1
|
Koyama T, Saeed U, Rewitz K, Halberg KV. The Integrative Physiology of Hormone Signaling: Insights from Insect Models. Physiology (Bethesda) 2025; 40:0. [PMID: 39887191 DOI: 10.1152/physiol.00030.2024] [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: 06/21/2024] [Revised: 11/18/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Hormones orchestrate virtually all physiological processes in animals and enable them to adjust internal responses to meet diverse physiological demands. Studies in both vertebrates and insects have uncovered many novel hormones and dissected the physiological mechanisms they regulate, demonstrating a remarkable conservation in endocrine signaling across the tree of life. In this review, we focus on recent advances in insect research, which have provided a more integrative view of the conserved interorgan communication networks that control physiology. These new insights have been driven by experimental advantages inherent to insects, which over the past decades have aligned with new technologies and sophisticated genetic tools, to transform insect genetic models into a powerful testbed for posing new questions and exploring longstanding issues in endocrine research. Here, we illustrate how insect studies have addressed classic questions in three main areas, hormonal control of growth and development, neuroendocrine regulation of ion and water balance, and hormonal regulation of behavior and metabolism, and how these discoveries have illuminated our fundamental understanding of endocrine signaling in animals. The application of integrative physiology in insect systems to questions in endocrinology and physiology is expanding and is poised to be a crucible of discovery, revealing fundamental mechanisms of hormonal regulation that underlie animal adaptations to their environments.
Collapse
Affiliation(s)
- Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Usama Saeed
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Bellampalli SS, Joshi V, Fonar G, Beyder A. Integration of signal transduction pathways in sensory epithelial enteroendocrine cells. Mol Biol Cell 2025; 36:re3. [PMID: 40408598 DOI: 10.1091/mbc.e24-03-0143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025] Open
Abstract
The gastrointestinal (GI) tract plays a crucial role in nutrient absorption, motility, and metabolism, while serving as a barrier against pathogens. The intestinal epithelial layer, particularly enteroendocrine cells (EEC), is pivotal in sensing luminal stimuli due to its strategic position. Although comprising only 1% of the GI epithelium, in total EEC comprises perhaps the largest endocrine organ that couples GI luminal stimuli with release of hormones and neurotransmitters such as serotonin, glucagon-like peptide-1 (GLP-1), and peptide-tyrosine-tyrosine (PYY), which regulate local and systemic physiology. In this review, we use EECs as a model to explore critical intracellular signaling pathways-centering around calcium (Ca²⁺) and cyclic adenosine monophosphate (cAMP). EECs utilize ionotropic and metabotropic receptors to detect luminal stimuli, leading to increased intracellular cytoplasmic Ca²⁺ and altered intracellular cAMP dynamics. Ca²⁺ influx is critical for the exocytosis of signaling molecules. The interplay between Ca²⁺ and cAMP signaling pathways enhances the release of these molecules, affecting both local and systemic responses. We make a point to discuss the conventional and emerging methodologies for measuring Ca²⁺ and cAMP dynamics in EECs, which is a rapidly expanding toolbox, containing genetically encoded indicators and resonance energy transfer techniques. Understanding these signaling cascades is essential for deciphering the complex roles of EECs in GI physiology and their impact on systemic health.
Collapse
Affiliation(s)
- Shreya S Bellampalli
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905
| | - Vikram Joshi
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Gennadiy Fonar
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
3
|
Zheng C, Zheng J, Wang X, Zhang Y, Ma X, He L. Two-pore-domain potassium channel Sandman regulates intestinal stem cell homeostasis and tumorigenesis in Drosophila melanogaster. J Genet Genomics 2025:S1673-8527(25)00147-X. [PMID: 40381822 DOI: 10.1016/j.jgg.2025.05.003] [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: 03/03/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Potassium channels regulate diverse biological processes, ranging from cell proliferation to immune responses. However, the functions of potassium homeostasis and its regulatory mechanisms in adult stem cells and tumors remain poorly characterized. Here, we identify Sandman, a two-pore-domain potassium channel in Drosophila, as an essential regulator for the proliferation of intestinal stem cells and malignant tumors, while dispensable for the normal development processes. Mechanistically, loss of sandman elevates intracellular K+ concentration, leading to growth inhibition. This phenotype is rescued by pharmacological reduction of intracellular K+ levels using the K+ ionophore. Conversely, overexpression of sandman triggers stem cell death in most regions of the midgut, inhibits tumor growth, and induces a Notch loss-of-function phenotype in the posterior midgut. These effects are mediated predominantly via the induction of endoplasmic reticulum (ER) stress, as demonstrated by the complete rescue of phenotypes through the co-expression of Ire1 or Xbp1s. Additionally, human homologs of Sandman demonstrated similar ER stress-inducing capabilities, suggesting an evolutionarily conserved relationship between this channel and ER stress. Together, our findings identify Sandman as a shared regulatory node that governs Drosophila adult stem cell dynamics and tumorigenesis through bioelectric homeostasis, and reveal a link between the two-pore potassium channel and ER stress signaling.
Collapse
Affiliation(s)
- Chen Zheng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jiadong Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Xin Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yue Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xianjue Ma
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China.
| | - Li He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
4
|
Asahina K, Zelikowsky M. Comparative Perspectives on Neuropeptide Function and Social Isolation. Biol Psychiatry 2025; 97:942-952. [PMID: 39892690 PMCID: PMC12048258 DOI: 10.1016/j.biopsych.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Chronic social isolation alters behavior across animal species. Genetic model organisms such as mice and flies provide crucial insight into the molecular and physiological effects of social isolation on brain cells and circuits. Here, we comparatively review recent findings regarding the function of conserved neuropeptides in social isolation in mice and flies. Analogous functions of 3 classes of neuropeptides-tachykinins, cholecystokinins, and neuropeptide Y/F-in the two model organisms suggest that these molecules may be involved in modulating behavioral changes induced by social isolation across a wider range of species, including humans. Comparative approaches armed with tools to dissect neuropeptidergic function can lead to an integrated understanding of the impacts of social isolation on brain circuits and behavior.
Collapse
Affiliation(s)
- Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California.
| | - Moriel Zelikowsky
- Department of Neurobiology, School of Medicine, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
5
|
Lee SH, Hwang D, Lee JW, Goo TW, Yun EY. The Identification of a Glucuronyltransferase-Related Gene, GlcAT-S, with Putative Mucus Protection and Anti-Inflammatory Effects from Gut-Damaged Drosophila by Dextran Sulfate Sodium (DSS). BIOLOGY 2025; 14:513. [PMID: 40427702 PMCID: PMC12109491 DOI: 10.3390/biology14050513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025]
Abstract
The intestinal epithelium, which is protected by mucosal surfaces composed of mucins and other glycoproteins, functions as a selective barrier that absorbs nutrients while preventing the translocation of harmful substances. To understand the mechanisms between mucosal disruption and tissue inflammation, we orally administrated a mucus-disrupting agent, dextran sodium sulfate, to Drosophila melanogaster and screened 63 differentially expressed genes (DEGs). Through a database search using bioinformatics tools (CHEA3 and WebGestalt), we identified ELK1 as a potential key transcription factor for the selected DEGs, and among the 63 DEGs, ELK1-related genes, B3GAT3, FIBP, and TENT2 (GlcAT-S, Fibp, and Wisp in Drosophila), were selected as the relevant genes that respond to mucus disruption. We confirmed that enterocyte (EC)-specific GlcAT-S knockdown by RNAi significantly reduced gut length and increased intestinal stem cell proliferation in Drosophila. Additionally, in EC-specific GlcAT-S-knockdown flies, it was observed that the mucus-production-related genes, Muc68D and Mur29B, were specifically reduced, whereas the inflammatory cytokines egr and upd3 were overexpressed. This study provides evidence that GlcAT-S is involved in the regulation of intestinal inflammation in Drosophila and plays a protective role against mucus disruption. Our findings suggest that GlcAT-S may be a potential therapeutic target for the treatment of intestinal inflammatory diseases such as IBD.
Collapse
Affiliation(s)
- Seung Hun Lee
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea; (S.H.L.); (D.H.); (J.-W.L.)
| | - Dooseon Hwang
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea; (S.H.L.); (D.H.); (J.-W.L.)
- Department of Future Food and Resources Technology, Donga University of Health, Yeongam 58439, Republic of Korea
| | - Jang-Won Lee
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea; (S.H.L.); (D.H.); (J.-W.L.)
| | - Tae-Won Goo
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju 38766, Republic of Korea
| | - Eun-Young Yun
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea; (S.H.L.); (D.H.); (J.-W.L.)
| |
Collapse
|
6
|
Knudsen C, Moriya A, Nakato E, Gulati R, Akiyama T, Nakato H. Chondroitin sulfate regulates proliferation of Drosophila intestinal stem cells. PLoS Genet 2025; 21:e1011686. [PMID: 40343906 PMCID: PMC12063844 DOI: 10.1371/journal.pgen.1011686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 04/10/2025] [Indexed: 05/11/2025] Open
Abstract
The basement membrane (BM) plays critical roles in stem cell maintenance and activity control. Here we show that chondroitin sulfate (CS), a major component of the Drosophila midgut BM, is required for proper control of intestinal stem cells (ISCs). Loss of Chsy, a critical CS biosynthetic gene, resulted in elevated levels of ISC proliferation during homeostasis, leading to midgut hyperplasia. Regeneration assays demonstrated that Chsy mutant ISCs failed to properly downregulate mitotic activity at the end of regeneration. We also found that CS is essential for the barrier integrity to prevent leakage of the midgut epithelium. CS is known to be polymerized by the action of the complex of Chsy and another critical protein, Chondroitin polymerizing factor (Chpf). We found that Chpf mutants show increased ISC division during midgut homeostasis and regeneration, similar to Chsy mutants. As Chpf is induced by a tissue damage during regeneration, our data suggest that Chpf functions with Chsy to facilitate CS remodeling and stimulate tissue repair. We propose that the completion of the repair of CS-containing BM acts as a prerequisite to properly terminate the regeneration process.
Collapse
Affiliation(s)
- Collin Knudsen
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ayano Moriya
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Eriko Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rishi Gulati
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Takuya Akiyama
- Department of Biology, The Porter Cancer Research Center, Indiana State University, Terre Haute, Indiana, United States of America
| | - Hiroshi Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
7
|
Sang Y, Ning X, Xu Q, Wang L, Yan Y, Zhang L, Bi X. Characterization of transcriptomics during aging and genes required for lifespan in Drosophila intestine. Sci Rep 2025; 15:14692. [PMID: 40287511 PMCID: PMC12033250 DOI: 10.1038/s41598-025-98888-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Aging is closely associated with imbalanced transcription. Regulated transcription in different organs is significantly different during aging, indicating that organ-specific transcriptomics is critical for understanding this process. Here we analyze the transcriptomics of the intestines of 3-, 15-, 30-, 40- and 50-days old female flies, which include young, middle-aged, and old flies. We find that the differential expression of protein-coding genes and lncRNAs is significant in aging, and fly age is characterized by well-separated gene expression trajectories. The highly clustered differentially expressed genes are connected to specific biological processes and signalling pathways. In particular, the Imd and Toll pathways are the top two immune signalling pathways that are highly regulated, and members with increased expression in the Imd pathway span all upstream activating events and include many ubiquitylation-associated factors and regulators of NF-κB factor Relish. Increased expression of Toll pathway members includes sensing mediators for all kinds of microorganisms and multiple proteases in the proteolytic processing cascade. Moreover, the expression of molecular markers of intestinal cells is greatly changed. Enterocyte markers are the most significantly influenced, and enteroendocrine markers AstA and NPF, as well as intestinal stem cell (ISC)/enteroblast (EB) markers Esg and Klu are expressed at low levels in young flies and much higher levels in aged flies. Furthermore, lncRNAs show similar expression trends and clustering patterns to those of protein-coding genes. Lastly, we find that ISC/EB-specific knock-down of 13 out of 19 genes that are highly differentially expressed reduces the lifespan of the fly. Together, the characterized transcriptomics and newly identified functional genes in aging will provide potential targets for preventing intestinal aging and associated disorders.
Collapse
Affiliation(s)
- Yan Sang
- Affiliated Hospital of Nantong University, School of Medicine, Nantong University, Nantong, 226001, China
| | - Xiufan Ning
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Qi Xu
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Lan Wang
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Yuhang Yan
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Lijiao Zhang
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Xiaolin Bi
- School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
8
|
Tsai M, Sun J, Alexandre C, Shapiro M, Franchet A, Li Y, Gould AP, Vincent JP, Stockinger B, Diny NL. Drosophila AHR limits tumor growth and stem cell proliferation in the intestine. Wellcome Open Res 2025; 10:38. [PMID: 40212817 PMCID: PMC11982807 DOI: 10.12688/wellcomeopenres.23515.2] [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] [Accepted: 04/23/2025] [Indexed: 04/29/2025] Open
Abstract
Background The aryl hydrocarbon receptor (AHR) plays important roles in intestinal homeostasis, limiting tumour growth and promoting differentiation in the intestinal epithelium. Spineless, the Drosophila homolog of AHR, has only been studied in the context of development but not in the adult intestine. Methods The role of Spineless in the Drosophila midgut was studied by overexpression or inactivation of Spineless in infection and tumour models and RNA sequencing of sorted midgut progenitor cells. Results We show that spineless is upregulated in the adult intestinal epithelium after infection with Pseudomonas entomophila ( P. e.). Spineless inactivation increased stem cell proliferation following infection-induced injury. Spineless overexpression limited intestinal stem cell proliferation and reduced survival after infection. In two tumour models, using either Notch RNAi or constitutively active Yorkie, Spineless suppressed tumour growth and doubled the lifespan of tumour-bearing flies. At the transcriptional level it reversed the gene expression changes induced in Yorkie tumours, counteracting cell proliferation and altered metabolism. Conclusions These findings demonstrate a new role for Spineless in the adult Drosophila midgut and highlight the evolutionarily conserved functions of AHR/Spineless in the control of proliferation and differentiation of the intestinal epithelium.
Collapse
Affiliation(s)
- Minghua Tsai
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Jiawei Sun
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | | | | | | | - Ying Li
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Alex P. Gould
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | | | | | - Nicola Laura Diny
- The Francis Crick Institute, London, England, NW1 1AT, UK
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, North Rhine-Westphalia, 53127, Germany
| |
Collapse
|
9
|
Li Y, Liu X, Guo L, Han K, Fang S, Wan X, Wang D, Xu X, Jiang L, Fan G, Xu M. SpaGRN: Investigating spatially informed regulatory paths for spatially resolved transcriptomics data. Cell Syst 2025; 16:101243. [PMID: 40179878 DOI: 10.1016/j.cels.2025.101243] [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: 11/27/2023] [Revised: 08/30/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Cells spatially organize into distinct cell types or functional domains through localized gene regulatory networks. However, current spatially resolved transcriptomics analyses fail to integrate spatial constraints and proximal cell influences, limiting the mechanistic understanding of tissue organization. Here, we introduce SpaGRN, a statistical framework that reconstructs cell-type- or functional-domain-specific, dynamic, and spatial regulons by coupling intracellular spatial regulatory causality with extracellular signaling path information. Benchmarking across synthetic and real datasets demonstrates SpaGRN's superior precision over state-of-the-art tools in identifying context-dependent regulons. Applied to diverse spatially resolved transcriptomics platforms (Stereo-seq, STARmap, MERFISH, CosMx, Slide-seq, and 10x Visium), complex cancerous samples, and 3D datasets of developing Drosophila embryos and larvae, SpaGRN not only provides a versatile toolkit for decoding receptor-mediated spatial regulons but also reveals spatiotemporal regulatory mechanisms underlying organogenesis and inflammation.
Collapse
Affiliation(s)
- Yao Li
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China
| | | | - Lidong Guo
- BGI Research, Qingdao 266555, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Han
- BGI Research, Qingdao 266555, China
| | - Shuangsang Fang
- BGI Research, Beijing 102601, China; BGI Research, Shenzhen 518083, China
| | - Xinjiang Wan
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, and Center of Deep Sea Research Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | | | - Xun Xu
- BGI Research, Wuhan 430074, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Ling Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Guangyi Fan
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| | - Mengyang Xu
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| |
Collapse
|
10
|
Ahrentløv N, Kubrak O, Lassen M, Malita A, Koyama T, Frederiksen AS, Sigvardsen CM, John A, Madsen PEH, Halberg KV, Nagy S, Imig C, Richter EA, Texada MJ, Rewitz K. Protein-responsive gut hormone tachykinin directs food choice and impacts lifespan. Nat Metab 2025:10.1038/s42255-025-01267-0. [PMID: 40229448 DOI: 10.1038/s42255-025-01267-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/06/2025] [Indexed: 04/16/2025]
Abstract
Animals select food based on hungers that reflect dynamic macronutrient needs, but the hormonal mechanisms underlying nutrient-specific appetite regulation remain poorly defined. Here, we identify tachykinin (Tk) as a protein-responsive gut hormone in Drosophila and female mice, regulated by conserved environmental and nutrient-sensing mechanisms. Protein intake activates Tk-expressing enteroendocrine cells (EECs), driving the release of gut Tk through mechanisms involving target of rapamycin (TOR) and transient receptor potential A1 (TrpA1). In flies, we delineate a pathway by which gut Tk controls selective appetite and sleep after protein ingestion, mediated by glucagon-like adipokinetic hormone (AKH) signalling to neurons and adipose tissue. This mechanism suppresses protein appetite, promotes sugar hunger and modulates wakefulness to align behaviour with nutritional needs. Inhibiting protein-responsive gut Tk prolongs lifespan through AKH, revealing a role for nutrient-dependent gut hormone signalling in longevity. Our results provide a framework for understanding EEC-derived nutrient-specific satiety signals and the role of gut hormones in regulating food choice, sleep and lifespan.
Collapse
Affiliation(s)
- Nadja Ahrentløv
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Olga Kubrak
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mette Lassen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Alina Malita
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Amalie S Frederiksen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Casper M Sigvardsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Alphy John
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Cordelia Imig
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation, Hellerup, Denmark
| | - Erik A Richter
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Puig-Barbe A, Dettmann S, Nirello VD, Moor H, Azami S, Edgar BA, Varga-Weisz P, Korzelius J, de Navascués J. A bHLH interaction code controls bipotential differentiation and self-renewal in the Drosophila gut. Cell Rep 2025; 44:115398. [PMID: 40089983 DOI: 10.1016/j.celrep.2025.115398] [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: 09/19/2023] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/18/2025] Open
Abstract
Multipotent adult stem cells balance self-renewal with differentiation into various cell types. How this balance is regulated at the transcriptional level is poorly understood. Here, we show that a network of basic helix-loop-helix (bHLH) transcription factors controls both stemness and bipotential differentiation in the Drosophila adult intestine. We find that homodimers of Daughterless (Da), a homolog of mammalian E proteins, maintain self-renewal of intestinal stem cells (ISCs), antagonizing the enteroendocrine fate promoted by heterodimers of Da and Scute (Sc; homolog of ASCL). The HLH factor Extramacrochaetae (Emc; homologous to Id proteins) promotes absorptive differentiation by titrating Da and Sc. Emc prevents the committed absorptive progenitor from dedifferentiating, underscoring the plasticity of these cells. Switching physical interaction partners in this way enables the active maintenance of stemness while priming stem cells for differentiation along two alternative fates. Such regulatory logic is likely operative in other bipotent stem cell systems.
Collapse
Affiliation(s)
- Aleix Puig-Barbe
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Svenja Dettmann
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; AbbVie Germany GmbH & Co. KG, 81 Mainzer Str., 65189 Wiesbaden, Frankfurt, Germany
| | - Vinícius Dias Nirello
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Helen Moor
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Sina Azami
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Straße 9B, 50931 Köln, Germany
| | - Bruce A Edgar
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Huntsman Cancer Institute & Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Patrick Varga-Weisz
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Jerome Korzelius
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Straße 9B, 50931 Köln, Germany; School of Biosciences, University of Kent, Canterbury CT2 7NZ, UK
| | - Joaquín de Navascués
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK.
| |
Collapse
|
12
|
Niosi A, Võ NH, Sundaramurthy P, Welch C, Penn A, Yuldasheva Y, Alfareh A, Rausch K, Amin-Rahbar T, Cavanaugh J, Yadav P, Peterson S, Brown R, Hu A, Ardon-Castro A, Nguyen D, Crawford R, Lee W, Morris EJ, Jensen MH, Mulligan K. Kismet/CHD7/CHD8 affects gut microbiota, mechanics, and the gut-brain axis in Drosophila melanogaster. Biophys J 2025; 124:933-941. [PMID: 38902926 PMCID: PMC11947469 DOI: 10.1016/j.bpj.2024.06.016] [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: 01/20/2024] [Revised: 05/17/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
The gut microbiome affects brain and neuronal development and may contribute to the pathophysiology of neurodevelopmental disorders. However, it is unclear how risk genes associated with such disorders affect gut physiology in a manner that could impact microbial colonization and how the mechanical properties of the gut tissue might play a role in gut-brain bidirectional communication. To address this, we used Drosophila melanogaster with a null mutation in the gene kismet, an ortholog of chromodomain helicase DNA-binding protein (CHD) family members CHD7 and CHD8. In humans, these are risk genes for neurodevelopmental disorders with co-occurring gastrointestinal symptoms. We found that kismet mutant flies have a significant increase in gastrointestinal transit time, indicating the functional homology of kismet with CHD7/CHD8 in vertebrates. Rheological characterization of dissected gut tissue revealed significant changes in the mechanics of kismet mutant gut elasticity, strain stiffening behavior, and tensile strength. Using 16S rRNA metagenomic sequencing, we also found that kismet mutants have reduced diversity and abundance of gut microbiota at every taxonomic level. To investigate the connection between the gut microbiome and behavior, we depleted gut microbiota in kismet mutant and control flies and quantified the flies' courtship behavior. Depletion of gut microbiota rescued courtship defects of kismet mutant flies, indicating a connection between gut microbiota and behavior. In striking contrast, depletion of the gut microbiome in the control strain reduced courtship activity, demonstrating that antibiotic treatment can have differential impacts on behavior and may depend on the status of microbial dysbiosis in the gut prior to depletion. We propose that Kismet influences multiple gastrointestinal phenotypes that contribute to the gut-microbiome-brain axis to influence behavior. We also suggest that gut tissue mechanics should be considered as an element in the gut-brain communication loop, both influenced by and potentially influencing the gut microbiome and neurodevelopment.
Collapse
Affiliation(s)
- Angelo Niosi
- Department of Biological Sciences, California State University, Sacramento, California
| | - Nguyên Henry Võ
- Department of Biological Sciences, California State University, Sacramento, California
| | | | - Chloe Welch
- Department of Biological Sciences, California State University, Sacramento, California
| | - Aliyah Penn
- Department of Biological Sciences, California State University, Sacramento, California
| | - Yelena Yuldasheva
- Department of Biological Sciences, California State University, Sacramento, California
| | - Adam Alfareh
- Department of Biological Sciences, California State University, Sacramento, California
| | - Kaitlyn Rausch
- Department of Biological Sciences, California State University, Sacramento, California
| | - Takhmina Amin-Rahbar
- Department of Biological Sciences, California State University, Sacramento, California
| | - Jeffery Cavanaugh
- Department of Physics and Astronomy, California State University, Sacramento, California
| | - Prince Yadav
- Department of Physics and Astronomy, California State University, Sacramento, California
| | - Stephanie Peterson
- Department of Biological Sciences, California State University, Sacramento, California
| | - Raina Brown
- Department of Biological Sciences, California State University, Sacramento, California
| | - Alain Hu
- Department of Biological Sciences, California State University, Sacramento, California
| | - Any Ardon-Castro
- Department of Biological Sciences, California State University, Sacramento, California
| | - Darren Nguyen
- Department of Biological Sciences, California State University, Sacramento, California
| | - Robert Crawford
- Department of Biological Sciences, California State University, Sacramento, California
| | - Wendy Lee
- Department of Computer Science, San Jose State University, San Jose, California
| | - Eliza J Morris
- Department of Physics and Astronomy, California State University, Sacramento, California
| | - Mikkel Herholdt Jensen
- Department of Physics and Astronomy, California State University, Sacramento, California.
| | - Kimberly Mulligan
- Department of Biological Sciences, California State University, Sacramento, California.
| |
Collapse
|
13
|
Nguyen QD, Fujii K, Ishibashi K, Hashiba H, Ohtsubo W, Kitazawa H, Tanimoto H, Fuse N, Kurata S. Regulation of Gut Starvation Responses Through Drosophila NP3253 Neurons. Genes Cells 2025; 30:e70005. [PMID: 39904737 DOI: 10.1111/gtc.70005] [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: 11/22/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
The "gut-brain axis," a bidirectional communication system between the gastrointestinal tract and the central nervous system, plays a crucial role in regulating complex physiological functions in response to nutrients, pathogens, and microbiota. However, the cellular and molecular mechanisms governing this regulation remain poorly understood. Using Drosophila melanogaster as a model organism, we previously identified NP3253 neurons, located in both the brain and gut, as key contributors to gut homeostasis during oral bacterial infection. Here, we found a novel role of NP3253 neurons in regulating starvation resistance. We observed that a subset of NP3253 neurons in the gut were activated during starvation. To investigate downstream effect, we conducted RNA-Seq analysis on the gut of adult flies with genetically silenced NP3253 neurons, comparing gene expression under starved and fed conditions. This analysis identified 26 genes differentially expressed in response to both starvation and NP3253 neuronal activity. Among these, CG12643, encoding an uncharacterized short peptide, was found to be essential for starvation resistance in the gut. Our findings demonstrate that NP3253 neurons modulate the gut gene expression in response to starvation, thereby supporting physiological adaptation to environmental stressors.
Collapse
Affiliation(s)
- Quang-Dat Nguyen
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kiho Fujii
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kentaro Ishibashi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haruka Hashiba
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wakako Ohtsubo
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Haruki Kitazawa
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Naoyuki Fuse
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
14
|
Hurcomb JD, Mukherjee A, Lindell AE, Popovic R, Yu Y, Patil KR, Loh SHY, Martins LM. Oral administration of aripiprazole to Drosophila causes intestinal toxicity. Dis Model Mech 2025; 18:dmm052180. [PMID: 40126029 PMCID: PMC11972071 DOI: 10.1242/dmm.052180] [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: 06/04/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Aripiprazole is a third-generation antipsychotic medication that was introduced to mitigate the poor tolerability of older antipsychotics. In contrast to the older antipsychotic drugs that act as dopamine receptor antagonists in the brain, aripiprazole functions as a partial agonist. Aripiprazole has been identified as an off-target inhibitor of mitochondrial respiratory complex I. We observed that patients prescribed aripiprazole often report gastrointestinal disturbances, but the mechanism underlying these side effects is not clear. We modelled the potential mitochondrial toxicity of aripiprazole in the gastrointestinal system using the fruit fly (Drosophila melanogaster). Aripiprazole consumption impaired Drosophila gut function and faecal output. It also reduced the mitochondrial membrane potential and increased reactive oxygen species (ROS) levels in intestinal cells. ROS activate the c-Jun N-terminal kinase (JNK) pathway, which induces cellular stress and cell death. Aripiprazole increased JNK activation in the intestinal cells of flies, resulting in cell death, which was suppressed by antioxidants. We conclude that aripiprazole activates the JNK pathway of cell death via mitochondrial ROS production. Using antioxidant supplements may help reduce aripiprazole-induced toxicity.
Collapse
Affiliation(s)
- James D. Hurcomb
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Amrita Mukherjee
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Anna E. Lindell
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Rebeka Popovic
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kiran R. Patil
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Samantha H. Y. Loh
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - L. Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
15
|
Sun C, Shao Y, Iqbal J. A comprehensive cell atlas of fall armyworm (Spodoptera frugiperda) larval gut and fat body via snRNA-Seq. Sci Data 2025; 12:250. [PMID: 39939604 PMCID: PMC11822134 DOI: 10.1038/s41597-025-04520-z] [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: 11/11/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
The midgut and fat body of insects control key physiological processes, including growth, digestion, metabolism, and stress response. Single-nucleus RNA sequencing (snRNA-seq) is a promising way to reveal organ complexity at the cellular level, yet data for lepidopteran insects are lacking. We utilized snRNA-seq to assess cellular diversity in the midgut and fat body of Spodoptera frugiperda. Our study identified 20 distinct clusters in the midgut, including enterocytes, enteroendocrine, stem-like cells, and muscle cells, and 27 clusters in the fat body, including adipocytes, hemocytes, and epithelial cells. This dataset, containing all identified cell types in midgut and fat body, is valuable for characterizing the cellular composition of these organs and uncovering new cell-specific biomarkers. This cellular atlas enhances our understanding of cellular heterogeneity of fat and midgut, serving as a basis for future functional and comparative analyses. As the first snRNA-seq study on the midgut and fat body of S. frugiperda, it will also support future research, contribute to lepidopteran studies, and aid in developing targeted pest control strategies.
Collapse
Affiliation(s)
- Chao Sun
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yongqi Shao
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Junaid Iqbal
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
16
|
Blanco Mendana J, Donovan M, O'Brien LG, Auch B, Garbe J, Gohl DM. Deterministic genetic barcoding for multiplexed behavioral and single-cell transcriptomic studies. eLife 2025; 12:RP88334. [PMID: 39908076 PMCID: PMC11798575 DOI: 10.7554/elife.88334] [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: 02/06/2025] Open
Abstract
Advances in single-cell sequencing technologies have provided novel insights into the dynamics of gene expression and cellular heterogeneity within tissues and have enabled the construction of transcriptomic cell atlases. However, linking anatomical information to transcriptomic data and positively identifying the cell types that correspond to gene expression clusters in single-cell sequencing data sets remains a challenge. We describe a straightforward genetic barcoding approach that takes advantage of the powerful genetic tools in Drosophila to allow in vivo tagging of defined cell populations. This method, called Targeted Genetically-Encoded Multiplexing (TaG-EM), involves inserting a DNA barcode just upstream of the polyadenylation site in a Gal4-inducible UAS-GFP construct so that the barcode sequence can be read out during single-cell sequencing, labeling a cell population of interest. By creating many such independently barcoded fly strains, TaG-EM enables positive identification of cell types in cell atlas projects, identification of multiplet droplets, and barcoding of experimental timepoints, conditions, and replicates. Furthermore, we demonstrate that TaG-EM barcodes can be read out using next-generation sequencing to facilitate population-scale behavioral measurements. Thus, TaG-EM has the potential to enable large-scale behavioral screens in addition to improving the ability to multiplex and reliably annotate single-cell transcriptomic experiments.
Collapse
Affiliation(s)
| | - Margaret Donovan
- University of Minnesota Genomics Center, MinneapolisMinneapolisUnited States
| | | | - Benjamin Auch
- University of Minnesota Genomics Center, MinneapolisMinneapolisUnited States
| | - John Garbe
- University of Minnesota Genomics Center, MinneapolisMinneapolisUnited States
| | - Daryl M Gohl
- University of Minnesota Genomics Center, MinneapolisMinneapolisUnited States
- Department of Genetics, Cell Biology, and Development, University of MinnesotaMinneapolisUnited States
| |
Collapse
|
17
|
Eldridge-Thomas BL, Bohere JG, Roubinet C, Barthelemy A, Samuels TJ, Teixeira FK, Kolahgar G. The transmembrane protein Syndecan is required for stem cell survival and maintenance of their nuclear properties. PLoS Genet 2025; 21:e1011586. [PMID: 39913561 PMCID: PMC11819509 DOI: 10.1371/journal.pgen.1011586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 02/12/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Tissue maintenance is underpinned by resident stem cells whose activity is modulated by microenvironmental cues. Using Drosophila as a simple model to identify regulators of stem cell behaviour and survival in vivo, we have identified novel connections between the conserved transmembrane proteoglycan Syndecan, nuclear properties and stem cell function. In the Drosophila midgut, Syndecan depletion in intestinal stem cells results in their loss from the tissue, impairing tissue renewal. At the cellular level, Syndecan depletion alters cell and nuclear shape, and causes nuclear lamina invaginations and DNA damage. In a second tissue, the developing Drosophila brain, live imaging revealed that Syndecan depletion in neural stem cells results in nuclear envelope remodelling defects which arise upon cell division. Our findings reveal a new role for Syndecan in the maintenance of nuclear properties in diverse stem cell types.
Collapse
Affiliation(s)
- Buffy L. Eldridge-Thomas
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Jerome G. Bohere
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Chantal Roubinet
- Université de Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, ERL U1305, Rennes, France
| | - Alexandre Barthelemy
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Tamsin J. Samuels
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Felipe Karam Teixeira
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Golnar Kolahgar
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Hu Y, Comjean A, Rodiger J, Chen W, Kim AR, Qadiri M, Gao C, Zirin J, Mohr S, Perrimon N. FlyRNAi.org 2025 update-expanded resources for new technologies and species. Nucleic Acids Res 2025; 53:D958-D965. [PMID: 39435987 PMCID: PMC11701652 DOI: 10.1093/nar/gkae917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024] Open
Abstract
The design, analysis and mining of large-scale 'omics studies with the goal of advancing biological and biomedical understanding require use of a range of bioinformatics tools, including approaches tailored to needs specific to a given species and/or technology. The FlyRNAi database at the Drosophila RNAi Screening Center and Transgenic RNAi Project (DRSC/TRiP) Functional Genomics Resources (https://fgr.hms.harvard.edu/tools) supports an increasingly broad group of technologies and species. Recently, for example, we expanded the database to include additional new data-centric resources that facilitate mining and analysis of single-cell transcriptomics. In addition, we have applied our approaches to CRISPR reagent and gene-centric bioinformatics approaches in Drosophila to arthropod vectors of infectious diseases. Building on our previous comprehensive reports on the FlyRNAi database, here we focus on new and updated resources with a primary focus on data-centric tools. Altogether, our suite of online resources supports various stages of functional genomics studies for Drosophila and other arthropods, and facilitate a wide range of reagent design, analysis, data mining and analysis approaches by biologists and biomedical experts studying Drosophila, other common genetic model species, arthropod vectors and/or human biology.
Collapse
Affiliation(s)
- Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jonathan Rodiger
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- LifeMine Therapeutics, 30 Acorn Park Dr, Cambridge, MA 02140, USA
| | - Weihang Chen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ah-Ram Kim
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Mujeeb Qadiri
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Chenxi Gao
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jonathan Zirin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Stephanie E Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
19
|
Fitzmeyer EA, Dutt TS, Pinaud S, Graham B, Gallichotte EN, Hill JL, Campbell CL, Ogg H, Howick V, Lawniczak MKN, Nishimura EO, Merkling SH, Henao-Tamayo M, Ebel GD. A single-cell atlas of the Culex tarsalis midgut during West Nile virus infection. PLoS Pathog 2025; 21:e1012855. [PMID: 39869679 PMCID: PMC11793825 DOI: 10.1371/journal.ppat.1012855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/04/2025] [Accepted: 12/20/2024] [Indexed: 01/29/2025] Open
Abstract
The mosquito midgut functions as a key interface between pathogen and vector. However, studies of midgut physiology and virus infection dynamics are scarce, and in Culex tarsalis-an extremely efficient vector of West Nile virus (WNV)-nonexistent. We performed single-cell RNA sequencing on Cx. tarsalis midguts, defined multiple cell types, and determined whether specific cell types are more permissive to WNV infection. We identified 20 cell states comprising 8 distinct cell types, consistent with existing descriptions of Drosophila and Aedes aegypti midgut physiology. Most midgut cell populations were permissive to WNV infection. However, there were higher levels of WNV RNA (vRNA) in enteroendocrine cells (EE), suggesting enhanced replication in this population. In contrast, proliferating intestinal stem cells (ISC) had the lowest levels of vRNA, a finding consistent with studies suggesting ISC proliferation in the midgut is involved in infection control. ISCs were also found to have a strong transcriptional response to WNV infection; genes involved in ribosome structure and biogenesis, and translation were significantly downregulated in WNV-infected ISC populations. Notably, we did not detect significant WNV-infection induced upregulation of canonical mosquito antiviral immune genes (e.g., AGO2, R2D2, etc.) at the whole-midgut level. Rather, we observed a significant positive correlation between immune gene expression levels and vRNA load in individual cells, suggesting that within midgut cells, high levels of vRNA may trigger antiviral responses. Our findings establish a Cx. tarsalis midgut cell atlas, and provide insight into midgut infection dynamics of WNV by characterizing cell-type specific enhancement/restriction of, and immune response to, infection at the single-cell level.
Collapse
Affiliation(s)
- Emily A. Fitzmeyer
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Taru S. Dutt
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Silvain Pinaud
- MIVEGEC, Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Barb Graham
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Emily N. Gallichotte
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Jessica L. Hill
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Corey L. Campbell
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Hunter Ogg
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Virginia Howick
- School of Biodiversity, One Health and Veterinary Medicine, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | | | - Erin Osborne Nishimura
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah Hélène Merkling
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Marcela Henao-Tamayo
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Gregory D. Ebel
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
20
|
Fitzmeyer EA, Dutt TS, Pinaud S, Graham B, Gallichotte EN, Hill JL, Campbell CL, Ogg H, Howick V, Lawniczak MKN, Osborne Nishimura E, Merkling SH, Henao-Tamayo M, Ebel GD. A single-cell atlas of the Culex tarsalis midgut during West Nile virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.603613. [PMID: 39091762 PMCID: PMC11291174 DOI: 10.1101/2024.07.23.603613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The mosquito midgut functions as a key interface between pathogen and vector. However, studies of midgut physiology and virus infection dynamics are scarce, and in Culex tarsalis - an extremely efficient vector of West Nile virus (WNV) - nonexistent. We performed single-cell RNA sequencing on Cx. tarsalis midguts, defined multiple cell types, and determined whether specific cell types are more permissive to WNV infection. We identified 20 cell states comprising 8 distinct cell types, consistent with existing descriptions of Drosophila and Aedes aegypti midgut physiology. Most midgut cell populations were permissive to WNV infection. However, there were higher levels of WNV RNA (vRNA) in enteroendocrine cells, suggesting enhanced replication in this population. In contrast, proliferating intestinal stem cells (ISC) had the lowest levels of vRNA, a finding consistent with studies suggesting ISC proliferation in the midgut is involved in infection control. ISCs were also found to have a strong transcriptional response to WNV infection; genes involved in ribosome structure and biogenesis, and translation were significantly downregulated in WNV-infected ISC populations. Notably, we did not detect significant WNV-infection induced upregulation of canonical mosquito antiviral immune genes (e.g., AGO2, R2D2, etc.) at the whole-midgut level. Rather, we observed a significant positive correlation between immune gene expression levels and vRNA load in individual cells, suggesting that within midgut cells, high levels of vRNA may trigger antiviral responses. Our findings establish a Cx. tarsalis midgut cell atlas, and provide insight into midgut infection dynamics of WNV by characterizing cell-type specific enhancement/restriction of, and immune response to, infection at the single-cell level.
Collapse
Affiliation(s)
- Emily A. Fitzmeyer
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Taru S. Dutt
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Silvain Pinaud
- MIVEGEC, Université de Montpellier, IRD, CNRS, Montpellier, France
| | - Barb Graham
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Emily N. Gallichotte
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Jessica L. Hill
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Corey L. Campbell
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Hunter Ogg
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Virginia Howick
- School of Biodiversity, One Health and Veterinary Medicine, Wellcome Centre for Integrative Parasitology, University of Glasgow, UK
| | | | - Erin Osborne Nishimura
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah Hélène Merkling
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, 75015 Paris, France
| | - Marcela Henao-Tamayo
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Gregory D. Ebel
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
21
|
Yan Y, Cheung E, Verzier LH, Appetecchia F, March S, Craven AR, Du E, Probst AS, Rinvee TA, de Vries LE, Kauffman J, Bhatia SN, Nelson E, Singh N, Peng D, Shaw WR, Catteruccia F. Mapping Plasmodium transitions and interactions in the Anopheles female. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623125. [PMID: 39605504 PMCID: PMC11601300 DOI: 10.1101/2024.11.12.623125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The human malaria parasite, Plasmodium falciparum , relies on Anopheles mosquitoes for transmission. Once ingested during blood feeding, most parasites die in the mosquito midgut lumen or during epithelium traversal. How surviving ookinetes interact with midgut cells and form oocysts is unknown, yet these steps are essential to initiate a remarkable, similarly uncharacterized growth process culminating in the production of thousands of infectious sporozoites. Here, using single-cell RNA sequencing of both parasites and mosquito cells across four time points and two metabolic conditions, we unveil key processes shaping developmental transitions and mosquito-parasite interactions occurring in the midgut. In depth functional analyses reveal processes regulating oocyst growth and identify the transcription factor Pf SIP2 as essential for sporozoite infection of human hepatocytes. By combining the analysis of shared mosquito-parasite barcodes with confocal microscopy, we discover that parasites preferentially interact with midgut progenitor cells during epithelial crossing, potentially using their basal location as an exit landmark. Additionally, we unveil tight connections between extracellular late oocysts and surrounding muscle cells that may ensure parasites adhere to the midgut without damaging it. Ultimately, our study provides fundamental insight into the molecular events characterizing previously inaccessible biological transitions and mosquito-parasite interactions, and identifies candidates for transmission-blocking strategies.
Collapse
|
22
|
Fei S, Awais MM, Zou J, Xia J, Wang Y, Kong Y, Feng M, Sun J. Single-nucleus RNA sequencing reveals midgut cellular heterogeneity and transcriptional profiles in Bombyx mori cytoplasmic polyhedrosis virus infection. INSECT SCIENCE 2024. [PMID: 39523555 DOI: 10.1111/1744-7917.13464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024]
Abstract
The gut is not only used by insects as an organ for the digestion of food and absorption of nutrients but also as an important barrier against the invasion and proliferation of pathogenic microorganisms. Bombyx mori cytoplasmic polyhedrosis virus (BmCPV), an insect-specific virus, predominantly colonizes the midgut epithelial cells of the silkworm, thereby jeopardizing its normal growth. However, there is limited knowledge of the cellular immune responses to viral infection and whether the infection is promoted or inhibited by different types of cells in the silkworm midgut. In this study, we used single-nucleus RNA sequencing to identify representative enteroendocrine cells, enterocytes, and muscle cell types in the silkworm midgut. In addition, by analyzing the transcriptional profiles of various subpopulations in the infected and uninfected groups, we found that BmCPV infection suppresses the response of the antiviral pathways and induces the expression of BmHSP70, which plays a role in promoting BmCPV replication. However, certain immune genes in the midgut of the silkworm, such as BmLebocin3, were induced upon viral infection, and downregulation of BmLEB3 using RNA interference promoted BmCPV replication in the midgut of B. mori. These results suggest that viral immune evasion and active host resistance coexist in BmCPV-infected silkworms. We reveal the richness of cellular diversity in the midgut of B. mori larvae by single-nucleus RNA sequencing analysis and provide new insights into the complex interactions between the host and the virus at the single-cell level.
Collapse
Affiliation(s)
- Shigang Fei
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mian Muhammad Awais
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jinglei Zou
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Junming Xia
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yeyuan Wang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yibing Kong
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
23
|
Chen J, Nouzová M, Noriega FG, Tatar M. Gut-to-brain regulation of Drosophila aging through neuropeptide F, insulin, and juvenile hormone. Proc Natl Acad Sci U S A 2024; 121:e2411987121. [PMID: 39413128 PMCID: PMC11513968 DOI: 10.1073/pnas.2411987121] [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: 06/26/2024] [Accepted: 09/09/2024] [Indexed: 10/18/2024] Open
Abstract
Dietary restriction (DR) slows aging in many animals, while in some cases, the sensory signals from diet alone are sufficient to retard or accelerate lifespan. The digestive tract is a candidate location to sense nutrients, where neuropeptides secreted by enteroendocrine cells (EEC) produce systemic signals in response to food. Here, we measure how Drosophila neuropeptide F (NPF) is secreted into adult circulation by EEC and find that specific EEC differentially respond to dietary sugar and yeast. Female lifespan is increased when gut NPF is genetically depleted, and this manipulation is sufficient to blunt the longevity benefit conferred by DR. Depletion of NPF receptors at insulin-producing neurons of the brain also increases female lifespan, consistent with observations where loss of gut NPF decreases neuronal insulin secretion. The longevity conferred by repressing gut NPF and brain NPF receptors is reversed by treating adults with a juvenile hormone (JH) analog. JH is produced by the adult corpora allata, and inhibition of the insulin receptor at this tissue decreases JH titer and extends lifespan in both males and females, while this longevity is restored to wild type by treating adults with a JH analog. Overall, EEC of the gut modulate Drosophila aging through interorgan communication mediated by a gut-brain-corpora allata axis, and insulin produced in the brain impacts lifespan through its control of JH titer. These data suggest that we consider how human incretins and their analogs, which are used to treat obesity and diabetes, may impact aging.
Collapse
Affiliation(s)
- Jiangtian Chen
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI02912
| | - Marcela Nouzová
- Institute of Parasitology, Laboratory of Molecular Biology and Physiology of Mosquitoes, Biology Centre Czech Academy of Sciences, České Budějovice37005, Czech Republic
| | - Fernando G. Noriega
- Department of Biological Sciences and Biomolecular Sciences Institute, Florida International University, Miami, FL33199
- Department of Parasitology, University of South Bohemia, České Budějovice37005, Czech Republic
| | - Marc Tatar
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI02912
| |
Collapse
|
24
|
Vonolfen MC, Meyer Zu Altenschildesche FL, Nam HJ, Brodesser S, Gyenis A, Buellesbach J, Lam G, Thummel CS, Storelli G. Drosophila HNF4 acts in distinct tissues to direct a switch between lipid storage and export in the gut. Cell Rep 2024; 43:114693. [PMID: 39235946 DOI: 10.1016/j.celrep.2024.114693] [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: 03/26/2024] [Revised: 07/15/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Nutrient digestion, absorption, and export must be coordinated in the gut to meet the nutritional needs of the organism. We used the Drosophila intestine to characterize the mechanisms that coordinate the fate of dietary lipids. We identified enterocytes specialized in absorbing and exporting lipids to peripheral organs. Distinct hepatocyte-like cells, called oenocytes, communicate with these enterocytes to adjust intestinal lipid storage and export. A single transcription factor, Drosophila hepatocyte nuclear factor 4 (dHNF4), supports this gut-liver axis. In enterocytes, dHNF4 maximizes dietary lipid export by preventing their sequestration in cytoplasmic lipid droplets. In oenocytes, dHNF4 promotes the expression of the insulin antagonist ImpL2 to activate Foxo and suppress lipid retention in enterocytes. Disruption of this switch between lipid storage and export is associated with intestinal inflammation, suggesting a lipidic origin for inflammatory bowel diseases. These studies establish dHNF4 as a central regulator of intestinal metabolism and inter-organ lipid trafficking.
Collapse
Affiliation(s)
- Maximilian C Vonolfen
- University of Cologne, Faculty of Mathematics and Natural Sciences, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Fenja L Meyer Zu Altenschildesche
- University of Cologne, Faculty of Mathematics and Natural Sciences, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Hyuck-Jin Nam
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5330, USA
| | - Susanne Brodesser
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Akos Gyenis
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Jan Buellesbach
- Institute for Evolution & Biodiversity, University of Münster, Hüfferstrasse 1, 48149 Münster, Germany
| | - Geanette Lam
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5330, USA
| | - Carl S Thummel
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112-5330, USA
| | - Gilles Storelli
- University of Cologne, Faculty of Mathematics and Natural Sciences, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute for Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
25
|
Hodgson JJ, Chen RY, Blissard GW, Buchon N. Viral and cellular determinants of polarized trafficking of viral envelope proteins from insect-specific and insect-vectored viruses in insect midgut and salivary gland cells. J Virol 2024; 98:e0054024. [PMID: 39162433 PMCID: PMC11406959 DOI: 10.1128/jvi.00540-24] [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: 03/21/2024] [Accepted: 07/09/2024] [Indexed: 08/21/2024] Open
Abstract
Systemic viral infection of insects typically begins with the primary infection of midgut epithelial cells (enterocytes) and subsequent transit of the progeny virus in an apical-to-basal orientation into the hemocoel. For insect-vectored viruses, an oppositely oriented process (basal-to-apical transit) occurs upon secondary infection of salivary glands and is necessary for virus transmission to non-insect hosts. To examine this inversely oriented virus transit in these polarized tissues, we assessed the intracellular trafficking of two model viral envelope proteins (baculovirus GP64 and vesicular stomatitis virus G) in the midgut and salivary gland cells of the model insect, Drosophila melanogaster. Using fly lines that inducibly express either GP64 or VSV G, we found that each protein, expressed alone, was trafficked basally in midgut enterocytes. In salivary gland cells, VSV G was trafficked apically in most but not all cells, whereas GP64 was consistently trafficked basally. We demonstrated that a YxxØ motif present in both proteins was critical for basal trafficking in midgut enterocytes but dispensable for trafficking in salivary gland cells. Using RNAi, we found that clathrin adaptor protein complexes AP-1 and AP-3, as well as seven Rab GTPases, were involved in polarized VSV G trafficking in midgut enterocytes. Our results indicate that these viral envelope proteins encode the requisite information and require no other viral factors for appropriately polarized trafficking. In addition, they exploit tissue-specific differences in protein trafficking pathways to facilitate virus egress in the appropriate orientation for establishing systemic infections and vectoring infection to other hosts. IMPORTANCE Viruses that use insects as hosts must navigate specific routes through different insect tissues to complete their life cycles. The routes may differ substantially depending on the life cycle of the virus. Both insect pathogenic viruses and insect-vectored viruses must navigate through the polarized cells of the midgut epithelium to establish a systemic infection. In addition, insect-vectored viruses must also navigate through the polarized salivary gland epithelium for transmission. Thus, insect-vectored viruses appear to traffic in opposite directions in these two tissues. In this study, we asked whether two viral envelope proteins (VSV G and baculovirus GP64) alone encode the signals necessary for the polarized trafficking associated with their respective life cycles. Using Drosophila as a model to examine tissue-specific polarized trafficking of these viral envelope proteins, we identified one of the virus-encoded signals and several host proteins associated with regulating the polarized trafficking in the midgut epithelium.
Collapse
Affiliation(s)
- Jeffrey J. Hodgson
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
- Boyce Thompson Institute at Cornell University, Ithaca, New York, USA
| | - Robin Y. Chen
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| | - Gary W. Blissard
- Boyce Thompson Institute at Cornell University, Ithaca, New York, USA
| | - Nicolas Buchon
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| |
Collapse
|
26
|
Zhang X, Zhang Q, Song X, Yang W, Cheng A, Zhang J, Dong W. Toxicity Evaluation of Potassium Sorbate In Vivo with Drosophila Melanogaster. INSECTS 2024; 15:703. [PMID: 39336671 PMCID: PMC11432522 DOI: 10.3390/insects15090703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Potassium sorbate (PS) is a preservative widely used in the food, pharmaceutical, and cosmetics industries. Improper and careless use of PS can lead to various health issues and potential environmental problems. Drosophila is capable of making rapid and sensitive responses to stress or other stimuli. Here we utilized Drosophila as a model organism to evaluate the potential toxicity of PS. Our study revealed that PS ingestion reduced the lifespan and fecundity of Drosophila. In addition, excessive PS ingestion led to cell apoptosis and ROS accumulation in the midgut. Furthermore, PS intake also enhanced the mitophagy of midgut cells. Strikingly, PS affected the cell differentiation progression as well, leading to the production of more enteroendocrine (EE) cells. We further demonstrated that the expression of notch (N), a vital player in intestinal stem cell (ISC) differentiation, was down-regulated in the midgut. This indicates that the differentiation progression was affected potentially by repressing the N expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Dong
- Shanxi Key Laboratory of Nucleic Acid Biopesticides, Research Institute of Applied Biology, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
27
|
Jena SG, Verma A, Engelhardt BE. Answering open questions in biology using spatial genomics and structured methods. BMC Bioinformatics 2024; 25:291. [PMID: 39232666 PMCID: PMC11375982 DOI: 10.1186/s12859-024-05912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
Genomics methods have uncovered patterns in a range of biological systems, but obscure important aspects of cell behavior: the shapes, relative locations, movement, and interactions of cells in space. Spatial technologies that collect genomic or epigenomic data while preserving spatial information have begun to overcome these limitations. These new data promise a deeper understanding of the factors that affect cellular behavior, and in particular the ability to directly test existing theories about cell state and variation in the context of morphology, location, motility, and signaling that could not be tested before. Rapid advancements in resolution, ease-of-use, and scale of spatial genomics technologies to address these questions also require an updated toolkit of statistical methods with which to interrogate these data. We present a framework to respond to this new avenue of research: four open biological questions that can now be answered using spatial genomics data paired with methods for analysis. We outline spatial data modalities for each open question that may yield specific insights, discuss how conflicting theories may be tested by comparing the data to conceptual models of biological behavior, and highlight statistical and machine learning-based tools that may prove particularly helpful to recover biological understanding.
Collapse
Affiliation(s)
- Siddhartha G Jena
- Department of Stem Cell and Regenerative Biology, Harvard, 7 Divinity Ave, Cambridge, MA, USA
| | - Archit Verma
- Gladstone Institutes, 1650 Owens Street, San Francisco, CA, 94158, USA
| | | |
Collapse
|
28
|
Arya SK, Harrison DA, Palli SR. Deciphering cellular heterogeneity in Spodoptera frugiperda midgut cell line through single cell RNA sequencing. Genomics 2024; 116:110898. [PMID: 39047877 DOI: 10.1016/j.ygeno.2024.110898] [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: 05/02/2024] [Revised: 06/24/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Using the 10x Genomics Chromium single-cell RNA sequencing (scRNA-seq) platform, we discovered unexpected heterogeneity in an established cell line developed from the midgut of the Fall armyworm, Spodoptera frugiperda, a major global pest. We analyzed the sequences of 18,794 cells and identified ten unique cellular clusters, including stem cells, enteroblasts, enterocytes and enteroendocrine cells, characterized by the expression of specific marker genes. Additionally, these studies addressed an important knowledge gap by investigating the expression of genes coding for respiratory and midgut membrane insecticide targets classified by the Insecticide Resistance Action Committee. Dual-fluorescence tagging method, fluorescence microscopy and fluorescence-activated cell sorting confirmed the expression of midgut cell type-specific genes. Stem cells were isolated from the heterogeneous population of SfMG-0617 cells. Our results, validated by KEGG and Gene Ontology analyses and supported by Monocle 3.0, advance the fields of midgut cellular biology and establish standards for scRNA-seq studies in non-model organisms.
Collapse
Affiliation(s)
- Surjeet Kumar Arya
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA
| | - Douglas A Harrison
- College of Arts & Science Imaging Center & Department of Biology, University of Kentucky, Lexington, KY 40546, USA
| | - Subba Reddy Palli
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA.
| |
Collapse
|
29
|
Glaser-Schmitt A, Lemoine M, Kaltenpoth M, Parsch J. Pervasive tissue-, genetic background-, and allele-specific gene expression effects in Drosophila melanogaster. PLoS Genet 2024; 20:e1011257. [PMID: 39178312 PMCID: PMC11376557 DOI: 10.1371/journal.pgen.1011257] [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: 04/14/2024] [Revised: 09/05/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
The pervasiveness of gene expression variation and its contribution to phenotypic variation and evolution is well known. This gene expression variation is context dependent, with differences in regulatory architecture often associated with intrinsic and environmental factors, and is modulated by regulatory elements that can act in cis (linked) or in trans (unlinked) relative to the genes they affect. So far, little is known about how this genetic variation affects the evolution of regulatory architecture among closely related tissues during population divergence. To address this question, we analyzed gene expression in the midgut, hindgut, and Malpighian tubule as well as microbiome composition in the two gut tissues in four Drosophila melanogaster strains and their F1 hybrids from two divergent populations: one from the derived, European range and one from the ancestral, African range. In both the transcriptome and microbiome data, we detected extensive tissue- and genetic background-specific effects, including effects of genetic background on overall tissue specificity. Tissue-specific effects were typically stronger than genetic background-specific effects, although the two gut tissues were not more similar to each other than to the Malpighian tubules. An examination of allele specific expression revealed that, while both cis and trans effects were more tissue-specific in genes expressed differentially between populations than genes with conserved expression, trans effects were more tissue-specific than cis effects. Despite there being highly variable regulatory architecture, this observation was robust across tissues and genetic backgrounds, suggesting that the expression of trans variation can be spatially fine-tuned as well as or better than cis variation during population divergence and yielding new insights into cis and trans regulatory evolution.
Collapse
Affiliation(s)
- Amanda Glaser-Schmitt
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marion Lemoine
- Department of Insect Symbiosis, Max-Planck-Institute for Chemical Ecology, Jena, Germany
| | - Martin Kaltenpoth
- Department of Insect Symbiosis, Max-Planck-Institute for Chemical Ecology, Jena, Germany
| | - John Parsch
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
30
|
Chen TY, Raduwan H, Marín-López A, Cui Y, Fikrig E. Zika virus exists in enterocytes and enteroendocrine cells of the Aedes aegypti midgut. iScience 2024; 27:110353. [PMID: 39055935 PMCID: PMC11269924 DOI: 10.1016/j.isci.2024.110353] [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: 04/10/2024] [Revised: 05/21/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The Aedes aegypti midgut is crucial for blood digestion, nutrition, reproduction, and pathogen interaction. Using single-cell RNA sequencing, we explored virus infection and transcriptomic changes at the cellular level. We identified 12 distinct cell clusters in the Ae. aegypti midgut post-Zika virus infection, including intestinal stem cells, enteroblasts, enteroendocrine cells (EE), and enterocytes (ECs). The virus was found mainly in specific subsets of ECs and EE. Infection altered transcriptional profiles related to metabolism, signaling, and immune responses. Functional studies highlighted three significantly differentially expressed genes in infected cells. Notably, silencing apolipophorin III reduced virus RNA copy number in the midgut, emphasizing the role of specific genes in viral infection. These findings enhance our understanding of mosquito midgut cell processes during Zika virus infection and suggest potential targets for vector control.
Collapse
Affiliation(s)
- Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yingjun Cui
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
31
|
Vetkama W, Tinikul R, Sobhon P, Tinikul Y. Differential expression of neuropeptide F in the digestive organs of female freshwater prawn, Macrobrachium rosenbergii, during the ovarian cycle. Cell Tissue Res 2024; 397:13-36. [PMID: 38592496 PMCID: PMC11231001 DOI: 10.1007/s00441-024-03893-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 03/19/2024] [Indexed: 04/10/2024]
Abstract
Neuropeptide F is a key hormone that controls feeding in invertebrates, including decapod crustaceans. We investigated the differential expression of Macrobrachium rosenbergii neuropeptide F (MrNPF) in the digestive organs of female prawns, M. rosenbergii, during the ovarian cycle. By using RT-qPCR, the expression of MrNPF mRNA in the esophagus (ESO), cardia (CD), and pylorus (PY) of the foregut (FG) gradually increased from stage II and peaked at stage III. In the midgut (MG), hindgut (HG), and hepatopancreas (HP), MrNPF mRNA increased from stage I, reaching a maximal level at stage II, and declined by about half at stages III and IV (P < 0.05). In the ESO, CD, and PY, strong MrNPF-immunoreactivities were seen in the epithelium, muscle, and lamina propria. Intense MrNPF-ir was found in the MG cells and the muscular layer. In the HG, MrNPF-ir was detected in the epithelium of the villi and gland regions, while MrNPF-ir was also more intense in the F-, R-, and B-cells in the HP. However, we found little colocalization between the MrNPF and PGP9.5/ChAT in digestive tissues, implying that most of the positive cells might not be neurons but could be digestive tract-associated endocrine cells that produce and secrete MrNPF to control digestive organ functions in feeding and utilizing feed. Taken together, our first findings indicated that MrNPF was differentially expressed in digestive organs in correlation with the ovarian cycle, suggesting an important link between MrNPF, the physiology of various digestive organs in feeding, and possibly ovarian maturation in female M. rosenbergii.
Collapse
Affiliation(s)
- Warinthip Vetkama
- Department of Anatomy, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi District, Bangkok 10400, Thailand
| | - Ruchanok Tinikul
- Department of Biochemistry and Center for Excellence in Protein and Enzyme Technology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi District, Bangkok 10400, Thailand
| | - Yotsawan Tinikul
- Department of Anatomy, Faculty of Science, Mahidol University, 272 Rama VI Road, Ratchathewi District, Bangkok 10400, Thailand.
| |
Collapse
|
32
|
Chen J, Nouzova M, Noriega FG, Tatar M. Gut-to-brain regulation of Drosophila aging through neuropeptide F, insulin and juvenile hormone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600832. [PMID: 38979180 PMCID: PMC11230353 DOI: 10.1101/2024.06.26.600832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Dietary restriction slows aging in many animals, while in some cases the sensory signals from diet alone are sufficient to retard or accelerate lifespan. The digestive tract is a candidate location to sense nutrients, where neuropeptides secreted by enteroendocrine cells (EEC) produce systemic signals in response to food. Here we measure how Drosophila neuropeptide F (NPF) is secreted into adult circulation by enteroendocrine cells and find that specific enteroendocrine cells differentially respond to dietary sugar and yeast. Lifespan is increased when gut NPF is genetically depleted, and this manipulation is sufficient to blunt the longevity benefit conferred by dietary restriction. Depletion of NPF receptors at insulin producing neurons of the brain also increases lifespan, consistent with observations where loss of gut NPF decreases neuronal insulin secretion. The longevity conferred by repressing gut NPF and brain NPF receptors is reversed by treating adults with a juvenile hormone (JH) analog. JH is produced by the adult corpora allata, and inhibition of the insulin receptor at this tissue decreases JH titer and extends lifespan, while this longevity is restored to wild type by treating adults with a JH analog. Overall, enteroendocrine cells of the gut modulate Drosophila aging through interorgan communication mediated by a gut-brain-corpora allata axis, and insulin produced in the brain impacts lifespan through its control of JH titer. These data suggest that we should consider how human incretins and their analogs, which are used to treat obesity and diabetes, may impact aging.
Collapse
Affiliation(s)
- Jiangtian Chen
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912 USA
| | - Marcela Nouzova
- Institute of Parasitology, Biology Centre CAS, České Budějovice, Czech Republic, 37005
| | - Fernando G. Noriega
- Department of Biological Sciences and Biomolecular Sciences Institute, Florida International University, Miami, FL, 33199 USA
- Department of Parasitology, University of South Bohemia, České Budějovice, 37005 Czech Republic
| | - Marc Tatar
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI 02912 USA
| |
Collapse
|
33
|
Wang S, Huang Y, Wang F, Han Q, Ren N, Wang X, Cui Y, Yuan Z, Xia H. A cell atlas of the adult female Aedes aegypti midgut revealed by single-cell RNA sequencing. Sci Data 2024; 11:587. [PMID: 38839790 PMCID: PMC11153528 DOI: 10.1038/s41597-024-03432-8] [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: 11/17/2023] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
Aedes aegypti is a primary vector for transmitting various arboviruses, including Yellow fever, dengue and Zika virus. The mosquito midgut is the principal organ for blood meal digestion, nutrient absorption and the initial site of arbovirus infection. Although a previous study delineated midgut's transcriptome of Ae. aegypti at the single-nucleus resolution, there still lacks an established protocol for isolating and RNA sequencing of single cells of Ae. aegypti midgut, which is required for investigating arbovirus-midgut interaction at the single-cell level. Here, we established an atlas of the midgut cells for Ae. aegypti by single-cell RNA sequencing. We annotated the cell clusters including intestinal stem cells/enteroblasts (ISC/EB), cardia cells (Cardia), enterocytes (EC, EC-like), enteroendocrine cells (EE), visceral muscle (VM), fat body cells (FBC) and hemocyte cells (HC). This study will provide a foundation for further studies of arbovirus infection in mosquito midgut at the single-cell level.
Collapse
Affiliation(s)
- Shunlong Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China
| | - Fei Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China
| | - Qian Han
- Hainan One Health Key Laboratory, Hainan University, Haikou, 570228, China
| | - Nanjie Ren
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyu Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjun Cui
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, 06520, USA.
| | - Zhiming Yuan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Han Xia
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430200, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Hubei Jiangxia Laboratory, Wuhan, 430207, China.
| |
Collapse
|
34
|
Chen J, Lin G, Ma K, Li Z, Liégeois S, Ferrandon D. A specific innate immune response silences the virulence of Pseudomonas aeruginosa in a latent infection model in the Drosophila melanogaster host. PLoS Pathog 2024; 20:e1012252. [PMID: 38833496 PMCID: PMC11178223 DOI: 10.1371/journal.ppat.1012252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/14/2024] [Accepted: 05/10/2024] [Indexed: 06/06/2024] Open
Abstract
Microbial pathogenicity often depends on the route of infection. For instance, P. aeruginosa or S. marcescens cause acute systemic infections when low numbers of bacteria are injected into D. melanogaster flies whereas flies succumb much slower to the continuous ingestion of these pathogens, even though both manage to escape from the gut compartment and reach the hemocoel. Here, we have developed a latent P. aeruginosa infection model by feeding flies on the bacteria for a short period. The bacteria stably colonize internal tissues yet hardly cause any damage since latently-infected flies live almost as long as noninfected control flies. The apparently dormant bacteria display particular characteristics in terms of bacterial colony morphology, composition of the outer cell wall, and motility. The virulence of these bacteria can however be reactivated upon wounding the host. We show that melanization but not the cellular or the systemic humoral response is the predominant host defense that establishes latency and may coerce the bacteria to a dormant state. In addition, the lasting activation of the melanization responses in latently-infected flies provides a degree of protection to the host against a secondary fungal infection. Latent infection by an ingested pathogen protects against a variety of homologous or heterologous systemic secondary infectious challenges, a situation previously described for the endosymbiotic Wolbachia bacteria, a guard against viral infections.
Collapse
Affiliation(s)
- Jing Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Guiying Lin
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Université de Strasbourg, Strasbourg, France
- Modèles Insectes de l’Immunité Innée, UPR 9022 du CNRS, Strasbourg, France
| | - Kaiyu Ma
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zi Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Samuel Liégeois
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Université de Strasbourg, Strasbourg, France
- Modèles Insectes de l’Immunité Innée, UPR 9022 du CNRS, Strasbourg, France
| | - Dominique Ferrandon
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Université de Strasbourg, Strasbourg, France
- Modèles Insectes de l’Immunité Innée, UPR 9022 du CNRS, Strasbourg, France
| |
Collapse
|
35
|
Singh A, Abhilasha KV, Acharya KR, Liu H, Nirala NK, Parthibane V, Kunduri G, Abimannan T, Tantalla J, Zhu LJ, Acharya JK, Acharya UR. A nutrient responsive lipase mediates gut-brain communication to regulate insulin secretion in Drosophila. Nat Commun 2024; 15:4410. [PMID: 38782979 PMCID: PMC11116528 DOI: 10.1038/s41467-024-48851-8] [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: 06/23/2022] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Pancreatic β cells secrete insulin in response to glucose elevation to maintain glucose homeostasis. A complex network of inter-organ communication operates to modulate insulin secretion and regulate glucose levels after a meal. Lipids obtained from diet or generated intracellularly are known to amplify glucose-stimulated insulin secretion, however, the underlying mechanisms are not completely understood. Here, we show that a Drosophila secretory lipase, Vaha (CG8093), is synthesized in the midgut and moves to the brain where it concentrates in the insulin-producing cells in a process requiring Lipid Transfer Particle, a lipoprotein originating in the fat body. In response to dietary fat, Vaha stimulates insulin-like peptide release (ILP), and Vaha deficiency results in reduced circulatory ILP and diabetic features including hyperglycemia and hyperlipidemia. Our findings suggest Vaha functions as a diacylglycerol lipase physiologically, by being a molecular link between dietary fat and lipid amplified insulin secretion in a gut-brain axis.
Collapse
Affiliation(s)
- Alka Singh
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | | | - Kathya R Acharya
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
- University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| | - Haibo Liu
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Niraj K Nirala
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Velayoudame Parthibane
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Thiruvaimozhi Abimannan
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Jacob Tantalla
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Jairaj K Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Usha R Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA.
| |
Collapse
|
36
|
Gao J, Zhang S, Deng P, Wu Z, Lemaitre B, Zhai Z, Guo Z. Dietary L-Glu sensing by enteroendocrine cells adjusts food intake via modulating gut PYY/NPF secretion. Nat Commun 2024; 15:3514. [PMID: 38664401 PMCID: PMC11045819 DOI: 10.1038/s41467-024-47465-4] [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: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.
Collapse
Affiliation(s)
- Junjun Gao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Deng
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhigang Wu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zongzhao Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, PR China.
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
37
|
Guo X, Wang C, Zhang Y, Wei R, Xi R. Cell-fate conversion of intestinal cells in adult Drosophila midgut by depleting a single transcription factor. Nat Commun 2024; 15:2656. [PMID: 38531872 DOI: 10.1038/s41467-024-46956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
The manipulation of cell identity by reprograming holds immense potential in regenerative medicine, but is often limited by the inefficient acquisition of fully functional cells. This problem can potentially be resolved by better understanding the reprogramming process using in vivo genetic models, which are currently scarce. Here we report that both enterocytes (ECs) and enteroendocrine cells (EEs) in adult Drosophila midgut show a surprising degree of cell plasticity. Depleting the transcription factor Tramtrack in the differentiated ECs can initiate Prospero-mediated cell transdifferentiation, leading to EE-like cells. On the other hand, depletion of Prospero in the differentiated EEs can lead to the loss of EE-specific transcription programs and the gain of intestinal progenitor cell identity, allowing cell cycle re-entry or differentiation into ECs. We find that intestinal progenitor cells, ECs, and EEs have a similar chromatin accessibility profile, supporting the concept that cell plasticity is enabled by pre-existing chromatin accessibility with switchable transcription programs. Further genetic analysis with this system reveals that the NuRD chromatin remodeling complex, cell lineage confliction, and age act as barriers to EC-to-EE transdifferentiation. The establishment of this genetically tractable in vivo model should facilitate mechanistic investigation of cell plasticity at the molecular and genetic level.
Collapse
Affiliation(s)
- Xingting Guo
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Chenhui Wang
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yongchao Zhang
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Ruxue Wei
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Rongwen Xi
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China.
| |
Collapse
|
38
|
Zhang G, Fu Y, Yang L, Ye F, Zhang P, Zhang S, Ma L, Li J, Wu H, Han X, Wang J, Guo G. Construction of single-cell cross-species chromatin accessibility landscapes with combinatorial-hybridization-based ATAC-seq. Dev Cell 2024; 59:793-811.e8. [PMID: 38330939 DOI: 10.1016/j.devcel.2024.01.015] [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: 06/01/2023] [Revised: 11/03/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024]
Abstract
Despite recent advances in single-cell genomics, the lack of maps for single-cell candidate cis-regulatory elements (cCREs) in non-mammal species has limited our exploration of conserved regulatory programs across vertebrates and invertebrates. Here, we developed a combinatorial-hybridization-based method for single-cell assay for transposase-accessible chromatin using sequencing (scATAC-seq) named CH-ATAC-seq, enabling the construction of single-cell accessible chromatin landscapes for zebrafish, Drosophila, and earthworms (Eisenia andrei). By integrating scATAC censuses of humans, monkeys, and mice, we systematically identified 152 distinct main cell types and around 0.8 million cell-type-specific cCREs. Our analysis provided insights into the conservation of neural, muscle, and immune lineages across species, while epithelial cells exhibited a higher organ-origin heterogeneity. Additionally, a large-scale gene regulatory network (GRN) was constructed in four vertebrates by integrating scRNA-seq censuses. Overall, our study provides a valuable resource for comparative epigenomics, identifying the evolutionary conservation and divergence of gene regulation across different species.
Collapse
Affiliation(s)
- Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Lei Yang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Peijing Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Shuang Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Lifeng Ma
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Jiaqi Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Xiaoping Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou 310058, China.
| | - Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China.
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310000, China; Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou 310058, China; Institute of Hematology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
39
|
Parasram K, Zuccato A, Shin M, Willms R, DeVeale B, Foley E, Karpowicz P. The emergence of circadian timekeeping in the intestine. Nat Commun 2024; 15:1788. [PMID: 38413599 PMCID: PMC10899604 DOI: 10.1038/s41467-024-45942-4] [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: 06/12/2023] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
The circadian clock is a molecular timekeeper, present from cyanobacteria to mammals, that coordinates internal physiology with the external environment. The clock has a 24-h period however development proceeds with its own timing, raising the question of how these interact. Using the intestine of Drosophila melanogaster as a model for organ development, we track how and when the circadian clock emerges in specific cell types. We find that the circadian clock begins abruptly in the adult intestine and gradually synchronizes to the environment after intestinal development is complete. This delayed start occurs because individual cells at earlier stages lack the complete circadian clock gene network. As the intestine develops, the circadian clock is first consolidated in intestinal stem cells with changes in Ecdysone and Hnf4 signalling influencing the transcriptional activity of Clk/cyc to drive the expression of tim, Pdp1, and vri. In the mature intestine, stem cell lineage commitment transiently disrupts clock activity in differentiating progeny, mirroring early developmental clock-less transitions. Our data show that clock function and differentiation are incompatible and provide a paradigm for studying circadian clocks in development and stem cell lineages.
Collapse
Affiliation(s)
- Kathyani Parasram
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Amy Zuccato
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Minjeong Shin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Reegan Willms
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Brian DeVeale
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Phillip Karpowicz
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, N9B 3P4, Canada.
| |
Collapse
|
40
|
Awais MM, Fei S, Xia J, Feng M, Sun J. Insights into midgut cell types and their crucial role in antiviral immunity in the lepidopteran model Bombyx mori. Front Immunol 2024; 15:1349428. [PMID: 38420120 PMCID: PMC10899340 DOI: 10.3389/fimmu.2024.1349428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
The midgut, a vital component of the digestive system in arthropods, serves as an interface between ingested food and the insect's physiology, playing a pivotal role in nutrient absorption and immune defense mechanisms. Distinct cell types, including columnar, enteroendocrine, goblet and regenerative cells, comprise the midgut in insects and contribute to its robust immune response. Enterocytes/columnar cells, the primary absorptive cells, facilitate the immune response through enzyme secretions, while regenerative cells play a crucial role in maintaining midgut integrity by continuously replenishing damaged cells and maintaining the continuity of the immune defense. The peritrophic membrane is vital to the insect's innate immunity, shielding the midgut from pathogens and abrasive food particles. Midgut juice, a mixture of digestive enzymes and antimicrobial factors, further contributes to the insect's immune defense, helping the insect to combat invading pathogens and regulate the midgut microbial community. The cutting-edge single-cell transcriptomics also unveiled previously unrecognized subpopulations within the insect midgut cells and elucidated the striking similarities between the gastrointestinal tracts of insects and higher mammals. Understanding the intricate interplay between midgut cell types provides valuable insights into insect immunity. This review provides a solid foundation for unraveling the complex roles of the midgut, not only in digestion but also in immunity. Moreover, this review will discuss the novel immune strategies led by the midgut employed by insects to combat invading pathogens, ultimately contributing to the broader understanding of insect physiology and defense mechanisms.
Collapse
Affiliation(s)
| | | | | | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
41
|
Vieira Contreras F, Auger GM, Müller L, Richter V, Huetteroth W, Seufert F, Hildebrand PW, Scholz N, Thum AS, Ljaschenko D, Blanco-Redondo B, Langenhan T. The adhesion G-protein-coupled receptor mayo/CG11318 controls midgut development in Drosophila. Cell Rep 2024; 43:113640. [PMID: 38180839 DOI: 10.1016/j.celrep.2023.113640] [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: 03/02/2023] [Revised: 11/14/2023] [Accepted: 12/16/2023] [Indexed: 01/07/2024] Open
Abstract
Adhesion G-protein-coupled receptors (aGPCRs) form a large family of cell surface molecules with versatile tasks in organ development. Many aGPCRs still await their functional and pharmacological deorphanization. Here, we characterized the orphan aGPCR CG11318/mayo of Drosophila melanogaster and found it expressed in specific regions of the gastrointestinal canal and anal plates, epithelial specializations that control ion homeostasis. Genetic removal of mayo results in tachycardia, which is caused by hyperkalemia of the larval hemolymph. The hyperkalemic effect can be mimicked by a raise in ambient potassium concentration, while normal potassium levels in mayoKO mutants can be restored by pharmacological inhibition of potassium channels. Intriguingly, hyperkalemia and tachycardia are caused non-cell autonomously through mayo-dependent control of enterocyte proliferation in the larval midgut, which is the primary function of this aGPCR. These findings characterize the ancestral aGPCR Mayo as a homeostatic regulator of gut development.
Collapse
Affiliation(s)
- Fernando Vieira Contreras
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Genevieve M Auger
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Lena Müller
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Vincent Richter
- Institute of Biology, Department of Genetics, Faculty of Life Sciences, Leipzig University, Talstraße 33, 04103 Leipzig, Germany
| | - Wolf Huetteroth
- Institute of Biology, Department of Genetics, Faculty of Life Sciences, Leipzig University, Talstraße 33, 04103 Leipzig, Germany
| | - Florian Seufert
- Institute for Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Peter W Hildebrand
- Institute for Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Andreas S Thum
- Institute of Biology, Department of Genetics, Faculty of Life Sciences, Leipzig University, Talstraße 33, 04103 Leipzig, Germany
| | - Dmitrij Ljaschenko
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Beatriz Blanco-Redondo
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany.
| | - Tobias Langenhan
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany; Institute of Biology, Faculty of Life Sciences, Leipzig University, Talstraße 33, 04103 Leipzig, Germany; Comprehensive Cancer Center Central Germany (CCCG), Germany.
| |
Collapse
|
42
|
Mancheno-Ferris A, Immarigeon C, Rivero A, Depierre D, Schickele N, Fosseprez O, Chanard N, Aughey G, Lhoumaud P, Anglade J, Southall T, Plaza S, Payre F, Cuvier O, Polesello C. Crosstalk between chromatin and Shavenbaby defines transcriptional output along the Drosophila intestinal stem cell lineage. iScience 2024; 27:108624. [PMID: 38174321 PMCID: PMC10762455 DOI: 10.1016/j.isci.2023.108624] [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: 01/26/2023] [Revised: 07/05/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
The transcription factor Shavenbaby (Svb), the only member of the OvoL family in Drosophila, controls the fate of various epithelial embryonic cells and adult stem cells. Post-translational modification of Svb produces two protein isoforms, Svb-ACT and Svb-REP, which promote adult intestinal stem cell renewal or differentiation, respectively. To define Svb mode of action, we used engineered cell lines and develop an unbiased method to identify Svb target genes across different contexts. Within a given cell type, Svb-ACT and Svb-REP antagonistically regulate the expression of a set of target genes, binding specific enhancers whose accessibility is constrained by chromatin landscape. Reciprocally, Svb-REP can influence local chromatin marks of active enhancers to help repressing target genes. Along the intestinal lineage, the set of Svb target genes progressively changes, together with chromatin accessibility. We propose that Svb-ACT-to-REP transition promotes enterocyte differentiation of intestinal stem cells through direct gene regulation and chromatin remodeling.
Collapse
Affiliation(s)
- Alexandra Mancheno-Ferris
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Control of cell shape remodeling team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Clément Immarigeon
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Control of cell shape remodeling team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Alexia Rivero
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Control of cell shape remodeling team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - David Depierre
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Naomi Schickele
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Olivier Fosseprez
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Nicolas Chanard
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Gabriel Aughey
- Imperial College London, Sir Ernst Chain Building, South Kensington Campus, London SW7 2AZ, UK
| | - Priscilla Lhoumaud
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
- Institut Jacques Monod, Université Paris Cité/CNRS, 15 rue Hélène Brion, 75205 Paris Cedex 13, France
| | - Julien Anglade
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Tony Southall
- Imperial College London, Sir Ernst Chain Building, South Kensington Campus, London SW7 2AZ, UK
| | - Serge Plaza
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Laboratoire de Recherche en Sciences Végétales, CNRS/UPS/INPT, 31320 Auzeville-Tolosane, France
| | - François Payre
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Control of cell shape remodeling team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Olivier Cuvier
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Chromatin Dynamics and Cell Proliferation team, CBI, CNRS, UPS, 31062 Toulouse, France
| | - Cédric Polesello
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
- Control of cell shape remodeling team, CBI, CNRS, UPS, 31062 Toulouse, France
| |
Collapse
|
43
|
Orchard I, Lange AB. The neuroendocrine and endocrine systems in insect - Historical perspective and overview. Mol Cell Endocrinol 2024; 580:112108. [PMID: 37956790 DOI: 10.1016/j.mce.2023.112108] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023]
Abstract
A complex cascade of events leads to the initiation and maintenance of a behavioral act in response to both internally and externally derived stimuli. These events are part of a transition of the animal into a new behavioral state, coordinated by chemicals that bias tissues and organs towards a new functional state of the animal. This form of integration is defined by the neuroendocrine (or neurosecretory) system and the endocrine system that release neurohormones or hormones, respectively. Here we describe the classical neuroendocrine and endocrine systems in insects to provide an historic perspective and overview of how neurohormones and hormones support plasticity in behavioral expression. Additionally, we describe peripheral tissues such as the midgut, epitracheal glands, and ovaries, which, whilst not necessarily being endocrine glands in the pure sense of the term, do produce and release hormones, thereby providing even more flexibility for inter-organ communication and regulation.
Collapse
Affiliation(s)
- Ian Orchard
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Rd., Mississauga, ON, L5L 1C6, Canada.
| | - Angela B Lange
- Department of Biology, University of Toronto Mississauga, 3359 Mississauga Rd., Mississauga, ON, L5L 1C6, Canada.
| |
Collapse
|
44
|
Wang Z, Yang Y, Li S, Ma W, Wang K, Soberón M, Yan S, Shen J, Francis F, Bravo A, Zhang J. JAK/STAT signaling regulated intestinal regeneration defends insect pests against pore-forming toxins produced by Bacillus thuringiensis. PLoS Pathog 2024; 20:e1011823. [PMID: 38236820 PMCID: PMC10796011 DOI: 10.1371/journal.ppat.1011823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/13/2023] [Indexed: 01/22/2024] Open
Abstract
A variety of coordinated host-cell responses are activated as defense mechanisms against pore-forming toxins (PFTs). Bacillus thuringiensis (Bt) is a worldwide used biopesticide whose efficacy and precise application methods limits its use to replace synthetic pesticides in agricultural settings. Here, we analyzed the intestinal defense mechanisms of two lepidopteran insect pests after intoxication with sublethal dose of Bt PFTs to find out potential functional genes. We show that larval intestinal epithelium was initially damaged by the PFTs and that larval survival was observed after intestinal epithelium regeneration. Further analyses showed that the intestinal regeneration caused by Cry9A protein is regulated through c-Jun NH (2) terminal kinase (JNK) and Janus tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathways. JAK/STAT signaling regulates intestinal regeneration through proliferation and differentiation of intestinal stem cells to defend three different Bt proteins including Cry9A, Cry1F or Vip3A in both insect pests, Chilo suppressalis and Spodoptera frugiperda. Consequently, a nano-biopesticide was designed to improve pesticidal efficacy based on the combination of Stat double stranded RNA (dsRNA)-nanoparticles and Bt strain. This formulation controlled insect pests with better effect suggesting its potential use to reduce the use of synthetic pesticides in agricultural settings for pest control.
Collapse
Affiliation(s)
- Zeyu Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanchao Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Sirui Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Weihua Ma
- National Key Laboratory of Crop Genetic Improvement, National Centre of Plant Gene Research, Huazhong Agricultural University, Wuhan, China
| | - Kui Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mario Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Shuo Yan
- Department of Plant Biosecurity and MOA Key Laboratory of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- Department of Plant Biosecurity and MOA Key Laboratory of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Frederic Francis
- Department of Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Alejandra Bravo
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Jie Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
45
|
Xia J, Fei S, Huang Y, Lai W, Yu Y, Liang L, Wu H, Swevers L, Sun J, Feng M. Single-nucleus sequencing of silkworm larval midgut reveals the immune escape strategy of BmNPV in the midgut during the late stage of infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 164:104043. [PMID: 38013005 DOI: 10.1016/j.ibmb.2023.104043] [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: 09/21/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
The midgut is an important barrier against microorganism invasion and proliferation, yet is the first tissue encountered when a baculovirus naturally invades the host. However, only limited knowledge is available how different midgut cell types contribute to the immune response and the clearance or promotion of viral infection. Here, single-nucleus RNA sequencing (snRNA seq) was employed to analyze the responses of various cell subpopulations in the silkworm larval midgut to B. mori nucleopolyhedrovirus (BmNPV) infection. We identified 22 distinct clusters representing enteroendocrine cells (EEs), enterocytes (ECs), intestinal stem cells (ISCs), Goblet cell-like and muscle cell types in the BmNPV-infected and uninfected silkworm larvae midgut at 72 h post infection. Further, our results revealed that the strategies for immune escape of BmNPV in the midgut at the late stage of infection include (1) inhibiting the response of antiviral pathways; (2) inhibiting the expression of antiviral host factors; (3) stimulating expression levels of genes promoting BmNPV replication. These findings suggest that the midgut, as the first line of defense against the invasion of the baculovirus, has dual characteristics of "resistance" and "tolerance". Our single-cell dataset reveals the diversity of silkworm larval midgut cells, and the transcriptome analysis provides insights into the interaction between host and virus infection at the single-cell level.
Collapse
Affiliation(s)
- Junming Xia
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Shigang Fei
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Yigui Huang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Wenxuan Lai
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Yue Yu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Lingying Liang
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Hailin Wu
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, National Centre for Scientific Research Demokritos, Institute of Biosciences and Applications, Athens, Greece.
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Min Feng
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
46
|
Qu S, Zhou X, Wang Z, Wei Y, Zhou H, Zhang X, Zhu Q, Wang Y, Yang Q, Jiang L, Ma Y, Gao Y, Kong L, Zhang L. The effects of methylphenidate and atomoxetine on Drosophila brain at single-cell resolution and potential drug repurposing for ADHD treatment. Mol Psychiatry 2024; 29:165-185. [PMID: 37957291 PMCID: PMC11078728 DOI: 10.1038/s41380-023-02314-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
The stimulant methylphenidate (MPH) and the non-stimulant atomoxetine (ATX) are frequently used for the treatment of attention-deficit/hyperactivity disorder (ADHD); however, the function of these drugs in different types of brain cells and their effects on related genes remain largely unknown. To address these questions, we built a pipeline for the simultaneous examination of the activity behavior and transcriptional responses of Drosophila melanogaster at single-cell resolution following drug treatment. We selected the Drosophila with significantly increased locomotor activities (hyperactivity-like behavior) following the administration of each drug in comparison with the control (same food as the drug-treated groups with 5% sucrose, yeast, and blue food dye solution) using EasyFlyTracker. Subsequently, single cell RNA sequencing (scRNASEQ) was used to capture the transcriptome of 82,917 cells, unsupervised clustering analysis of which yielded 28 primary cell clusters representing the major cell types in adult Drosophila brain. Indeed, both neuronal and glial cells responded to MPH and ATX. Further analysis of differentially expressed genes (DEGs) revealed distinct transcriptional changes associated with these two drugs, such as two well-studied dopamine receptor genes (Dop2R and DopEcR) were responsive to MPH but not to ATX at their optimal doses, in addition to genes involved in dopamine metabolism pathways such as Syt1, Sytalpha, Syt7, and Ih in different cell types. More importantly, MPH also suppressed the expression of genes encoding other neurotransmitter receptors and synaptic signaling molecules in many cell types, especially those for Glu and GABA, while the responsive effects of ATX were much weaker. In addition to monoaminergic neuronal transmitters, other neurotransmitters have also shown a similar pattern with respect to a stronger effect associated with MPH than with ATX. Moreover, we identified four distinct glial cell subtypes responsive to the two drugs and detected a greater number of differentially expressed genes associated with ensheathing and astrocyte-like glia. Furthermore, our study provides a rich resource of candidate target genes, supported by drug set enrichment analysis (P = 2.10E-4; hypergeometric test), for the further exploration of drug repurposing. The whole list of candidates can be found at ADHDrug ( http://adhdrug.cibr.ac.cn/ ). In conclusion, we propose a fast and cost-efficient pipeline to explore the underlying molecular mechanisms of ADHD drug treatment in Drosophila brain at single-cell resolution, which may further facilitate drug repurposing applications.
Collapse
Affiliation(s)
- Susu Qu
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
| | - Xiangyu Zhou
- Chinese Institute for Brain Research, Beijing, China
| | - Zhicheng Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Yi Wei
- Chinese Institute for Brain Research, Beijing, China
| | - Han Zhou
- Chinese Institute for Brain Research, Beijing, China
| | | | - Qingjie Zhu
- Chinese Institute for Brain Research, Beijing, China
| | - Yanmin Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Quanjun Yang
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Likun Jiang
- Department of Computer Science, Xiamen University, Xiamen, China
| | - Yuan Ma
- Chinese Institute for Brain Research, Beijing, China
| | - Yuan Gao
- Chinese Institute for Brain Research, Beijing, China
| | - Lei Kong
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
47
|
Sun C, Shao Y, Iqbal J. Insect Insights at the Single-Cell Level: Technologies and Applications. Cells 2023; 13:91. [PMID: 38201295 PMCID: PMC10777908 DOI: 10.3390/cells13010091] [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: 11/22/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Single-cell techniques are a promising way to unravel the complexity and heterogeneity of transcripts at the cellular level and to reveal the composition of different cell types and functions in a tissue or organ. In recent years, advances in single-cell RNA sequencing (scRNA-seq) have further changed our view of biological systems. The application of scRNA-seq in insects enables the comprehensive characterization of both common and rare cell types and cell states, the discovery of new cell types, and revealing how cell types relate to each other. The recent application of scRNA-seq techniques to insect tissues has led to a number of exciting discoveries. Here we provide an overview of scRNA-seq and its application in insect research, focusing on biological applications, current challenges, and future opportunities to make new discoveries with scRNA-seq in insects.
Collapse
Affiliation(s)
- Chao Sun
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Yongqi Shao
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junaid Iqbal
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
48
|
Mathes N, Comas M, Bleul R, Everaert K, Hermle T, Wiekhorst F, Knittel P, Sperling RA, Vidal X. Nitrogen-vacancy center magnetic imaging of Fe 3O 4 nanoparticles inside the gastrointestinal tract of Drosophila melanogaster. NANOSCALE ADVANCES 2023; 6:247-255. [PMID: 38125606 PMCID: PMC10729879 DOI: 10.1039/d3na00684k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/25/2023] [Indexed: 12/23/2023]
Abstract
Widefield magnetometry based on nitrogen-vacancy centers enables high spatial resolution imaging of magnetic field distributions without a need for spatial scanning. In this work, we show nitrogen-vacancy center magnetic imaging of Fe3O4 nanoparticles within the gastrointestinal tract of Drosophila melanogaster larvae. Vector magnetic field imaging based on optically detected magnetic resonance is carried out on dissected larvae intestine organs containing accumulations of externally loaded magnetic nanoparticles. The distribution of the magnetic nanoparticles within the tissue can be clearly deduced from the magnetic stray field measurements. Spatially resolved magnetic imaging requires the nitrogen-vacancy centers to be very close to the sample making the technique particularly interesting for thin tissue samples. This study is a proof of principle showing the capability of nitrogen-vacancy center magnetometry as a technique to detect magnetic nanoparticle distributions in Drosophila melanogaster larvae that can be extended to other biological systems.
Collapse
Affiliation(s)
- Niklas Mathes
- Fraunhofer Institute of Applied Solid State Physics IAF Freiburg Germany
| | - Maria Comas
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg Hugstetter Straße 55 79106 Freiburg Germany
| | - Regina Bleul
- Fraunhofer Institute for Microengineering and Microsystems IMM Carl-Zeiss-Str. 18-20 55129 Mainz Germany
| | - Katrijn Everaert
- Physikalisch-Technische Bundesanstalt Abbestraße 2-12 Berlin Germany
- Department of Solid State Sciences, Ghent University Krijgslaan 281/S1 Ghent Belgium
| | - Tobias Hermle
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg Hugstetter Straße 55 79106 Freiburg Germany
| | - Frank Wiekhorst
- Physikalisch-Technische Bundesanstalt Abbestraße 2-12 Berlin Germany
| | - Peter Knittel
- Fraunhofer Institute of Applied Solid State Physics IAF Freiburg Germany
| | - Ralph A Sperling
- Fraunhofer Institute for Microengineering and Microsystems IMM Carl-Zeiss-Str. 18-20 55129 Mainz Germany
| | - Xavier Vidal
- Fraunhofer Institute of Applied Solid State Physics IAF Freiburg Germany
- TECNALIA, Basque Research and Technology Alliance (BRTA) Derio 48160 Spain
| |
Collapse
|
49
|
Kosakamoto H, Obata F, Kuraishi J, Aikawa H, Okada R, Johnstone JN, Onuma T, Piper MDW, Miura M. Early-adult methionine restriction reduces methionine sulfoxide and extends lifespan in Drosophila. Nat Commun 2023; 14:7832. [PMID: 38052797 PMCID: PMC10698029 DOI: 10.1038/s41467-023-43550-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/10/2023] [Indexed: 12/07/2023] Open
Abstract
Methionine restriction (MetR) extends lifespan in various organisms, but its mechanistic understanding remains incomplete. Whether MetR during a specific period of adulthood increases lifespan is not known. In Drosophila, MetR is reported to extend lifespan only when amino acid levels are low. Here, by using an exome-matched holidic medium, we show that decreasing Met levels to 10% extends Drosophila lifespan with or without decreasing total amino acid levels. MetR during the first four weeks of adult life only robustly extends lifespan. MetR in young flies induces the expression of many longevity-related genes, including Methionine sulfoxide reductase A (MsrA), which reduces oxidatively-damaged Met. MsrA induction is foxo-dependent and persists for two weeks after cessation of the MetR diet. Loss of MsrA attenuates lifespan extension by early-adulthood MetR. Our study highlights the age-dependency of the organismal response to specific nutrients and suggests that nutrient restriction during a particular period of life is sufficient for healthspan extension.
Collapse
Affiliation(s)
- Hina Kosakamoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Fumiaki Obata
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan.
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| | - Junpei Kuraishi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hide Aikawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Rina Okada
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Joshua N Johnstone
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Taro Onuma
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, 650-0047, Japan
| | - Matthew D W Piper
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
50
|
Quintero M, Bangi E. Disruptions in cell fate decisions and transformed enteroendocrine cells drive intestinal tumorigenesis in Drosophila. Cell Rep 2023; 42:113370. [PMID: 37924517 PMCID: PMC10841758 DOI: 10.1016/j.celrep.2023.113370] [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: 01/31/2023] [Revised: 07/11/2023] [Accepted: 10/18/2023] [Indexed: 11/06/2023] Open
Abstract
Most epithelial tissues are maintained by stem cells that produce the different cell lineages required for proper tissue function. Constant communication between different cell types ensures precise regulation of stem cell behavior and cell fate decisions. These cell-cell interactions are often disrupted during tumorigenesis, but mechanisms by which they are co-opted to support tumor growth in different genetic contexts are poorly understood. Here, we introduce PromoterSwitch, a genetic platform we established to generate large, transformed clones derived from individual adult Drosophila intestinal stem/progenitor cells. We show that cancer-driving genetic alterations representing common colon tumor genome landscapes disrupt cell fate decisions within transformed tissue and result in the emergence of abnormal cell fates. We also show that transformed enteroendocrine cells, a differentiated, hormone-secreting cell lineage, support tumor growth by regulating intestinal stem cell proliferation through multiple genotype-dependent mechanisms, which represent potential vulnerabilities that could be exploited for therapy.
Collapse
Affiliation(s)
- Maria Quintero
- Department of Biological Science, Florida State University, Tallahassee, FL 32304, USA
| | - Erdem Bangi
- Department of Biological Science, Florida State University, Tallahassee, FL 32304, USA.
| |
Collapse
|