/journal.pgen.1009709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/19/2021] [Accepted: 07/12/2021] [Indexed: 12/02/2022] Open
Abstract
Drosophila larval hematopoiesis occurs in a specialized multi-lobed organ called the lymph gland. Extensive characterization of the organ has provided mechanistic insights into events related to developmental hematopoiesis. Spanning from the thoracic to the abdominal segment of the larvae, this organ comprises a pair of primary, secondary, and tertiary lobes. Much of our understanding arises from the studies on the primary lobe, while the secondary and tertiary lobes have remained mostly unexplored. Previous studies have inferred that these lobes are composed of progenitors that differentiate during pupation; however, the mechanistic basis of this extended progenitor state remains unclear. This study shows that posterior lobe progenitors are maintained by a local signaling center defined by Ubx and Collier in the tertiary lobe. This Ubx zone in the tertiary lobe shares several markers with the niche of the primary lobe. Ubx domain regulates the homeostasis of the posterior lobe progenitors in normal development and an immune-challenged scenario. Our study establishes the lymph gland as a model to tease out how the progenitors interface with the dual niches within an organ during development and disorders.
Collapse
Affiliation(s)
- Aditya Kanwal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| | - Pranav Vijay Joshi
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| | - Lolitika Mandal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab, India
| |
Collapse
|
6
|
Kurihara M, Komatsu K, Awane R, Inoue YH. Loss of Histone Locus Bodies in the Mature Hemocytes of Larval Lymph Gland Result in Hyperplasia of the Tissue in mxc Mutants of Drosophila. Int J Mol Sci 2020; 21:E1586. [PMID: 32111032 PMCID: PMC7084650 DOI: 10.3390/ijms21051586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 01/22/2023] Open
Abstract
Mutations in the multi sex combs (mxc) gene in Drosophila results in malignant hyperplasia in larval hematopoietic tissues, called lymph glands (LG). mxc encodes a component of the histone locus body (HLB) that is essential for cell cycle-dependent transcription and processing of histone mRNAs. The mammalian nuclear protein ataxia-telangiectasia (NPAT) gene, encoded by the responsible gene for ataxia telangiectasia, is a functional Mxc orthologue. However, their roles in tumorigenesis are unclear. Genetic analyses of the mxc mutants and larvae having LG-specific depletion revealed that a reduced activity of the gene resulted in the hyperplasia, which is caused by hyper-proliferation of immature LG cells. The depletion of mxc in mature hemocytes of the LG resulted in the hyperplasia. Furthermore, the inhibition of HLB formation was required for LG hyperplasia. In the mutant larvae, the total mRNA levels of the five canonical histones decreased, and abnormal forms of polyadenylated histone mRNAs, detected rarely in normal larvae, were generated. The ectopic expression of the polyadenylated mRNAs was sufficient for the reproduction of the hyperplasia. The loss of HLB function, especially 3-end processing of histone mRNAs, is critical for malignant LG hyperplasia in this leukemia model in Drosophila. We propose that mxc is involved in the activation to induce adenosine deaminase-related growth factor A (Adgf-A), which suppresses immature cell proliferation in LG.
Collapse
Affiliation(s)
| | | | | | - Yoshihiro H. Inoue
- Department of Insect Biomedical Research, Center for Advanced Insect Research Promotion, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-0962, Japan; (M.K.); (K.K.); (R.A.)
| |
Collapse
|
7
|
Panettieri S, Paddibhatla I, Chou J, Rajwani R, Moore RS, Goncharuk T, John G, Govind S. Discovery of aspirin-triggered eicosanoid-like mediators in a Drosophila metainflammation blood tumor model. J Cell Sci 2019; 133:jcs.236141. [PMID: 31562189 DOI: 10.1242/jcs.236141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/20/2019] [Indexed: 12/18/2022] Open
Abstract
Epidemiologic studies have linked the use of aspirin to a decline in chronic inflammation that underlies many human diseases, including some cancers. Aspirin reduces the levels of cyclooxygenase-mediated pro-inflammatory prostaglandins, promotes the production of pro-resolution molecules, and triggers the production of anti-inflammatory electrophilic mono-oxygenated (EFOX) lipid mediators. We investigated the effects of aspirin in fruit fly models of chronic inflammation. Ectopic Toll/NF-κB and JAK/STAT signaling in mutant D. melanogaster results in overproliferation of hematopoietic blood progenitors resulting in the formation of granuloma-like tumors. Ectopic JAK-STAT signaling also leads to metabolic inflammation. We report that aspirin-treated mutant flies experience reduction in metabolic inflammation, mitosis, ectopic immune signaling, and macrophage infiltration. Moreover, these flies synthesize 13-HODE, and aspirin triggers 13-oxoODE (13-EFOX-L2) production. Providing the precursor of 13-HODE, linoleic acid, or performing targeted knockdown of the transcription factor STAT in inflammatory blood cells, boosts 13-EFOX-L2 levels while decreasing metabolic inflammation. Thus, hematopoietic cells regulate metabolic inflammation in flies, and their effects can be reversed by pharmaceutical or dietary intervention, suggesting deep phylogenetic conservation in the ability of animals to resolve inflammation and repair tissue damage. These findings can help identify novel treatment targets in humans.
Collapse
Affiliation(s)
- Silvio Panettieri
- Department of Chemistry & Biochemistry, The City College of New York, New York, NY 10031, USA.,PhD Program in Chemistry, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Indira Paddibhatla
- PhD Program in Biology, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA.,Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Jennifer Chou
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Roma Rajwani
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Rebecca S Moore
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - Tamara Goncharuk
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA
| | - George John
- Department of Chemistry & Biochemistry, The City College of New York, New York, NY 10031, USA .,PhD Program in Chemistry, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Shubha Govind
- Biology Department, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA .,PhD Programs in Biology & Biochemistry, The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
8
|
Araki M, Kurihara M, Kinoshita S, Awane R, Sato T, Ohkawa Y, Inoue YH. Anti-tumour effects of antimicrobial peptides, components of the innate immune system, against haematopoietic tumours in Drosophila mxc mutants. Dis Model Mech 2019; 12:dmm037721. [PMID: 31160313 PMCID: PMC6602314 DOI: 10.1242/dmm.037721] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 05/10/2019] [Indexed: 12/21/2022] Open
Abstract
The innate immune response is the first line of defence against microbial infections. In Drosophila, two major pathways of the innate immune system (the Toll- and Imd-mediated pathways) induce the synthesis of antimicrobial peptides (AMPs) within the fat body. Recently, it has been reported that certain cationic AMPs exhibit selective cytotoxicity against human cancer cells; however, little is known about their anti-tumour effects. Drosophila mxcmbn1 mutants exhibit malignant hyperplasia in a larval haematopoietic organ called the lymph gland (LG). Here, using RNA-seq analysis, we found many immunoresponsive genes, including those encoding AMPs, to be upregulated in these mutants. Downregulation of these pathways by either a Toll or imd mutation enhanced the tumour phenotype of the mxc mutants. Conversely, ectopic expression of each of five different AMPs in the fat body significantly suppressed the LG hyperplasia phenotype in the mutants. Thus, we propose that the Drosophila innate immune system can suppress the progression of haematopoietic tumours by inducing AMP gene expression. Overexpression of any one of the five AMPs studied resulted in enhanced apoptosis in mutant LGs, whereas no apoptotic signals were detected in controls. We observed that two AMPs, Drosomycin and Defensin, were taken up by circulating haemocyte-like cells, which were associated with the LG regions and showed reduced cell-to-cell adhesion in the mutants. By contrast, the AMP Diptericin was directly localised at the tumour site without intermediating haemocytes. These results suggest that AMPs have a specific cytotoxic effect that enhances apoptosis exclusively in the tumour cells.
Collapse
Affiliation(s)
- Mayo Araki
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Massanori Kurihara
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Suzuko Kinoshita
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Rie Awane
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| | - Tetsuya Sato
- Medical Institute of Bioregulation, Kyushu University, Kyushu 812-0054, Japan
| | - Yasuyuki Ohkawa
- Medical Institute of Bioregulation, Kyushu University, Kyushu 812-0054, Japan
| | - Yoshihiro H Inoue
- Department of Insect Biomedical Research, Centre for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto 606-0962, Japan
| |
Collapse
|
9
|
Cell Adhesion-Mediated Actomyosin Assembly Regulates the Activity of Cubitus Interruptus for Hematopoietic Progenitor Maintenance in Drosophila. Genetics 2019; 212:1279-1300. [PMID: 31138608 PMCID: PMC6707476 DOI: 10.1534/genetics.119.302209] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/20/2019] [Indexed: 12/13/2022] Open
Abstract
The actomyosin network is involved in crucial cellular processes including morphogenesis, cell adhesion, apoptosis, proliferation, differentiation, and collective cell migration in Drosophila, Caenorhabditiselegans, and mammals. Here, we demonstrate that Drosophila larval blood stem-like progenitors require actomyosin activity for their maintenance. Genetic loss of the actomyosin network from progenitors caused a decline in their number. Likewise, the progenitor population increased upon sustained actomyosin activation via phosphorylation by Rho-associated kinase. We show that actomyosin positively regulates larval blood progenitors by controlling the maintenance factor Cubitus interruptus (Ci). Overexpression of the maintenance signal via a constitutively activated construct (ci.HA) failed to sustain Ci-155 in the absence of actomyosin components like Zipper (zip) and Squash (sqh), thus favoring protein kinase A (PKA)-independent regulation of Ci activity. Furthermore, we demonstrate that a change in cortical actomyosin assembly mediated by DE-cadherin modulates Ci activity, thereby determining progenitor status. Thus, loss of cell adhesion and downstream actomyosin activity results in desensitization of the progenitors to Hh signaling, leading to their differentiation. Our data reveal how cell adhesion and the actomyosin network cooperate to influence patterning, morphogenesis, and maintenance of the hematopoietic stem-like progenitor pool in the developing Drosophila hematopoietic organ.
Collapse
|
10
|
Banerjee U, Girard JR, Goins LM, Spratford CM. Drosophila as a Genetic Model for Hematopoiesis. Genetics 2019; 211:367-417. [PMID: 30733377 PMCID: PMC6366919 DOI: 10.1534/genetics.118.300223] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
In this FlyBook chapter, we present a survey of the current literature on the development of the hematopoietic system in Drosophila The Drosophila blood system consists entirely of cells that function in innate immunity, tissue integrity, wound healing, and various forms of stress response, and are therefore functionally similar to myeloid cells in mammals. The primary cell types are specialized for phagocytic, melanization, and encapsulation functions. As in mammalian systems, multiple sites of hematopoiesis are evident in Drosophila and the mechanisms involved in this process employ many of the same molecular strategies that exemplify blood development in humans. Drosophila blood progenitors respond to internal and external stress by coopting developmental pathways that involve both local and systemic signals. An important goal of these Drosophila studies is to develop the tools and mechanisms critical to further our understanding of human hematopoiesis during homeostasis and dysfunction.
Collapse
Affiliation(s)
- Utpal Banerjee
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
- Molecular Biology Institute, University of California, Los Angeles, California 90095
- Department of Biological Chemistry, University of California, Los Angeles, California 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| | - Juliet R Girard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Lauren M Goins
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Carrie M Spratford
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| |
Collapse
|
11
|
Baldeosingh R, Gao H, Wu X, Fossett N. Hedgehog signaling from the Posterior Signaling Center maintains U-shaped expression and a prohemocyte population in Drosophila. Dev Biol 2018; 441:132-145. [PMID: 29966604 PMCID: PMC6064674 DOI: 10.1016/j.ydbio.2018.06.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/15/2022]
Abstract
Hematopoietic progenitor choice between multipotency and differentiation is tightly regulated by intrinsic factors and extrinsic signals from the surrounding microenvironment. The Drosophila melanogaster hematopoietic lymph gland has emerged as a powerful tool to investigate mechanisms that regulate hematopoietic progenitor choice in vivo. The lymph gland contains progenitor cells, which share key characteristics with mammalian hematopoietic progenitors such as quiescence, multipotency and niche-dependence. The lymph gland is zonally arranged, with progenitors located in medullary zone, differentiating cells in the cortical zone, and the stem cell niche or Posterior Signaling Center (PSC) residing at the base of the medullary zone (MZ). This arrangement facilitates investigations into how signaling from the microenvironment controls progenitor choice. The Drosophila Friend of GATA transcriptional regulator, U-shaped, is a conserved hematopoietic regulator. To identify additional novel intrinsic and extrinsic regulators that interface with U-shaped to control hematopoiesis, we conducted an in vivo screen for factors that genetically interact with u-shaped. Smoothened, a downstream effector of Hedgehog signaling, was one of the factors identified in the screen. Here we report our studies that characterized the relationship between Smoothened and U-shaped. We showed that the PSC and Hedgehog signaling are required for U-shaped expression and that U-shaped is an important intrinsic progenitor regulator. These observations identify a potential link between the progenitor regulatory machinery and extrinsic signals from the PSC. Furthermore, we showed that both Hedgehog signaling and the PSC are required to maintain a subpopulation of progenitors. This led to a delineation of PSC-dependent versus PSC-independent progenitors and provided further evidence that the MZ progenitor population is heterogeneous. Overall, we have identified a connection between a conserved hematopoietic master regulator and a putative stem cell niche, which adds to our understanding of how signals from the microenvironment regulate progenitor multipotency.
Collapse
Affiliation(s)
- Rajkumar Baldeosingh
- Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hongjuan Gao
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Xiaorong Wu
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nancy Fossett
- Graduate Program in Life Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
12
|
Liu K, Zhang J, Wang H. Small ubiquitin-like modifier/sentrin-specific peptidase 1 associates with chemotherapy and is a risk factor for poor prognosis of non-small cell lung cancer. J Clin Lab Anal 2018; 32:e22611. [PMID: 30043429 DOI: 10.1002/jcla.22611] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/31/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND SUMO/sentrin-specific peptidase 1 (SENP1) was associated with radioresistance of cancer cells and was upregulated in non-small cell lung cancer (NSCLC). This study was to investigate the association of SENP1 with resistance of NSCLC tumor to chemoradiotherapy. METHODS Sentrin-specific peptidase 1 expression profile was detected using the immunohistochemistry and quantitative real-time PCR (qRT-PCR) analyses. The relative expression level of SENP1 mRNA was detected using qRT-PCR. The response to chemoradiotherapy was evaluated according to the Response Evaluation Criteria in Solid Tumors. RESULTS AND CONCLUSION When compared with adjacent non-tumor tissues, the overexpression of SENP1 mRNA and protein in NSCLC tumor tissues was determined using qRT-PCR and immunochemistry. Based on the chemoradiotherapy response rate, we found that NSCLC patients with higher SENP1 expression showed lower rates of complete response and higher partial and non-response rate to chemoradiotherapy. In the overall survival analysis, we found patients with high SENP1 expression showed significant shorter survival time compared with those with low SENP1 expression. In the multivariate Cox regression model, we found SENP1 overexpression, TNM stage, and lymph metastasis were independent risk factors for poor prognosis of NSCLC. SENP1 overexpression contributed to chemoradiotherapy resistance of NSCLC. The overexpression of SENP1 could be used as a risk factor for the poor prognosis of NSCLC.
Collapse
Affiliation(s)
- Keyuan Liu
- Department of Thoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou City, Zhejiang Province, China
| | - Jing Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou City, Zhejiang Province, China
| | - Hao Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
13
|
Khadilkar RJ, Vogl W, Goodwin K, Tanentzapf G. Modulation of occluding junctions alters the hematopoietic niche to trigger immune activation. eLife 2017; 6:28081. [PMID: 28841136 PMCID: PMC5597334 DOI: 10.7554/elife.28081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/24/2017] [Indexed: 12/04/2022] Open
Abstract
Stem cells are regulated by signals from their microenvironment, or niche. During Drosophila hematopoiesis, a niche regulates prohemocytes to control hemocyte production. Immune challenges activate cell-signalling to initiate the cellular and innate immune response. Specifically, certain immune challenges stimulate the niche to produce signals that induce prohemocyte differentiation. However, the mechanisms that promote prohemocyte differentiation subsequent to immune challenges are poorly understood. Here we show that bacterial infection induces the cellular immune response by modulating occluding-junctions at the hematopoietic niche. Occluding-junctions form a permeability barrier that regulates the accessibility of prohemocytes to niche derived signals. The immune response triggered by infection causes barrier breakdown, altering the prohemocyte microenvironment to induce immune cell production. Moreover, genetically induced barrier ablation provides protection against infection by activating the immune response. Our results reveal a novel role for occluding-junctions in regulating niche-hematopoietic progenitor signalling and link this mechanism to immune cell production following infection.
Collapse
Affiliation(s)
- Rohan J Khadilkar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Katharine Goodwin
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
14
|
The Friend of GATA Transcriptional Co-Regulator, U-Shaped, Is a Downstream Antagonist of Dorsal-Driven Prohemocyte Differentiation in Drosophila. PLoS One 2016; 11:e0155372. [PMID: 27163255 PMCID: PMC4862636 DOI: 10.1371/journal.pone.0155372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 04/27/2016] [Indexed: 12/11/2022] Open
Abstract
Recent studies suggest that mammalian hematopoietic stem and progenitor cells (HSPCs) respond directly to infection and inflammatory signaling. These signaling pathways also regulate HSPCs during steady-state conditions (absence of infection), and dysregulation may lead to cancer or age-related loss of progenitor repopulation capacity. Toll-like receptors (TLRs) are a major class of pathogen recognition receptors, and are expressed on the surface of immune effector cells and HSPCs. TLR/NF-κB activation promotes HSPCs differentiation; however, the mechanisms by which this signaling pathway alters the intrinsic transcriptional landscape are not well understood. Although Drosophila prohemocytes are the functional equivalent of mammalian HSPCs, a prohemocyte-specific function for Toll signaling has not been reported. Using Drosophila transgenics, we identified prohemocyte-specific roles for Toll pathway members, Dorsal and Cactus. We showed that Dorsal is required to limit the size of the progenitor pool. Additionally, we showed that activation of Toll signaling in prohemocytes drives differentiation in a manner that is analogous to TLR/NF-κB-driven HSPC differentiation. This was accomplished by showing that over-expression of Dorsal, or knockdown of Cactus, promotes differentiation. We also investigated whether Dorsal and Cactus control prohemocyte differentiation by regulating a key intrinsic prohemocyte factor, U-shaped (Ush), which is known to promote multipotency and block differentiation. We showed that Dorsal repressed Ush expression levels to promote differentiation, whereas Cactus maintained Ush levels to block differentiation. Additionally, we showed that another Toll antagonist, Lesswright, also maintained the level of Ush to block differentiation and promote proliferative quiescence. Collectively, these results identify a novel role for Ush as a downstream target of Toll signaling.
Collapse
|
15
|
Constanzo JD, Deng M, Rindhe S, Tang KJ, Zhang CC, Scaglioni PP. Pias1 is essential for erythroid and vascular development in the mouse embryo. Dev Biol 2016; 415:98-110. [PMID: 27155222 DOI: 10.1016/j.ydbio.2016.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/24/2016] [Accepted: 04/18/2016] [Indexed: 01/26/2023]
Abstract
The protein inhibitor of activated STAT-1 (PIAS1) is one of the few known SUMO E3 ligases. PIAS1 has been implicated in several biological processes including repression of innate immunity and DNA repair. However, PIAS1 function during development and tissue differentiation has not been studied. Here, we report that Pias1 is required for proper embryonic development. Approximately 90% of Pias1 null embryos die in utero between E10.5 and E12.5. We found significant apoptosis within the yolk sac (YS) blood vessels and concomitant loss of red blood cells (RBCs) resulting in profound anemia. In addition, Pias1 loss impairs YS angiogenesis and results in defective capillary plexus formation and blood vessel occlusions. Moreover, heart development is impaired as a result of loss of myocardium muscle mass. Accordingly, we found that Pias1 expression in primary myoblasts enhances the induction of cardiac muscle genes MyoD, Myogenin and Myomaker. PIAS1 protein regulation of cardiac gene transcription is dependent on transcription factors Myocardin and Gata-4. Finally, endothelial cell specific inactivation of Pias1 in vivo impairs YS erythrogenesis, angiogenesis and recapitulates loss of myocardium muscle mass. However, these defects are not sufficient to recapitulate the lethal phenotype of Pias1 null embryos. These findings highlight Pias1 as an essential gene for YS erythropoiesis and vasculogenesis in vivo.
Collapse
Affiliation(s)
- Jerfiz D Constanzo
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mi Deng
- Departments of Physiology, and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Smita Rindhe
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ke-Jing Tang
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pulmonary Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Cheng-Cheng Zhang
- Departments of Physiology, and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pier Paolo Scaglioni
- Department of Internal Medicine and Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
16
|
Benmimoun B, Haenlin M, Waltzer L. Blood cell progenitor maintenance: Collier barks out of the niche. Fly (Austin) 2016; 9:160-4. [PMID: 26925971 DOI: 10.1080/19336934.2016.1151130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Drosophila lymph gland, a larval haematopoietic organ, has emerged as a popular model to study regulatory mechanisms controlling blood cell progenitor fate. In this organ, the Posterior Signaling Center (PSC), a small group of cells expressing the EBF transcription factor Collier, has been proposed to act as a niche required for progenitor maintenance. Accordingly, several reports showed that PSC size/activity modulation impacts on blood cell differentiation. Yet our recent results challenge this model. Indeed, we found that PSC ablation does not affect haematopoietic progenitor maintenance. This unexpected result led us to reinvestigate the role of the PSC and collier in hematopoiesis. Consistent with previous findings, the PSC appears required for the production of a specialized blood cell type in response to parasitization. Moreover, our results indicate that the massive blood cell differentiation observed in collier mutant larvae is not due to the lack of PSC but to collier expression within the haematopoietic progenitors. We thus propose a paradigm shift whereby larval blood cell progenitor maintenance is largely independent of the PSC but requires the cell-autonomous function of collier.
Collapse
Affiliation(s)
- Billel Benmimoun
- a Université de Toulouse; UPS; CBD (Center de Biologie du Développement) France CNRS, CBD UMR5547 , F-31062 Toulouse , France.,b Present address: The Francis Crick Institute ; London WC2A 3LY , UK
| | - Marc Haenlin
- a Université de Toulouse; UPS; CBD (Center de Biologie du Développement) France CNRS, CBD UMR5547 , F-31062 Toulouse , France
| | - Lucas Waltzer
- a Université de Toulouse; UPS; CBD (Center de Biologie du Développement) France CNRS, CBD UMR5547 , F-31062 Toulouse , France
| |
Collapse
|
17
|
Lin CJ, Smibert P, Zhao X, Hu JF, Ramroop J, Kellner SM, Benton MA, Govind S, Dedon PC, Sternglanz R, Lai EC. An extensive allelic series of Drosophila kae1 mutants reveals diverse and tissue-specific requirements for t6A biogenesis. RNA (NEW YORK, N.Y.) 2015; 21:2103-2118. [PMID: 26516084 PMCID: PMC4647464 DOI: 10.1261/rna.053934.115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/14/2015] [Indexed: 06/03/2023]
Abstract
N(6)-threonylcarbamoyl-adenosine (t6A) is one of the few RNA modifications that is universally present in life. This modification occurs at high frequency at position 37 of most tRNAs that decode ANN codons, and stabilizes cognate anticodon-codon interactions. Nearly all genetic studies of the t6A pathway have focused on single-celled organisms. In this study, we report the isolation of an extensive allelic series in the Drosophila ortholog of the core t6A biosynthesis factor Kae1. kae1 hemizygous larvae exhibit decreases in t6A that correlate with allele strength; however, we still detect substantial t6A-modified tRNAs even during the extended larval phase of null alleles. Nevertheless, complementation of Drosophila Kae1 and other t6A factors in corresponding yeast null mutants demonstrates that these metazoan genes execute t6A synthesis. Turning to the biological consequences of t6A loss, we characterize prominent kae1 melanotic masses and show that they are associated with lymph gland overgrowth and ectopic generation of lamellocytes. On the other hand, kae1 mutants exhibit other phenotypes that reflect insufficient tissue growth. Interestingly, whole-tissue and clonal analyses show that strongly mitotic tissues such as imaginal discs are exquisitely sensitive to loss of kae1, whereas nonproliferating tissues are less affected. Indeed, despite overt requirements of t6A for growth of many tissues, certain strong kae1 alleles achieve and sustain enlarged body size during their extended larval phase. Our studies highlight tissue-specific requirements of the t6A pathway in a metazoan context and provide insights into the diverse biological roles of this fundamental RNA modification during animal development and disease.
Collapse
Affiliation(s)
- Ching-Jung Lin
- Department of Developmental Biology, Sloan-Kettering Institute, New York, New York 10065, USA
| | - Peter Smibert
- Department of Developmental Biology, Sloan-Kettering Institute, New York, New York 10065, USA Research School of Biological Sciences, The Australian National University, Acton ACT 2601, Australia
| | - Xiaoyu Zhao
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Jennifer F Hu
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | - Johnny Ramroop
- Department of Biology, The City College of the City University of New York, New York 10031, USA The Graduate Center of the City University of New York, New York 10016, USA
| | - Stefanie M Kellner
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | - Matthew A Benton
- Research School of Biological Sciences, The Australian National University, Acton ACT 2601, Australia
| | - Shubha Govind
- Department of Biology, The City College of the City University of New York, New York 10031, USA The Graduate Center of the City University of New York, New York 10016, USA
| | - Peter C Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | - Rolf Sternglanz
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794, USA
| | - Eric C Lai
- Department of Developmental Biology, Sloan-Kettering Institute, New York, New York 10065, USA
| |
Collapse
|
18
|
The EBF transcription factor Collier directly promotes Drosophila blood cell progenitor maintenance independently of the niche. Proc Natl Acad Sci U S A 2015; 112:9052-7. [PMID: 26150488 DOI: 10.1073/pnas.1423967112] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The maintenance of stem or progenitor cell fate relies on intrinsic factors as well as local cues from the cellular microenvironment and systemic signaling. In the lymph gland, an hematopoietic organ in Drosophila larva, a group of cells called the Posterior Signaling Centre (PSC), whose specification depends on the EBF transcription factor Collier (Col) and the HOX factor Antennapedia (Antp), has been proposed to form a niche required to maintain the pool of hematopoietic progenitors (prohemocytes). In contrast with this model, we show here that genetic ablation of the PSC does not cause an increase in blood cell differentiation or a loss of blood cell progenitors. Furthermore, although both col and Antp mutant larvae are devoid of PSC, the massive prohemocyte differentiation observed in col mutant is not phenocopied in Antp mutant. Interestingly, beside its expression in the PSC, Col is also expressed at low levels in prohemocytes and we show that this expression persists in PSC-ablated and Antp mutant larvae. Moreover, targeted knockdown and rescue experiments indicate that Col expression is required in the prohemocytes to prevent their differentiation. Together, our findings show that the PSC is dispensable for blood cell progenitor maintenance and reveal the key role of the conserved transcription factor Col as an intrinsic regulator of hematopoietic progenitor fate.
Collapse
|
19
|
Heavner ME, Hudgins AD, Rajwani R, Morales J, Govind S. Harnessing the natural Drosophila-parasitoid model for integrating insect immunity with functional venomics. CURRENT OPINION IN INSECT SCIENCE 2014; 6:61-67. [PMID: 25642411 PMCID: PMC4309977 DOI: 10.1016/j.cois.2014.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Drosophila species lack most hallmarks of adaptive immunity yet are highly successful against an array of natural microbial pathogens and metazoan enemies. When attacked by figitid parasitoid wasps, fruit flies deploy robust, multi-faceted innate immune responses and overcome many attackers. In turn, parasitoids have evolved immunosuppressive strategies to match, and more frequently to overcome, their hosts. We present methods to examine the evolutionary dynamics underlying anti-parasitoid host defense by teasing apart the specialized immune-modulating venoms of figitid parasitoids and, in turn, possibly delineating the roles of individual venom molecules. This combination of genetic, phylogenomic, and "functional venomics" methods in the Drosophila-parasitoid model should allow entomologists and immunologists to tackle important outstanding questions with implications across disciplines and to pioneer translational applications in agriculture and medicine.
Collapse
Affiliation(s)
- Mary E. Heavner
- The Graduate Center, City University of New York, New York, 10016
- Department of Biology, The City College of New York, City University of New York, New York, New York, 10031
| | - Adam D. Hudgins
- Department of Biology, The City College of New York, City University of New York, New York, New York, 10031
| | - Roma Rajwani
- Department of Biology, The City College of New York, City University of New York, New York, New York, 10031
| | - Jorge Morales
- Department of Biology, The City College of New York, City University of New York, New York, New York, 10031
| | - Shubha Govind
- The Graduate Center, City University of New York, New York, 10016
- Department of Biology, The City College of New York, City University of New York, New York, New York, 10031
| |
Collapse
|
20
|
ARF1-GTP regulates Asrij to provide endocytic control of Drosophila blood cell homeostasis. Proc Natl Acad Sci U S A 2014; 111:4898-903. [PMID: 24707047 DOI: 10.1073/pnas.1303559111] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Drosophila melanogaster larval hematopoiesis is a well-established model to study mechanisms that regulate hematopoietic niche maintenance and control of blood cell precursor (prohemocyte) differentiation. Molecules that perturb niche function affect the balance between prohemocytes and differentiated hemocytes. The conserved hemocyte-specific endosomal protein Asrij is essential for niche function and prohemocyte maintenance. Elucidating how subcellular trafficking molecules can regulate signaling presents an important challenge. Here we show that Asrij function is mediated by the Ras family GTPase Arf79F, the Drosophila homolog of ADP ribosylation factor 1 (ARF1), essential for clathrin coat assembly, Golgi architecture, and vesicular trafficking. ARF1 is expressed in the larval lymph gland and in circulating hemocytes and interacts with Asrij. ARF1-depleted lymph glands show loss of niche cells and prohemocyte maintenance with increased differentiation. Inhibiting ARF1 activation by knocking down its guanine nucleotide exchange factor (Gartenzwerg) or overexpressing its GTPAse-activating protein showed that ARF1-GTP is essential for regulating niche size and maintaining stemness. Activated ARF1 regulates Asrij levels in blood cells thereby mediating Asrij function. Asrij controls crystal cell differentiation by affecting Notch trafficking. ARF1 perturbation also leads to aberrant Notch trafficking and the Notch intracellular domain is stalled in sorting endosomes. Thus, ARF1 can regulate Drosophila blood cell homeostasis by regulating Asrij endocytic function. ARF1 also regulates signals arising from the niche and differentiated cells by integrating the insulin-mediated and PDGF-VEGF receptor signaling pathways. We propose that the conserved ARF1-Asrij endocytic axis modulates signals that govern hematopoietic development. Thus, Asrij affords tissue-specific control of global mechanisms involved in molecular traffic.
Collapse
|
21
|
An unexpected link between notch signaling and ROS in restricting the differentiation of hematopoietic progenitors in Drosophila. Genetics 2013; 197:471-83. [PMID: 24318532 DOI: 10.1534/genetics.113.159210] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A fundamental question in hematopoietic development is how multipotent progenitors achieve precise identities, while the progenitors themselves maintain quiescence. In Drosophila melanogaster larvae, multipotent hematopoietic progenitors support the production of three lineages, exhibit quiescence in response to cues from a niche, and from their differentiated progeny. Infection by parasitic wasps alters the course of hematopoiesis. Here we address the role of Notch (N) signaling in lamellocyte differentiation in response to wasp infection. We show that Notch activity is moderately high and ubiquitous in all cells of the lymph gland lobes, with crystal cells exhibiting the highest levels. Wasp infection reduces Notch activity, which results in fewer crystal cells and more lamellocytes. Robust lamellocyte differentiation is induced even in N mutants. Using RNA interference knockdown of N, Serrate, and neuralized (neur), and twin clone analysis of a N null allele, we show that all three genes inhibit lamellocyte differentiation. However, unlike its cell-autonomous function in crystal cell development, Notch's inhibitory influence on lamellocyte differentiation is not cell autonomous. High levels of reactive oxygen species in the lymph gland lobes, but not in the niche, accompany N(RNAi)-induced lamellocyte differentiation and lobe dispersal. Our results define a novel dual role for Notch signaling in maintaining competence for basal hematopoiesis: while crystal cell development is encouraged, lamellocytic fate remains repressed. Repression of Notch signaling in fly hematopoiesis is important for host defense against natural parasitic wasp infections. These findings can serve as a model to understand how reactive oxygen species and Notch signals are integrated and interpreted in vivo.
Collapse
|
22
|
Abstract
The Toll signaling pathway has a highly conserved function in innate immunity and is regulated by multiple factors that fine tune its activity. One such factor is β-arrestin Kurtz (Krz), which we previously implicated in the inhibition of developmental Toll signaling in the Drosophila melanogaster embryo. Another level of controlling Toll activity and immune system homeostasis is by protein sumoylation. In this study, we have uncovered a link between these two modes of regulation and show that Krz affects sumoylation via a conserved protein interaction with a SUMO protease, Ulp1. Loss of function of krz or Ulp1 in Drosophila larvae results in a similar inflammatory phenotype, which is manifested as increased lamellocyte production; melanotic mass formation; nuclear accumulation of Toll pathway transcriptional effectors, Dorsal and Dif; and expression of immunity genes, such as Drosomycin. Moreover, mutations in krz and Ulp1 show dosage-sensitive synergistic genetic interactions, suggesting that these two proteins are involved in the same pathway. Using Dorsal sumoylation as a readout, we found that altering Krz levels can affect the efficiency of SUMO deconjugation mediated by Ulp1. Our results demonstrate that β-arrestin controls Toll signaling and systemic inflammation at the level of sumoylation.
Collapse
|
23
|
Gueguen G, Kalamarz ME, Ramroop J, Uribe J, Govind S. Polydnaviral ankyrin proteins aid parasitic wasp survival by coordinate and selective inhibition of hematopoietic and immune NF-kappa B signaling in insect hosts. PLoS Pathog 2013; 9:e1003580. [PMID: 24009508 PMCID: PMC3757122 DOI: 10.1371/journal.ppat.1003580] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 07/07/2013] [Indexed: 12/04/2022] Open
Abstract
Polydnaviruses are mutualists of their parasitoid wasps and express genes in immune cells of their Lepidopteran hosts. Polydnaviral genomes carry multiple copies of viral ankyrins or vankyrins. Vankyrin proteins are homologous to IκB proteins, but lack sequences for regulated degradation. We tested if Ichnoviral Vankyrins differentially impede Toll-NF-κB-dependent hematopoietic and immune signaling in a heterologous in vivo Drosophila, system. We first show that hematopoiesis and the cellular encapsulation response against parasitoid wasps are tightly-linked via NF-κB signaling. The niche, which neighbors the larval hematopoietic progenitors, responds to parasite infection. Drosophila NF-κB proteins are expressed in the niche, and non cell-autonomously influence fate choice in basal and parasite-activated hematopoiesis. These effects are blocked by the Vankyrin I2-vank-3, but not by P-vank-1, as is the expression of a NF-κB target transgene. I2-vank-3 and P-vank-1 differentially obstruct cellular and humoral inflammation. Additionally, their maternal expression weakens ventral embryonic patterning. We propose that selective perturbation of NF-κB-IκB interactions in natural hosts of parasitic wasps negatively impacts the outcome of hematopoietic and immune signaling and this immune deficit contributes to parasite survival and species success in nature. Parasitoid wasps are insects whose development takes place within the body of other insects. To survive, wasp larvae must overcome the immune defenses of their hosts. How parasitic wasps overcome host immunity is not fully understood even though we know that different strategies using venoms, virus-like particles, or viruses are involved. A unique class of viruses, called polydnaviruses is found in two families of wasps that comprise more than 20,000 wasp species. The genomes of polydnaviruses encode proteins with ankyrin repeats. Ankyrin repeats are also found in Cactus, the inhibitor protein of NF-κB signaling in Drosophila. Viral ankyrin proteins, or Vankyrins, however, lack the amino acid sequences necessary for turnover found in Cactus and mammalian IκB family members. We show that Vankyrins produced by polydnaviruses of a parasitic wasp that attacks caterpillars of many common agricultural pests can block NF-κB signaling in fruit fly larvae. This inhibition supports parasite success. Our work highlights the crucial role of NF-κB signaling across insect taxa in insect-insect and insect-virus interactions. Studies of polydnaviral ankyrin proteins in Drosophila reveal that immune-suppressive viruses may block both cellular and humoral immunity in insects to win the biological ‘arms race’.
Collapse
Affiliation(s)
- Gwenaelle Gueguen
- Biology Department, The City College of the City University of New York, New York, New York, United States of America
| | - Marta E. Kalamarz
- Biology Department, The City College of the City University of New York, New York, New York, United States of America
- The Graduate Center of the City University of New York, New York, New York, United States of America
| | - Johnny Ramroop
- Biology Department, The City College of the City University of New York, New York, New York, United States of America
- The Graduate Center of the City University of New York, New York, New York, United States of America
| | - Jeffrey Uribe
- Biology Department, The City College of the City University of New York, New York, New York, United States of America
| | - Shubha Govind
- Biology Department, The City College of the City University of New York, New York, New York, United States of America
- The Graduate Center of the City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
24
|
Ivanschitz L, De Thé H, Le Bras M. PML, SUMOylation, and Senescence. Front Oncol 2013; 3:171. [PMID: 23847762 PMCID: PMC3701148 DOI: 10.3389/fonc.2013.00171] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 06/14/2013] [Indexed: 11/15/2022] Open
Abstract
Since its discovery, 25 years ago, promyelocytic leukemia (PML) has been an enigma. Implicated in the oncogenic PML/RARA fusion, forming elusive intranuclear domains, triggering cell death or senescence, controlled by and perhaps controlling SUMOylation… there are multiple PML-related issues. Here we review the reciprocal interactions between PML, senescence, and SUMOylation, notably in the context of cellular transformation.
Collapse
Affiliation(s)
- Lisa Ivanschitz
- University Paris Diderot, Sorbonne Paris Cité, Hôpital St. Louis , Paris , France ; INSERM UMR 944, Equipe labellisée par la Ligue Nationale contre le Cancer, Institut Universitaire d'Hématologie, Hôpital St. Louis , Paris , France ; CNRS UMR 7212, Hôpital St. Louis , Paris , France
| | | | | |
Collapse
|
25
|
Fossett N. Signal transduction pathways, intrinsic regulators, and the control of cell fate choice. Biochim Biophys Acta Gen Subj 2012; 1830:2375-84. [PMID: 22705942 DOI: 10.1016/j.bbagen.2012.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 05/10/2012] [Accepted: 06/07/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Information regarding changes in organismal status is transmitted to the stem cell regulatory machinery by a limited number of signal transduction pathways. Consequently, these pathways derive their functional specificity through interactions with stem cell intrinsic master regulators, notably transcription factors. Identifying the molecular underpinnings of these interactions is critical to understanding stem cell function. SCOPE OF REVIEW This review focuses on studies in Drosophila that identify the gene regulatory basis for interactions between three different signal transduction pathways and an intrinsic master transcriptional regulator in the context of hematopoietic stem-like cell fate choice. Specifically, the interface between the GATA:FOG regulatory complex and the JAK/STAT, BMP, and Hedgehog pathways is examined. MAJOR CONCLUSIONS The GATA:FOG complex coordinates information transmitted by at least three different signal transduction pathways as a means to control stem-like cell fate choice. This illustrates emerging principles concerning regulation of stem cell function and describes a gene regulatory link between changes in organismal status and stem cell response. GENERAL SIGNIFICANCE The Drosophila model system offers a powerful approach to identify the molecular basis of how stem cells receive, interpret, and then respond to changes in organismal status. This article is part of a Special Issue entitled: Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Nancy Fossett
- Center for Vascular and Inflammatory Diseases and the Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|