1
|
Singh A, van den Burgh M, Boopathy V, van Nierop Y Sanchez P, Bageritz J, Lohmann I, Domsch K. Autonomous function of Antennapedia in adult muscle precursors directly connects Hox genes to adult muscle development. Development 2025; 152:DEV204341. [PMID: 39918891 DOI: 10.1242/dev.204341] [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: 08/20/2024] [Accepted: 01/16/2025] [Indexed: 02/09/2025]
Abstract
The evolutionarily conserved Hox genes define segment identities along the anterior-posterior axis and are expressed in most cell types within each segment, performing specific functions tailored to cellular needs. It has been suggested previously that Drosophila adult flight muscles in the second thoracic segment (T2) develop without direct Hox gene input, relying instead on ectodermal signals to shape their identity. However, our research, leveraging single-cell transcriptomics of Drosophila wing discs and Hox perturbation experiments using CRISPR technology and gain-of-function assays, unveiled a more intricate regulatory landscape. We found that the Hox protein Antennapedia (Antp) is essential for adult flight muscle development, acting in two crucial ways: by regulating the cell cycle rate of adult muscle precursors (AMPs) through repression of proliferation genes, and by guiding flight muscle fate via regulation of Hedgehog (Hh) signalling during cell fate establishment. Antp, along with its co-factor Apterous (Ap), directly interacts with the patched (ptc) locus to control its expression in AMPs. These findings challenge the notion of T2 as a 'Hox-free' zone, highlighting the indispensable role of low-level Antp expression in adult muscle development.
Collapse
Affiliation(s)
- Aakriti Singh
- COS, Developmental Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Meike van den Burgh
- COS, Developmental Biology, Heidelberg University, 69120 Heidelberg, Germany
| | | | | | - Josephine Bageritz
- COS, Stem Cell Niche Heterogeneity, Heidelberg University, 69120 Heidelberg, Germany
| | - Ingrid Lohmann
- COS, Developmental Biology, Heidelberg University, 69120 Heidelberg, Germany
| | - Katrin Domsch
- COS, Developmental Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
2
|
Cruz J, Sun WY, Verbeke A, Hariharan IK. Single-cell transcriptomics of X-ray irradiated Drosophila wing discs reveals heterogeneity related to cell-cycle status and cell location. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.10.627868. [PMID: 39990483 PMCID: PMC11844406 DOI: 10.1101/2024.12.10.627868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Even seemingly homogeneous populations of cells can express phenotypic diversity in response to environmental changes. Thus, X-ray irradiation of tissues composed of diverse cell types can have complex outcomes. We have used single-cell RNA-sequencing to study the effects of X-ray radiation on the Drosophila wing imaginal disc, a relatively simple tissue composed mostly of epithelial cells. Transcriptomic clustering of cells collected from the wing disc generates clusters that are mainly grouped based on proximodistal cell location. To quantify heterogeneity of gene expression among clusters, we adapted a metric used to study market concentration, the Herfindahl-Hirschman Index. Genes involved in DNA damage repair, defense against reactive oxygen species, cell cycle progression, and apoptosis are expressed relatively uniformly. In contrast, genes encoding a subset of ligands, notably cytokines that activate the JAK/STAT pathway, some transcription factors including Ets21C, previously implicated in regeneration, and several signaling proteins are expressed more regionally. Though the radiation-responsive transcription factor p53 is expressed relatively uniformly in the wing disc, several regionally-induced genes still require p53 function, indicating that regional and radiation-induced factors combine to regulate their expression. We also examined heterogeneity within regions using a clustering approach based on cell cycle gene expression. A subpopulation of cells, characterized by high levels of tribbles expression, is amplified in irradiated discs. Remarkably, this subpopulation accounts for a considerable fraction of radiation-induced gene expression, indicating that cellular responses are non-uniform even within regions. Thus, both inter-regional and intra-regional heterogeneity are important features of tissue responses to X-ray radiation.
Collapse
Affiliation(s)
- Joyner Cruz
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Willam Y. Sun
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Alexandra Verbeke
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Iswar K. Hariharan
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| |
Collapse
|
3
|
Li Y, Liu D, Zhang S, Zhou J, Li S. Outstretched wing is controlled by intestinal enteroblasts-derived unpaired 2 cytokine signaling in Drosophila. FASEB J 2024; 38:e70227. [PMID: 39636270 DOI: 10.1096/fj.202402392r] [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: 10/08/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
The outstretched wing phenotype in Drosophila melanogaster can be induced by various genetic mutations and environmental perturbations, yet the role of gut-derived signals in coordinating wing development remains largely unexplored. In this study, we demonstrate that Upd2, secreted from the gut to the wing discs, plays a crucial role in regulating the outstretched wing phenotype. The intestinal precursor cell driver esg-Gal4 exhibits low levels of leaky expression, even in the presence of Gal80ts at room temperature (25°C). This leaky expression of TDP-43, Notch, and Yki in intestinal precursor cells leads to a held-out wing phenotype, shortened lifespan, and impaired locomotor function. Although esg-Gal4 is expressed in imaginal discs, overexpression of TDP-43, Notch, or Yki using the wing-specific driver does not result in the outstretched wing. Furthermore, our data indicate that genetic alterations associated with the spread-out wing phenotype originate in enteroblasts (EBs) during early development. RNA sequencing analysis with guts from third instar larvae revealed that the JAK-STAT pathway ligand Upd2 is among the most significantly downregulated transcripts. Notably, ectopic expression of Upd2 in EBs partially rescued the abnormal held-out wing phenotype induced by TDP-43, Notch, and Yki overexpression. Together, our findings identify gut-derived Upd2 cytokine signaling as a key mediator of the outstretched wing phenotype, providing evidence for gut-to-wing communication axis during Drosophila development.
Collapse
Affiliation(s)
- Yu Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Dongyue Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Shengliang Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Jinglan Zhou
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Shuangxi Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| |
Collapse
|
4
|
Mönch TC, Smylla TK, Brändle F, Preiss A, Nagel AC. Novel Genome-Engineered H Alleles Differentially Affect Lateral Inhibition and Cell Dichotomy Processes during Bristle Organ Development. Genes (Basel) 2024; 15:552. [PMID: 38790181 PMCID: PMC11121709 DOI: 10.3390/genes15050552] [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: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Hairless (H) encodes the major antagonist in the Notch signaling pathway, which governs cellular differentiation of various tissues in Drosophila. By binding to the Notch signal transducer Suppressor of Hairless (Su(H)), H assembles repressor complexes onto Notch target genes. Using genome engineering, three new H alleles, HFA, HLLAA and HWA were generated and a phenotypic series was established by several parameters, reflecting the residual H-Su(H) binding capacity. Occasionally, homozygous HWA flies develop to adulthood. They were compared with the likewise semi-viable HNN allele affecting H-Su(H) nuclear entry. The H homozygotes were short-lived, sterile and flightless, yet showed largely normal expression of several mitochondrial genes. Typical for H mutants, both HWA and HNN homozygous alleles displayed strong defects in wing venation and mechano-sensory bristle development. Strikingly, however, HWA displayed only a loss of bristles, whereas bristle organs of HNN flies showed a complete shaft-to-socket transformation. Apparently, the impact of HWA is restricted to lateral inhibition, whereas that of HNN also affects the respective cell type specification. Notably, reduction in Su(H) gene dosage only suppressed the HNN bristle phenotype, but amplified that of HWA. We interpret these differences as to the role of H regarding Su(H) stability and availability.
Collapse
Affiliation(s)
- Tanja C. Mönch
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany; (T.C.M.); (T.K.S.); (F.B.)
| | - Thomas K. Smylla
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany; (T.C.M.); (T.K.S.); (F.B.)
| | - Franziska Brändle
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany; (T.C.M.); (T.K.S.); (F.B.)
| | - Anette Preiss
- Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany;
| | - Anja C. Nagel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany; (T.C.M.); (T.K.S.); (F.B.)
| |
Collapse
|
5
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
6
|
Palumbo RJ, Yang Y, Feigon J, Hanes SD. Catalytic activity of the Bin3/MePCE methyltransferase domain is dispensable for 7SK snRNP function in Drosophila melanogaster. Genetics 2024; 226:iyad203. [PMID: 37982586 PMCID: PMC10763541 DOI: 10.1093/genetics/iyad203] [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: 05/31/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Methylphosphate Capping Enzyme (MePCE) monomethylates the gamma phosphate at the 5' end of the 7SK noncoding RNA, a modification thought to protect 7SK from degradation. 7SK serves as a scaffold for assembly of a snRNP complex that inhibits transcription by sequestering the positive elongation factor P-TEFb. While much is known about the biochemical activity of MePCE in vitro, little is known about its functions in vivo, or what roles-if any-there are for regions outside the conserved methyltransferase domain. Here, we investigated the role of Bin3, the Drosophila ortholog of MePCE, and its conserved functional domains in Drosophila development. We found that bin3 mutant females had strongly reduced rates of egg-laying, which was rescued by genetic reduction of P-TEFb activity, suggesting that Bin3 promotes fecundity by repressing P-TEFb. bin3 mutants also exhibited neuromuscular defects, analogous to a patient with MePCE haploinsufficiency. These defects were also rescued by genetic reduction of P-TEFb activity, suggesting that Bin3 and MePCE have conserved roles in promoting neuromuscular function by repressing P-TEFb. Unexpectedly, we found that a Bin3 catalytic mutant (Bin3Y795A) could still bind and stabilize 7SK and rescue all bin3 mutant phenotypes, indicating that Bin3 catalytic activity is dispensable for 7SK stability and snRNP function in vivo. Finally, we identified a metazoan-specific motif (MSM) outside of the methyltransferase domain and generated mutant flies lacking this motif (Bin3ΔMSM). Bin3ΔMSM mutant flies exhibited some-but not all-bin3 mutant phenotypes, suggesting that the MSM is required for a 7SK-independent, tissue-specific function of Bin3.
Collapse
Affiliation(s)
- Ryan J Palumbo
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Yuan Yang
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Juli Feigon
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Steven D Hanes
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
7
|
Peng Q, Wang Y, Xiao Y, Chang H, Luo S, Wang D, Rong YS. Drosophila Amus and Bin3 methylases functionally replace mammalian MePCE for capping and the stabilization of U6 and 7SK snRNAs. SCIENCE ADVANCES 2023; 9:eadj9359. [PMID: 38100593 PMCID: PMC10848712 DOI: 10.1126/sciadv.adj9359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023]
Abstract
U6 and 7SK snRNAs have a 5' cap, believed to be essential for their stability and maintained by mammalian MePCE or Drosophila Bin3 enzymes. Although both proteins are required for 7SK stability, loss of neither destabilizes U6, casting doubts on the function of capping U6. Here, we show that the Drosophila Amus protein, homologous to both proteins, is essential for U6 but not 7SK stability. The loss of U6 is rescued by the expression of an Amus-MePCE hybrid protein harboring the methyltransferase domain from MePCE, highlighting the conserved function of the two proteins as the U6 capping enzyme. Our investigations in human cells establish a dependence of both U6 and 7SK stability on MePCE, resolving a long-standing uncertainty. While uncovering a division of labor of Bin3/MePCE/Amus proteins, we found a "Bin3-Box" domain present only in enzymes associated with 7SK regulation. Targeted mutagenesis confirms its importance for Bin3 function, revealing a possible conserved element in 7SK but not U6 biology.
Collapse
Affiliation(s)
- Qiu Peng
- MOE Key Lab of Rare Pediatric Diseases, Hengyang College of Medicine, University of South China, Hengyang, China
| | - Yiqing Wang
- MOE Key Lab of Rare Pediatric Diseases, Hengyang College of Medicine, University of South China, Hengyang, China
| | - Ying Xiao
- MOE Key Lab of Rare Pediatric Diseases, Hengyang College of Medicine, University of South China, Hengyang, China
| | - Hua Chang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shishi Luo
- MOE Key Lab of Rare Pediatric Diseases, Hengyang College of Medicine, University of South China, Hengyang, China
| | - Danling Wang
- MOE Key Lab of Rare Pediatric Diseases, Hengyang College of Medicine, University of South China, Hengyang, China
| | - Yikang S. Rong
- MOE Key Lab of Rare Pediatric Diseases, Hengyang College of Medicine, University of South China, Hengyang, China
| |
Collapse
|
8
|
Palumbo RJ, Hanes SD. Catalytic activity of the Bin3/MEPCE methyltransferase domain is dispensable for 7SK snRNP function in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.01.543302. [PMID: 37333392 PMCID: PMC10274667 DOI: 10.1101/2023.06.01.543302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Methylphosphate Capping Enzyme (MEPCE) monomethylates the gamma phosphate at the 5' end of the 7SK noncoding RNA, a modification thought to protect 7SK from degradation. 7SK serves as a scaffold for assembly of a snRNP complex that inhibits transcription by sequestering the positive elongation factor P-TEFb. While much is known about the biochemical activity of MEPCE in vitro, little is known about its functions in vivo, or what roles- if any-there are for regions outside the conserved methyltransferase domain. Here, we investigated the role of Bin3, the Drosophila ortholog of MEPCE, and its conserved functional domains in Drosophila development. We found that bin3 mutant females had strongly reduced rates of egg-laying, which was rescued by genetic reduction of P-TEFb activity, suggesting that Bin3 promotes fecundity by repressing P-TEFb. bin3 mutants also exhibited neuromuscular defects, analogous to a patient with MEPCE haploinsufficiency. These defects were also rescued by genetic reduction of P-TEFb activity, suggesting that Bin3 and MEPCE have conserved roles in promoting neuromuscular function by repressing P-TEFb. Unexpectedly, we found that a Bin3 catalytic mutant (Bin3Y795A) could still bind and stabilize 7SK and rescue all bin3 mutant phenotypes, indicating that Bin3 catalytic activity is dispensable for 7SK stability and snRNP function in vivo. Finally, we identified a metazoan-specific motif (MSM) outside of the methyltransferase domain and generated mutant flies lacking this motif (Bin3ΔMSM). Bin3ΔMSM mutant flies exhibited some-but not all-bin3 mutant phenotypes, suggesting that the MSM is required for a 7SK-independent, tissue-specific function of Bin3.
Collapse
Affiliation(s)
- Ryan J Palumbo
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University 750 East Adams Street, 4283 Weiskotten Hall, Syracuse, New York, 13210
| | - Steven D Hanes
- Department of Biochemistry & Molecular Biology, SUNY Upstate Medical University 750 East Adams Street, 4283 Weiskotten Hall, Syracuse, New York, 13210
| |
Collapse
|
9
|
Houtman A, Gruber S, Reisert H, Amini M, Fiore C, Gonzalez P, Han V, Jazic A, Kusupholnand M, Miller M, Nam J, Wang Z, Yu Y, Dong P, Oak ASW, Sharma A, Spana EP. Characterization of the tilt (tt) phenotype in Drosophila melanogaster. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000788. [PMID: 37193546 PMCID: PMC10183093 DOI: 10.17912/micropub.biology.000788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/16/2023] [Accepted: 04/27/2023] [Indexed: 05/18/2023]
Abstract
In the early 20th century, Calvin Bridges and Thomas Morgan identified a number of spontaneous mutations that displayed visible phenotypes in adult flies and subsequent analysis of these mutations over the past century have provided fundamental insights into subdisciplines of biology such as genetics, developmental, and cell biology. One of the mutations they identified in 1915 was named tilt ( tt ) and was described by Bridges and Morgan as having two visible phenotype characteristics in the wing. The wings were "held out at a wider angle from the body" and had a break in wing vein L3. Subsequent analysis of the tilt phenotype identified another phenotype: the wings were missing a varying number of campaniform sensilla on L3. Though Bridges and Morgan provided an ink drawing of the wing posture phenotype, only the vein and campaniform sensilla loss images have been published. Here we confirm and document the tilt phenotypes that have been previously described. We also show the penetrance of these phenotypes: the vein break and the distinct outward wing posture have decreased since its discovery.
Collapse
Affiliation(s)
- Arno Houtman
- Duke Kunshan University, Kunshan, Jiangsu, China
| | - Samuel Gruber
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Hailey Reisert
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Mina Amini
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Caroline Fiore
- Department of Biology, Duke University, Durham, North Carolina, United States
| | | | - Veronica Han
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Aeva Jazic
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Mie Kusupholnand
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Max Miller
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Jiung Nam
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Ziqin Wang
- Duke Kunshan University, Kunshan, Jiangsu, China
| | - Yang Yu
- Duke Kunshan University, Kunshan, Jiangsu, China
| | - Peter Dong
- Department of Biology, Duke University, Durham, North Carolina, United States
| | - Allen S. W. Oak
- Department of Biology, Duke University, Durham, North Carolina, United States
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Arun Sharma
- Department of Biology, Duke University, Durham, North Carolina, United States
- Department of Biomedical Sciences; Board of Governors Regenerative Medicine Institute; and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Eric P Spana
- Department of Biology, Duke University, Durham, North Carolina, United States
| |
Collapse
|
10
|
Wang Y, Zhou L, Liang W, Dang Z, Wang S, Zhang Y, Zhao P, Lu Z. Cytokine receptor DOME controls wing disc development in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 148:103828. [PMID: 36002096 DOI: 10.1016/j.ibmb.2022.103828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
In multicellular organisms, the JAK/STAT signaling pathway is involved in cell proliferation, differentiation, apoptosis, and immune regulation. Through activation of the Stat92E transcription factor, JAK/STAT signaling induced proper wing development in Drosophila. Domeless (DOME) was the first identified invertebrate JAK/STAT receptor. However, the function of DOME in Bombyx mori development remains unclear, especially in wing morphogenesis. In this study, we isolated the cytokine receptor DOME gene in B. mori and evaluated its function in DOME-knockout models. We found that overexpression of DOME at the cellular level upregulated the expression of JAK/STAT pathway-related genes, promoted proliferation, and inhibited apoptosis. The results of the interference with DOME had the opposite effects with those of overexpression at the cellular level. Using CRISPR/Cas9 technology, we constructed a DOME-knockout transgenic silkworm strain (KO-DOME) and found that the wings of the pupa and moth stages were vesicle-shaped and smaller than those of the wild-type silkworm. Some KO-DOME silkworms were unable to extend their wings from the pupal case after eclosion. We detected the expression of cyclin and apoptosis-related genes in the wing disc of the moth stage and found that some cyclin genes, such as CyclinA, CyclinB, and CyclinD, were downregulated, whereas apoptotic genes, such as Caspase1, Caspase3, and Caspase8, were upregulated. We propose that DOME regulates cell proliferation and apoptosis by affecting the JAK/STAT signaling pathway, ultimately influencing the development of wing discs. Our study provides empirical evidence for the biological function of the silkworm DOME gene, which is essential for the normal development of wings.
Collapse
Affiliation(s)
- Yaping Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China.
| | - Li Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Wenjuan Liang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Zhuo Dang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Shiyuan Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China
| | - Yan Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715, PR China
| | - Ping Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715, PR China
| | - Zhongyan Lu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, 400715, PR China; Biological Science Research Center, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
11
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
12
|
Ledru M, Clark CA, Brown J, Verghese S, Ferrara S, Goodspeed A, Su TT. Differential gene expression analysis identified determinants of cell fate plasticity during radiation-induced regeneration in Drosophila. PLoS Genet 2022; 18:e1009989. [PMID: 34990447 PMCID: PMC8769364 DOI: 10.1371/journal.pgen.1009989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/19/2022] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Ionizing radiation (IR) is used to treat half of all cancer patients because of its ability to kill cells. IR, however, can induce stem cell-like properties in non-stem cancer cells, potentiating tumor regrowth and reduced therapeutic success. We identified previously a subpopulation of cells in Drosophila larval wing discs that exhibit IR-induced stem cell-like properties. These cells reside in the future wing hinge, are resistant to IR-induced apoptosis, and are capable of translocating, changing fate, and participating in regenerating the pouch that suffers more IR-induced apoptosis. We used here a combination of lineage tracing, FACS-sorting of cells that change fate, genome-wide RNAseq, and functional testing of 42 genes, to identify two key changes that are required cell-autonomously for IR-induced hinge-to-pouch fate change: (1) repression of hinge determinants Wg (Drosophila Wnt1) and conserved zinc-finger transcription factor Zfh2 and (2) upregulation of three ribosome biogenesis factors. Additional data indicate a role for Myc, a transcriptional activator of ribosome biogenesis genes, in the process. These results provide a molecular understanding of IR-induced cell fate plasticity that may be leveraged to improve radiation therapy. Ionizing radiation (IR) is used to treat half of all cancer patients because of its ability to kill cells but treatment failures are common because tumors grow back (regenerate). Here, we asked which changes in the properties of cells facilitate regeneration in Drosophila (fruit flies) after exposure to radiation. We identified six genes whose products increase or decrease the regenerative potential of cells. These results help us understand how tissues regenerate after IR damage and will aid in designing better therapies that involve radiation.
Collapse
Affiliation(s)
- Michelle Ledru
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Caitlin A. Clark
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Jeremy Brown
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Shilpi Verghese
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Sarah Ferrara
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Andrew Goodspeed
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
13
|
Yamazoe T, Nakahara Y, Katsube H, Inoue YH. Expression of Human Mutant Preproinsulins Induced Unfolded Protein Response, Gadd45 Expression, JAK-STAT Activation, and Growth Inhibition in Drosophila. Int J Mol Sci 2021; 22:12038. [PMID: 34769468 PMCID: PMC8584581 DOI: 10.3390/ijms222112038] [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: 09/27/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Mutations in the insulin gene (INS) are frequently associated with human permanent neonatal diabetes mellitus. However, the mechanisms underlying the onset of this genetic disease is not sufficiently decoded. We induced expression of two types of human mutant INSs in Drosophila using its ectopic expression system and investigated the resultant responses in development. Expression of the wild-type preproinsulin in the insulin-producing cells (IPCs) throughout the larval stage led to a stimulation of the overall and wing growth. However, ectopic expression of human mutant preproinsulins, hINSC96Y and hINSLB15YB16delinsH, neither of which secreted from the β-cells, could not stimulate the Drosophila growth. Furthermore, neither of the mutant polypeptides induced caspase activation leading to apoptosis. Instead, they induced expression of several markers indicating the activation of unfolded protein response, such as ER stress-dependent Xbp1 mRNA splicing and ER chaperone induction. We newly found that the mutant polypeptides induced the expression of Growth arrest and DNA-damage-inducible 45 (Gadd45) in imaginal disc cells. ER stress induced by hINSC96Y also activated the JAK-STAT signaling, involved in inflammatory responses. Collectively, we speculate that the diabetes-like growth defects appeared as a consequence of the human mutant preproinsulin expression was involved in dysfunction of the IPCs, rather than apoptosis.
Collapse
Affiliation(s)
| | | | | | - Yoshihiro H. Inoue
- Department of Insect Biomedical Research, Kyoto Institute of Technology, Matsugasaki, Kyoto 606-0962, Sakyo, Japan; (T.Y.); (Y.N.); (H.K.)
| |
Collapse
|
14
|
Takemura M, Lu YS, Nakato E, Nakato H. Endogenous epitope tagging of a JAK/STAT ligand Unpaired1 in Drosophila. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 34651105 PMCID: PMC8506834 DOI: 10.17912/micropub.biology.000387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/03/2022]
Abstract
Unpaired1 (Upd1) is a ligand of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in Drosophila. In this study, using the CRISPR/Cas9 technique, we generate a transgenic fly strain in which a hemagglutinin (HA) epitope tag sequence is inserted into the endogenous locus of the upd1 gene. Anti-HA antibody staining confirms that the distribution of the epitope-tagged Upd1::HA in various tissues is consistent with upd1 expression patterns revealed by previous studies. This transgenic fly strain will be useful in studying the expression, localization, and association partners of Upd1, and thus will contribute to understanding how activation of the JAK/STAT pathway is regulated.
Collapse
Affiliation(s)
- Masahiko Takemura
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis USA
| | - Yi-Si Lu
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis USA
| | - Eriko Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis USA
| | - Hiroshi Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis USA
| |
Collapse
|
15
|
Zhang XJ, Li DD, Xu GF, Chen YQ, Zheng SC. Signal transducer and activator of transcription is involved in the expression regulation of ecdysteroid-induced insulin-like growth factor-like peptide in the pupal wing disc of silkworm, Bombyx mori. INSECT SCIENCE 2020; 27:1186-1197. [PMID: 31724818 DOI: 10.1111/1744-7917.12736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 06/10/2023]
Abstract
In insects, 20-hydroxyecdysone (20E) and insulin-like growth factor-like peptides (IGFLPs) regulate the development of imaginal discs. However, how IGFLPs are up-regulated to impact the development of the pupal wing disc is still unclear. In this study, we investigated the expression regulation of IGFLP in the pupal wing disc of silkworm, Bombyx mori. We confirmed that B. mori IGFLP (BmIGFLP) was mainly expressed in the pupal wing disc and the expression of BmIGFLP could be significantly induced by 20E. Bioinformatics analysis of BmIGFLP promoter sequence revealed three cis-regulation elements (CREs) of signal transducer and activator of transcription (STAT), which is a key component in the Janus-activated kinase / STAT pathway. Luciferase activity assays showed that two CREs enhanced the transcriptional activity of BmIGFLP. Electrophoretic mobility shift and chromatin immunoprecipitation assays demonstrated that BmSTAT proteins in the nuclear extracts of B. mori pupal wing discs and BmN cells could only bind to the STAT CRE3, indicating that STAT CRE3 activated by BmSTAT enhances BmIGFLP expression at pupal stages. Although 20E could not enhance the expression of BmSTAT, 20E enhanced the nucleus translocation of BmSTAT to bind with the STAT CRE3 in the BmIGFLP promoter. The increase of transcriptional activity of the STAT CRE3 by overexpression of BmSTAT and addition of 20E in BmN cells confirmed this result. Taken together, all data indicate that BmSTAT is one of the transcription factors activating 20E-induced BmIGFLP expression in the pupal wing disc.
Collapse
Affiliation(s)
- Xiao-Juan Zhang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Dong-Dong Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Guan-Feng Xu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Ya-Qing Chen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Si-Chun Zheng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangzhou Key Laboratory of Insect Development Regulation and Applied Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
16
|
Powers N, Srivastava A. JAK/STAT signaling is involved in air sac primordium development of Drosophila melanogaster. FEBS Lett 2019; 593:658-669. [PMID: 30854626 DOI: 10.1002/1873-3468.13355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/17/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022]
Abstract
The dorsal thoracic air sacs in fruit flies (Drosophila melanogaster) are functionally and developmentally comparable to human lungs. The progenitors of these structures, air sac primordia (ASPs), invasively propagate into wing imaginal disks, employing mechanisms similar to those that promote metastasis in malignant tumors. We investigated whether Janus kinase/signal transducer and activator of transcription JAK/STAT signaling plays a role in the directed morphogenesis of ASPs. We find that JAK/STAT signaling occurs in ASP tip cells and misexpression of core components in the JAK/STAT signaling cascade significantly impedes ASP development. We further identify Upd2 as an activating ligand for JAK/STAT activity in the ASP. Together, these data constitute a considerable step forward in understanding the role of JAK/STAT signaling in ASPs and similar structures in mammalian models.
Collapse
Affiliation(s)
- Nathan Powers
- Department of Biology and Biotechnology Center, Western Kentucky University, Bowling Green, KY, USA
| | - Ajay Srivastava
- Department of Biology and Biotechnology Center, Western Kentucky University, Bowling Green, KY, USA
| |
Collapse
|
17
|
Narbonne-Reveau K, Maurange C. Developmental regulation of regenerative potential in Drosophila by ecdysone through a bistable loop of ZBTB transcription factors. PLoS Biol 2019; 17:e3000149. [PMID: 30742616 PMCID: PMC6386533 DOI: 10.1371/journal.pbio.3000149] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/22/2019] [Accepted: 01/30/2019] [Indexed: 01/08/2023] Open
Abstract
In many organisms, the regenerative capacity of tissues progressively decreases as development progresses. However, the developmental mechanisms that restrict regenerative potential remain unclear. In Drosophila, wing imaginal discs become unable to regenerate upon damage during the third larval stage (L3). Here, we show that production of ecdysone after larvae reach their critical weight (CW) terminates the window of regenerative potential by acting on a bistable loop composed of two antagonistic Broad-complex/Tramtrack/Bric-à-brac Zinc-finger (ZBTB) genes: chinmo and broad (br). Around mid L3, ecdysone signaling silences chinmo and activates br to switch wing epithelial progenitors from a default self-renewing to a differentiation-prone state. Before mid L3, Chinmo promotes a strong regenerative response upon tissue damage. After mid L3, Br installs a nonpermissive state that represses regeneration. Transient down-regulation of ecdysone signaling or Br in late L3 larvae enhances chinmo expression in damaged cells that regain the capacity to regenerate. This work unveils a mechanism that ties the self-renewing and regenerative potential of epithelial progenitors to developmental progression. This study finds that the loss of regeneration potential in Drosophila wing imaginal discs is induced by the production of the steroid hormone ecdysone after the larva reaches its critical weight. Manipulating ecdysone signaling or the downstream transcription factors can uncouple regenerative properties from developmental progression. While some organisms exhibit remarkable regenerative abilities throughout their life, many animals, including mammals, present limited regenerative potential that progressively decreases during development. Understanding the mechanisms underlying this progressive loss is important to devise therapeutic approaches aiming at facilitating the regeneration of a damaged tissue throughout life. The fruitfly Drosophila is a powerful model organism to address such questions. Indeed, while tissues, such as imaginal discs, can fully regenerate if damaged during early development, they fail to do so upon damages during late development. We show here that restriction of regenerative potential occurring during midlarval stages is due to the production of a steroid hormone, named ecdysone. By genetically manipulating ecdysone signaling, we can uncouple regenerative abilities from developmental progression. In particular, we show that ecdysone signaling triggers a switch in the sequential expression of two transcription factors, Chinmo and Broad, that positively and negatively regulate the competence for imaginal disc regeneration, respectively. Our work therefore identifies a key developmental signal that restricts regenerative potential in insects and opens new perspectives on elucidating how regeneration-permissive transcriptional programs are locked as development progresses.
Collapse
Affiliation(s)
| | - Cédric Maurange
- Aix Marseille Université, CNRS, IBDM, UMR 7288, Marseille, France
- * E-mail:
| |
Collapse
|
18
|
Herrera SC, Bach EA. JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates. Development 2019; 146:dev167643. [PMID: 30696713 PMCID: PMC6361132 DOI: 10.1242/dev.167643] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
The JAK/STAT pathway is a conserved metazoan signaling system that transduces cues from extracellular cytokines into transcriptional changes in the nucleus. JAK/STAT signaling is best known for its roles in immunity. However, recent work has demonstrated that it also regulates critical homeostatic processes in germline and somatic stem cells, as well as regenerative processes in several tissues, including the gonad, intestine and appendages. Here, we provide an overview of JAK/STAT signaling in stem cells and regeneration, focusing on Drosophila and highlighting JAK/STAT pathway functions in proliferation, survival and cell competition that are conserved between Drosophila and vertebrates.
Collapse
Affiliation(s)
- Salvador C Herrera
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Erika A Bach
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
19
|
Sui L, Alt S, Weigert M, Dye N, Eaton S, Jug F, Myers EW, Jülicher F, Salbreux G, Dahmann C. Differential lateral and basal tension drive folding of Drosophila wing discs through two distinct mechanisms. Nat Commun 2018; 9:4620. [PMID: 30397306 PMCID: PMC6218478 DOI: 10.1038/s41467-018-06497-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 09/05/2018] [Indexed: 12/26/2022] Open
Abstract
Epithelial folding transforms simple sheets of cells into complex three-dimensional tissues and organs during animal development. Epithelial folding has mainly been attributed to mechanical forces generated by an apically localized actomyosin network, however, contributions of forces generated at basal and lateral cell surfaces remain largely unknown. Here we show that a local decrease of basal tension and an increased lateral tension, but not apical constriction, drive the formation of two neighboring folds in developing Drosophila wing imaginal discs. Spatially defined reduction of extracellular matrix density results in local decrease of basal tension in the first fold; fluctuations in F-actin lead to increased lateral tension in the second fold. Simulations using a 3D vertex model show that the two distinct mechanisms can drive epithelial folding. Our combination of lateral and basal tension measurements with a mechanical tissue model reveals how simple modulations of surface and edge tension drive complex three-dimensional morphological changes.
Collapse
Affiliation(s)
- Liyuan Sui
- Institute of Genetics, Technische Universität Dresden, 01062, Dresden, Germany
| | - Silvanus Alt
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187, Dresden, Germany
- The Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Martin Weigert
- Center for Systems Biology Dresden (CSBD), Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Natalie Dye
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Suzanne Eaton
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Biotechnologisches Zentrum, Technische Universität Dresden, Tatzberg 47/49, 01309, Dresden, Germany
| | - Florian Jug
- Center for Systems Biology Dresden (CSBD), Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Eugene W Myers
- Center for Systems Biology Dresden (CSBD), Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Department of Computer Science, Technische Universität Dresden, 01062, Dresden, Germany
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187, Dresden, Germany
- Center for Systems Biology Dresden (CSBD), Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Guillaume Salbreux
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187, Dresden, Germany.
- The Francis Crick Institute, 1 Midland Road, NW1 1AT, London, UK.
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062, Dresden, Germany.
| |
Collapse
|
20
|
Worley MI, Alexander LA, Hariharan IK. CtBP impedes JNK- and Upd/STAT-driven cell fate misspecifications in regenerating Drosophila imaginal discs. eLife 2018; 7:30391. [PMID: 29372681 PMCID: PMC5823544 DOI: 10.7554/elife.30391] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/19/2018] [Indexed: 12/27/2022] Open
Abstract
Regeneration following tissue damage often necessitates a mechanism for cellular re-programming, so that surviving cells can give rise to all cell types originally found in the damaged tissue. This process, if unchecked, can also generate cell types that are inappropriate for a given location. We conducted a screen for genes that negatively regulate the frequency of notum-to-wing transformations following genetic ablation and regeneration of the wing pouch, from which we identified mutations in the transcriptional co-repressor C-terminal Binding Protein (CtBP). When CtBP function is reduced, ablation of the pouch can activate the JNK/AP-1 and JAK/STAT pathways in the notum to destabilize cell fates. Ectopic expression of Wingless and Dilp8 precede the formation of the ectopic pouch, which is subsequently generated by recruitment of both anterior and posterior cells near the compartment boundary. Thus, CtBP stabilizes cell fates following damage by opposing the destabilizing effects of the JNK/AP-1 and JAK/STAT pathways.
Collapse
Affiliation(s)
- Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Larissa A Alexander
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
21
|
JAK/STAT controls organ size and fate specification by regulating morphogen production and signalling. Nat Commun 2017; 8:13815. [PMID: 28045022 PMCID: PMC5216089 DOI: 10.1038/ncomms13815] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/02/2017] [Indexed: 01/20/2023] Open
Abstract
A stable pool of morphogen-producing cells is critical for the development of any organ or tissue. Here we present evidence that JAK/STAT signalling in the Drosophila wing promotes the cycling and survival of Hedgehog-producing cells, thereby allowing the stable localization of the nearby BMP/Dpp-organizing centre in the developing wing appendage. We identify the inhibitor of apoptosis dIAP1 and Cyclin A as two critical genes regulated by JAK/STAT and contributing to the growth of the Hedgehog-expressing cell population. We also unravel an early role of JAK/STAT in guaranteeing Wingless-mediated appendage specification, and a later one in restricting the Dpp-organizing activity to the appendage itself. These results unveil a fundamental role of the conserved JAK/STAT pathway in limb specification and growth by regulating morphogen production and signalling, and a function of pro-survival cues and mitogenic signals in the regulation of the pool of morphogen-producing cells in a developing organ.
Collapse
|
22
|
Pflugfelder G, Eichinger F, Shen J. T-Box Genes in Drosophila Limb Development. Curr Top Dev Biol 2017; 122:313-354. [DOI: 10.1016/bs.ctdb.2016.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Tamori Y, Suzuki E, Deng WM. Epithelial Tumors Originate in Tumor Hotspots, a Tissue-Intrinsic Microenvironment. PLoS Biol 2016; 14:e1002537. [PMID: 27584724 PMCID: PMC5008749 DOI: 10.1371/journal.pbio.1002537] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/27/2016] [Indexed: 12/28/2022] Open
Abstract
Malignant tumors are caused by uncontrolled proliferation of transformed mutant cells that have lost the ability to maintain tissue integrity. Although a number of causative genetic backgrounds for tumor development have been discovered, the initial steps mutant cells take to escape tissue integrity and trigger tumorigenesis remain elusive. Here, we show through analysis of conserved neoplastic tumor-suppressor genes (nTSGs) in Drosophila wing imaginal disc epithelia that tumor initiation depends on tissue-intrinsic local cytoarchitectures, causing tumors to consistently originate in a specific region of the tissue. In this “tumor hotspot” where cells constitute a network of robust structures on their basal side, nTSG-deficient cells delaminate from the apical side of the epithelium and begin tumorigenic overgrowth by exploiting endogenous Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling activity. Conversely, in other regions, the “tumor coldspot” nTSG-deficient cells are extruded toward the basal side and undergo apoptosis. When the direction of delamination is reversed through suppression of RhoGEF2, an activator of the Rho family small GTPases, and JAK/STAT is activated ectopically in these coldspot nTSG-deficient cells, tumorigenesis is induced. These data indicate that two independent processes, apical delamination and JAK/STAT activation, are concurrently required for the initiation of nTSG-deficient-induced tumorigenesis. Given the conservation of the epithelial cytoarchitecture, tumorigenesis may be generally initiated from tumor hotspots by a similar mechanism. A genetic study in Drosophila reveals a mechanism of tumorigenesis by which pro-tumor cells initiate dysplastic tumor growth at specific "hotspot" locations in epithelial tissues. Transformed mutant cells (pro-tumor cells) can evolve through a multistep process in which they become tumorigenic and invasive. Many genes that are involved in the different steps towards cancer development have been identified; however, how certain mutant cells destroy normal tissue organization and undergo uncontrolled proliferation during the initial stages of this process remains largely unclear. Using the epithelial tissue of the wing imaginal discs of the fruit fly (Drosophila melanogaster) larvae as a model system, we have analyzed these initial stages of inducing tumors by depletion of a neoplastic tumor suppressor gene (nTSG). We discovered that these tumors always originate from specific regions of the epithelial tissue of the wing disc. We show that in other regions that we dubbed “tumor coldspots” and that lack specific cellular structures, pro-tumor cells are eliminated from the epithelial tissue by the surrounding cells. However, in “tumor hotspots,” cells constitute specific structures in their basal side, and we found that pro-tumor cells successfully avoid potential elimination and deviate from the apical side of the tissue, initiating tumorous overgrowth. Our findings reveal the molecular and cellular mechanisms underlying the initial steps of tumorigenesis at tumor hotspots in the Drosophila imaginal wing discs.
Collapse
Affiliation(s)
- Yoichiro Tamori
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
- Structural Biology Center, National Institute of Genetics and Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 1111 Yata, Mishima, Japan
- * E-mail: (YT); (WMD)
| | - Emiko Suzuki
- Structural Biology Center, National Institute of Genetics and Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 1111 Yata, Mishima, Japan
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
- * E-mail: (YT); (WMD)
| |
Collapse
|
24
|
Wang D, Li L, Lu J, Liu S, Shen J. Complementary expression of optomotor-blind and the Iroquois complex promotes fold formation to separate wing notum and hinge territories. Dev Biol 2016; 416:225-234. [DOI: 10.1016/j.ydbio.2016.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/13/2016] [Accepted: 05/16/2016] [Indexed: 01/05/2023]
|
25
|
Štorgel N, Krajnc M, Mrak P, Štrus J, Ziherl P. Quantitative Morphology of Epithelial Folds. Biophys J 2016; 110:269-77. [PMID: 26745429 PMCID: PMC4825108 DOI: 10.1016/j.bpj.2015.11.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/08/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
The shape of spatially modulated epithelial morphologies such as villi and crypts is usually associated with the epithelium-stroma area mismatch leading to buckling. We propose an alternative mechanical model based on intraepithelial stresses generated by differential tensions of apical, lateral, and basal sides of cells as well as on the elasticity of the basement membrane. We use it to theoretically study longitudinal folds in simple epithelia and we identify four types of corrugated morphologies: compact, invaginated, evaginated, and wavy. The obtained tissue contours and thickness profiles are compared to epithelial folds observed in invertebrates and vertebrates, and for most samples, the agreement is within the estimated experimental error. Our model establishes the groove-crest modulation of tissue thickness as a morphometric parameter that can, together with the curvature profile, be used to estimate the relative differential apicobasal tension in the epithelium.
Collapse
Affiliation(s)
- Nick Štorgel
- Jožef Stefan Institute, Ljubljana, Slovenia; Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | | | - Polona Mrak
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jasna Štrus
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Primož Ziherl
- Jožef Stefan Institute, Ljubljana, Slovenia; Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia; Erwin Schrödinger International Institute for Mathematical Physics, University of Vienna, Vienna, Austria
| |
Collapse
|
26
|
La Fortezza M, Schenk M, Cosolo A, Kolybaba A, Grass I, Classen AK. JAK/STAT signalling mediates cell survival in response to tissue stress. Development 2016; 143:2907-19. [DOI: 10.1242/dev.132340] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Tissue homeostasis relies on the ability of tissues to respond to stress. Tissue regeneration and tumour models in Drosophila have shown that JNK is a prominent stress-response pathway promoting injury-induced apoptosis and compensatory proliferation. A central question remaining unanswered is how both responses are balanced by activation of a single pathway. JAK/STAT signalling, a potential JNK target, is implicated in promoting compensatory proliferation. While we observe JAK/STAT activation in imaginal discs upon damage, our data demonstrates that JAK/STAT and its downstream effector Zfh2 promote survival of JNK-signalling cells instead. The JNK component fos and the pro-apoptotic gene hid are regulated in a JAK/STAT-dependent manner. This molecular pathway restrains JNK-induced apoptosis and spatial propagation of JNK-signalling, thereby limiting the extent of tissue damage, as well as facilitating systemic and proliferative responses to injury. We find that the pro-survival function of JAK/STAT also drives tumour growth under conditions of chronic stress. Our study defines JAK/STAT function in tissue stress and illustrates how crosstalk between conserved signalling pathways establishes an intricate equilibrium between proliferation, apoptosis and survival to restore tissue homeostasis.
Collapse
Affiliation(s)
- Marco La Fortezza
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Madlin Schenk
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Andrea Cosolo
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Addie Kolybaba
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Isabelle Grass
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Anne-Kathrin Classen
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
27
|
Tate AT, Graham AL. Dynamic Patterns of Parasitism and Immunity across Host Development Influence Optimal Strategies of Resource Allocation. Am Nat 2015; 186:495-512. [DOI: 10.1086/682705] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
28
|
Pleiotropy of the Drosophila JAK pathway cytokine Unpaired 3 in development and aging. Dev Biol 2014; 395:218-31. [PMID: 25245869 DOI: 10.1016/j.ydbio.2014.09.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 12/22/2022]
Abstract
The Janus kinase (JAK) pathway is an essential, highly re-utilized developmental signaling cascade found in most metazoans. In vertebrates, the JAK intracellular cascade mediates signaling by dozens of cytokines and growth factors. In Drosophila, the Unpaired (Upd) family, encoded by three tandemly duplicated genes, is the only class of ligands associated with JAK stimulation. Unpaired has a central role in activation of JAK for most pathway functions, while Unpaired 2 regulates body size through insulin signaling. We show here that the third member of the family, unpaired 3 (upd3), overlaps upd in expression in some tissues and is essential for a subset of JAK-mediated developmental functions. First, consistent with the known requirements of JAK signaling in gametogenesis, we find that mutants of upd3 show an age-dependent impairment of fertility in both sexes. In oogenesis, graded JAK activity stimulated by Upd specifies the fates of the somatic follicle cells. As upd3 mutant females age, defects arise that can be attributed to perturbations of the terminal follicle cells, which require the highest levels of JAK activation. Therefore, in oogenesis, the activities of Upd and Upd3 both appear to quantitatively contribute to specification of those follicle cell fates. Furthermore, the sensitization of upd3 mutants to age-related decline in fertility can be used to investigate reproductive senescence. Second, loss of Upd3 during imaginal development results in defects of adult structures, including reduced eye size and abnormal wing and haltere posture. The outstretched wing and small eye phenotypes resemble classical alleles referred to as outstretched (os) mutations that have been previously ascribed to upd. However, we show that os alleles affect expression of both upd and upd3 and map to untranscribed regions, suggesting that they disrupt regulatory elements shared by both genes. Thus the upd region serves as a genetically tractable model for coordinate regulation of tandemly duplicated gene families that are commonly found in higher eukaryotes.
Collapse
|
29
|
Hombría JCG, Sotillos S. JAK-STAT pathway in Drosophila morphogenesis: From organ selector to cell behavior regulator. JAKSTAT 2013; 2:e26089. [PMID: 24069568 PMCID: PMC3772120 DOI: 10.4161/jkst.26089] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 11/19/2022] Open
Abstract
One of the main contributions of Drosophila to the JAK-STAT field is the study of morphogenesis. JAK-STAT signaling controls the formation of many different structures through surprisingly different morphogenetic behaviors that include induction of cell rearrangements, invagination, folding of tissues, modulation of cell shape, and migration. This variability may be explained by the many transcription factors and signaling molecules STAT regulates at early stages of development. But is STAT just acting as an upstream inducer of morphogenesis or does it have a more direct role in controlling cell behaviors? Here we review what is known about how the canonical phosphorylation of STAT contributes to shaping the embryonic and imaginal structures.
Collapse
|
30
|
Zeidler MP, Bausek N. The Drosophila JAK-STAT pathway. JAKSTAT 2013; 2:e25353. [PMID: 24069564 PMCID: PMC3772116 DOI: 10.4161/jkst.25353] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 05/23/2013] [Indexed: 02/03/2023] Open
Abstract
The conservation of signaling cascades between humans and Drosophila, over more than 500 million years of evolutionary time, means that the genetic tractability of the fly can be used to its full advantage to understand the functional requirements for JAK-STAT pathway signaling across species. Here we review the background to how the pathway was first identified and the first characterization of JAK-STAT pathway phenotypes in the Drosophila system, highlighting the molecular, functional, and disease-related conservation of the pathway.
Collapse
Affiliation(s)
- Martin P Zeidler
- MRC Centre for Development and Biomedical Genetics and the Department of Biomedical Science; The University of Sheffield; Sheffield, UK
| | | |
Collapse
|