1
|
Liang SJ, Wang K, Mao DB, Xie LW, Zhu DJ. Inhibition of the Wnt/β‑catenin signaling pathway and SOX9 by XAV939 did not alleviate inflammation in a dextran sulfate sodium‑induced ulcerative colitis model. Exp Ther Med 2025; 29:24. [PMID: 39650775 PMCID: PMC11619566 DOI: 10.3892/etm.2024.12774] [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: 06/26/2024] [Accepted: 09/12/2024] [Indexed: 12/11/2024] Open
Abstract
The Wnt/β-catenin signaling pathway has been reported to be hyperactivated during the pathogenesis of ulcerative colitis (UC). The present study aimed to explore the therapeutic efficacy of the Wnt/β-catenin signaling inhibitor XAV939 in mitigating UC symptoms. Utilizing a dextran sulfate sodium (DSS)-induced UC mouse model, the present study aimed to evaluate the impact of XAV939 on intestinal morphology through hematoxylin and eosin staining and to measure the expression levels of critical proteins in the Wnt/β-catenin signaling cascade. XAV939 did not exert a significant influence on the morphological features and inflammatory status of the intestinal epithelium. However, XAV939 was found to effectively suppress the Wnt/β-catenin signaling pathway and its downstream target SOX9. This suppression implied a reduction in the differentiation of intestinal stem cells into secretory cell progenitor cells. Additionally, XAV939 was ineffective at reversing the DSS-induced decrease in expression levels of Villin and peroxisome proliferator-activated receptor γ, which suggested that it did not facilitate the differentiation of intestinal absorptive cells. The present findings indicated that the Wnt/β-catenin signaling pathway may not be the predominant mechanism in the pathogenesis of DSS-induced UC.
Collapse
Affiliation(s)
- Shao-Jie Liang
- Maternal and Children's Health Research Institute, Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, Guangdong 528300, P.R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Kun Wang
- Maternal and Children's Health Research Institute, Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, Guangdong 528300, P.R. China
| | - Da-Bin Mao
- Maternal and Children's Health Research Institute, Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, Guangdong 528300, P.R. China
| | - Li-Wei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, Guangdong 510075, P.R. China
| | - Da-Jian Zhu
- Maternal and Children's Health Research Institute, Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, Guangdong 528300, P.R. China
| |
Collapse
|
2
|
Vieira CS, Bisogno S, Salvemini M, Loza Telleria E, Volf P. Azadirachtin disrupts ecdysone signaling and alters sand fly immunity. Parasit Vectors 2024; 17:526. [PMID: 39707409 DOI: 10.1186/s13071-024-06589-8] [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: 09/03/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Leishmaniasis is a group of neglected vector-borne diseases transmitted by phlebotomine sand flies. Leishmania parasites must overcome various defenses in the sand fly midgut, including the insects's immune response. Insect immunity is regulated by the ecdysone hormone, which binds to its nuclear receptor (EcR) and activates the transcription of genes involved in insect immunity. However, the role of ecdysone in sand fly immunity has never been studied. Phlebotomus perniciosus is a natural vector of Leishmania infantum; here, we manipulated its neuroendocrine system using azadirachtin (Aza), a natural compound known to affect ecdysone synthesis. METHODS Phlebotomus perniciosus larvae and adult females were fed on food containing either Aza alone or Aza plus ecdysone, and the effects on mortality and ecdysis were evaluated. Genes related to ecdysone signaling and immunity were identified in P. perniciosus, and the expression of antimicrobial peptides (AMPs), EcR, the ecdysone-induced genes Eip74EF and Eip75B, and the transcription factor serpent were analyzed using quantitative polymerase chain reaction (PCR). RESULTS Aza treatment inhibited molting of first-instar (L1) larvae to L2, with only 10% of larvae molting compared to 95% in the control group. Serpent and Eip74EF, attacin, defensin 1, and defensin 2 genes were downregulated by Aza treatment in larvae. Similarly, Aza-treated adult females also presented suppression of ecdysone signaling-related genes and the AMPs attacin and defensin 2. Notably, all gene repression caused by Aza was reversed by adding ecdysone concomitantly with Aza to the larval or female food, indicating that these genes are effective markers for ecdysone repression. CONCLUSIONS These results highlight the critical role of ecdysone in regulating the development and immunity of P. perniciosus, which potentially could interfere with Leishmania infection.
Collapse
Affiliation(s)
- Cecilia Stahl Vieira
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Sara Bisogno
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marco Salvemini
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Erich Loza Telleria
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
3
|
Landis GN, Baybutt B, Das S, Fan Y, Olsen K, Yan K, Tower J. Mifepristone and rapamycin have non-additive benefits for life span in mated female Drosophila. Fly (Austin) 2024; 18:2419151. [PMID: 39440794 PMCID: PMC11514543 DOI: 10.1080/19336934.2024.2419151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
The drugs mifepristone and rapamycin were compared for their relative ability to increase the life span of mated female Drosophila melanogaster. Titration of rapamycin indicated an optimal concentration of approximately 50 μM, which increased median life span here by average +81%. Meta-analysis of previous mifepristone titrations indicated an optimal concentration of approximately 466 μM, which increased median life span here by average +114%. Combining mifepristone with various concentrations of rapamycin did not produce further increases in life span, and instead reduced life span relative to either drug alone. Assay of maximum midgut diameter indicated that rapamycin was equally efficacious as mifepristone in reducing mating-induced midgut hypertrophy. The mito-QC mitophagy reporter is a previously described green fluorescent protein (GFP)-mCherry fusion protein targeted to the outer mitochondrial membrane. Inhibition of GFP fluorescence by the acidic environment of the autophagolysosome yields an increased red/green fluorescence ratio indicative of increased mitophagy. Creation of a multi-copy mito-QC reporter strain facilitated assay in live adult flies, as well as in dissected midgut tissue. Mifepristone was equally efficacious as rapamycin in activating the mito-QC mitophagy reporter in the adult female fat-body and midgut. The data suggest that mifepristone and rapamycin act through a common pathway to increase mated female Drosophila life span, and implicate increased mitophagy and decreased midgut hypertrophy in that pathway.
Collapse
Affiliation(s)
- Gary N. Landis
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Britta Baybutt
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Shoham Das
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Yijie Fan
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Kate Olsen
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Karissa Yan
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
4
|
Wang X, Guo Y, Lin P, Yu M, Song S, Xu W, Kong D, Wang Y, Zhang Y, Lu F, Xie Q, Ma X. Nuclear receptor E75/NR1D2 promotes tumor malignant transformation by integrating Hippo and Notch pathways. EMBO J 2024; 43:6336-6363. [PMID: 39516282 DOI: 10.1038/s44318-024-00290-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Hormone therapy resistance and the ensuing aggressive tumor progression present a significant clinical challenge. However, the mechanisms underlying the induction of tumor malignancy upon inhibition of steroid hormone signaling remain poorly understood. Here, we demonstrate that Drosophila malignant epithelial tumors show a similar reduction in ecdysone signaling, the main steroid hormone pathway. Our analysis of ecdysone-induced downstream targets reveals that overexpression of the nuclear receptor E75, particularly facilitates the malignant transformation of benign tumors. Genome-wide DNA binding profiles and biochemistry data reveal that E75 not only binds to the transcription factors of both Hippo and Notch pathways, but also exhibits widespread co-binding to their target genes, thus contributing to tumor malignancy. We further validated these findings by demonstrating that depletion of NR1D2, the mammalian homolog of E75, inhibits the activation of Hippo and Notch target genes, impeding glioblastoma progression. Together, our study unveils a novel mechanism by which hormone inhibition promotes tumor malignancy, and describes an evolutionarily conserved role of the oncogene E75/NR1D2 in integration of Hippo and Notch pathway activity during tumor progression.
Collapse
Affiliation(s)
- Xianping Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Yifan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Peng Lin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sha Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Du Kong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Yin Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Yanxiao Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Fei Lu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Qi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
5
|
Kandelouei T, Houghton ME, Lewis MR, Keller CC, Marchetti M, Kang X, Edgar BA. Mating and ecdysone signaling modify growth, metabolism, and digestive efficiency in the female Drosophila gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624434. [PMID: 39605564 PMCID: PMC11601506 DOI: 10.1101/2024.11.19.624434] [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
Adaptive changes in organ size and physiology occur in most adult animals, but how these changes are regulated is not well understood. Previous research found that mating in Drosophila females drives not only increases in gut size and stem cell proliferation but also alters feeding behavior, intestinal gene expression, and whole-body lipid storage, suggesting altered gut metabolism. Here, we show that mating dramatically alters female gut metabolism and digestive function. In addition to promoting a preference for a high-protein diet, mating also altered levels of TCA cycle intermediates and fatty acids in the gut, increased total gut lipids and protein, reduced relative carbohydrate levels, and enhanced the efficiency of protein digestion relative to carbohydrate digestion. The expression of genes that mediate each of these metabolic processes was similarly altered. In addition, we noted the mating-dependent downregulation of oxidative stress response and autophagy genes. Mating-dependent increases in ecdysone signaling played an important role in re-programming many, but not all, of these changes in the female gut. This study contributes to our understanding of how steroid signaling alters gut physiology to adapt to the demands of reproduction.
Collapse
Affiliation(s)
- Tahmineh Kandelouei
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, U.S.A
| | - Madeline E. Houghton
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, U.S.A
| | - Mitchell R. Lewis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, U.S.A
| | - Caroline C. Keller
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, U.S.A
| | - Marco Marchetti
- Eccles Institute of Human Genetics, 3i-UCGD Bioinformatics Core, University of Utah, Salt Lake City, Utah, U.S.A
| | - Xiaoyu Kang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, U.S.A
| | - Bruce A. Edgar
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, U.S.A
| |
Collapse
|
6
|
Wang N, Wang K, Lei L, Zhang L, Guo X, Xu B, Wang Y, Wang C. Juvenile hormone III improves honeybee resistance to imidacloprid by protecting the midgut. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117280. [PMID: 39515199 DOI: 10.1016/j.ecoenv.2024.117280] [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: 07/28/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Imidacloprid is one of the most commonly used insecticides and seriously threatens the survival of honeybees. Juvenile hormone III (JHIII), an important hormone in honeybees, plays a central role in the life cycle of honeybees; however, the mechanism by which JHIII responds to imidacloprid stress has not been determined. In this study, the resistance of Apis cerana cerana (Acc) to imidacloprid was greater than that of Apis mellifera (Am), and the application of JHIII significantly increased the resistance of both species of honeybees. RTqPCR revealed that two genes, juvenile hormone acid methyltransferase (JHAMT) and methyl farnesoate epoxidase (MFE), which are involved in JHIII biosynthesis, are key genes for improving resistance to imidacloprid via JHIII in honeybees. Silencing JHAMT and MFE with RNAi resulted in a lower content of JHIII in Acc and reduced resistance to imidacloprid. The exogenous application of JHIII compensated for the reduced resistance phenotype caused by gene silencing. Furthermore, histological examination of paraffin sections revealed that imidacloprid treatment caused loose intestinal cell arrangement, slight damage to columnar digestive cells and muscle cells, and chromatin condensation in basal layer cells. JHIII treatment reduced the degree of damage to the honeybee midgut cells and improved resistance to imidacloprid. The results of this study provide a new perspective and molecular basis for the study of the regulation of JHIII and the toxicity of imidacloprid in honeybees.
Collapse
Affiliation(s)
- Nana Wang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Kunrong Wang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Li Lei
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Longtao Zhang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Xingqi Guo
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Ying Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China.
| | - Chen Wang
- College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, China.
| |
Collapse
|
7
|
Box AM, Ramesh NA, Nandakumar S, Church SJ, Prasad D, Afrakhteh A, Taichman RS, Buttitta L. Cell cycle variants during Drosophila male accessory gland development. G3 (BETHESDA, MD.) 2024; 14:jkae089. [PMID: 38683731 PMCID: PMC11228851 DOI: 10.1093/g3journal/jkae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 01/31/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024]
Abstract
The Drosophila melanogaster male accessory gland (AG) is a functional analog of the mammalian prostate and seminal vesicles containing two secretory epithelial cell types, termed main and secondary cells. This tissue is responsible for making and secreting seminal fluid proteins and other molecules that contribute to successful reproduction. The cells of this tissue are binucleate and polyploid, due to variant cell cycles that include endomitosis and endocycling during metamorphosis. Here, we provide evidence of additional cell cycle variants in this tissue. We show that main cells of the gland are connected by ring canals that form after the penultimate mitosis, and we describe an additional post-eclosion endocycle required for gland maturation that is dependent on juvenile hormone signaling. We present evidence that the main cells of the D. melanogaster AG undergo a unique cell cycle reprogramming throughout organ development that results in step-wise cell cycle truncations culminating in cells containing two octoploid nuclei with under-replicated heterochromatin in the mature gland. We propose this tissue as a model to study developmental and hormonal temporal control of cell cycle variants in terminally differentiating tissues.
Collapse
Affiliation(s)
- Allison M Box
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| | - Navyashree A Ramesh
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| | - Shyama Nandakumar
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| | - Samuel Jaimian Church
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| | - Dilan Prasad
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| | - Ariana Afrakhteh
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| | - Russell S Taichman
- Department of Periodontology, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Laura Buttitta
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, 1105 N. University Ave. Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Landis GN, Bell HS, Peng OK, Fan Y, Yan K, Baybutt B, Tower J. Conditional Inhibition of Eip75B Eliminates the Effects of Mating and Mifepristone on Lifespan in Female Drosophila. Cells 2024; 13:1123. [PMID: 38994975 PMCID: PMC11240670 DOI: 10.3390/cells13131123] [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: 05/11/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024] Open
Abstract
Mating in female Drosophila melanogaster causes midgut hypertrophy and reduced lifespan, and these effects are blocked by the drug mifepristone. Eip75B is a transcription factor previously reported to have pleiotropic effects on Drosophila lifespan. Because Eip75B null mutations are lethal, conditional systems and/or partial knock-down are needed to study Eip75B effects in adults. Previous studies showed that Eip75B is required for adult midgut cell proliferation in response to mating. To test the possible role of Eip75B in mediating the lifespan effects of mating and mifepristone, a tripartite FLP-recombinase-based conditional system was employed that provides controls for genetic background. Expression of a Hsp70-FLP transgene was induced in third instar larvae by a brief heat pulse. The FLP recombinase catalyzed the recombination and activation of an Actin5C-GAL4 transgene. The GAL4 transcription factor in turn activated expression of a UAS-Eip75B-RNAi transgene. Inhibition of Eip75B activity was confirmed by loss of midgut hypertrophy upon mating, and the lifespan effects of both mating and mifepristone were eliminated. In addition, the negative effects of mifepristone on egg production were eliminated. The data indicate that Eip75B mediates the effects of mating and mifepristone on female midgut hypertrophy, egg production, and lifespan.
Collapse
Affiliation(s)
| | | | | | | | | | | | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, USA
| |
Collapse
|
9
|
Grmai L, Jimenez E, Baxter E, Doren MV. Steroid signaling controls sex-specific development in an invertebrate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573099. [PMID: 38187640 PMCID: PMC10769319 DOI: 10.1101/2023.12.22.573099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
In vertebrate sexual development, two important steroid hormones, testosterone and estrogen, regulate the sex-specific development of many tissues. In contrast, invertebrates utilize a single steroid hormone, ecdysone, to regulate developmental timing in both sexes. However, here we show that in Drosophila melanogaster, sex-specific ecdysone (E) signaling controls important aspects of gonad sexual dimorphism. Rather than being regulated at the level of hormone production, hormone activity is regulated cell-autonomously through sex-specific hormone reception. Ecdysone receptor (EcR) expression is restricted to the developing ovary and is repressed in the testis at a time when ecdysone initiates ovary morphogenesis. Interestingly, EcR expression is regulated downstream of the sex determination factor Doublesex (Dsx), the founding member of the Dsx/Mab3 Related Transcription Factor (DMRT) family that regulates gonad development in all animals. E signaling is required for normal ovary development1,2, and ectopic activation of E signaling in the testis antagonized stem cell niche identity and feminized somatic support cells, which were transformed into follicle-like cells. This work demonstrates that invertebrates can also use steroid hormone signaling to control sex-specific development. Further, it may help explain recent work showing that vertebrate sexual development is surprisingly cell-autonomous. For example, chickens utilize testosterone and estrogen to control sex-specific development, but when they have a mixture of cells with male and female genotypes, the male cells develop as male and the female cells develop as female despite exposure to the same circulating hormones3. Sex-specific regulation of steroid hormone signaling may well underly such cell-autonomous sexual fate choices in vertebrates as it does in Drosophila.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erin Jimenez
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ellen Baxter
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Taracena-Agarwal ML, Walter-Nuno AB, Bottino-Rojas V, Mejia APG, Xu K, Segal S, Dotson EM, Oliveira PL, Paiva-Silva GO. Juvenile Hormone as a contributing factor in establishing midgut microbiota for fecundity and fitness enhancement in adult female Aedes aegypti. Commun Biol 2024; 7:687. [PMID: 38839829 PMCID: PMC11153597 DOI: 10.1038/s42003-024-06334-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: 07/13/2023] [Accepted: 05/15/2024] [Indexed: 06/07/2024] Open
Abstract
Understanding the factors influencing mosquitoes' fecundity and longevity is important for designing better and more sustainable vector control strategies, as these parameters can impact their vectorial capacity. Here, we address how mating affects midgut growth in Aedes aegypti, what role Juvenile Hormone (JH) plays in this process, and how it impacts the mosquito's immune response and microbiota. Our findings reveal that mating and JH induce midgut growth. Additionally, the establishment of a native bacterial population in the midgut due to JH-dependent suppression of the immune response has important reproductive outcomes. Specific downregulation of AMPs with an increase in bacteria abundance in the gut results in increased egg counts and longer lifespans. Overall, these findings provide evidence of a cross-talk between JH response, gut epithelial tissue, cell cycle regulation, and the mechanisms governing the trade-offs between nutrition, immunity, and reproduction at the cellular level in the mosquito gut.
Collapse
Affiliation(s)
- Mabel L Taracena-Agarwal
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil.
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA.
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA.
| | - Ana Beatriz Walter-Nuno
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Vanessa Bottino-Rojas
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | | | - Kelsey Xu
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
| | - Steven Segal
- Entomology Department, Cornell University, College of Agriculture and Life Sciences, Ithaca, NY, USA
| | - Ellen M Dotson
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Pedro L Oliveira
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Gabriela O Paiva-Silva
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil.
| |
Collapse
|
11
|
Kurogi Y, Mizuno Y, Kamiyama T, Niwa R. The intestinal stem cell/enteroblast-GAL4 driver, escargot-GAL4, also manipulates gene expression in the juvenile hormone-synthesizing organ of Drosophila melanogaster. Sci Rep 2024; 14:9631. [PMID: 38671036 PMCID: PMC11053112 DOI: 10.1038/s41598-024-60269-2] [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: 01/12/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
Intestinal stem cells (ISCs) of the fruit fly, Drosophila melanogaster, offer an excellent genetic model to explore homeostatic roles of ISCs in animal physiology. Among available genetic tools, the escargot (esg)-GAL4 driver, expressing the yeast transcription factor gene, GAL4, under control of the esg gene promoter, has contributed significantly to ISC studies. This driver facilitates activation of genes of interest in proximity to a GAL4-binding element, Upstream Activating Sequence, in ISCs and progenitor enteroblasts (EBs). While esg-GAL4 has been considered an ISC/EB-specific driver, recent studies have shown that esg-GAL4 is also active in other tissues, such as neurons and ovaries. Therefore, the ISC/EB specificity of esg-GAL4 is questionable. In this study, we reveal esg-GAL4 expression in the corpus allatum (CA), responsible for juvenile hormone (JH) production. When driving the oncogenic gene, RasV12, esg-GAL4 induces overgrowth in ISCs/EBs as reported, but also increases CA cell number and size. Consistent with this observation, animals alter expression of JH-response genes. Our data show that esg-GAL4-driven gene manipulation can systemically influence JH-mediated animal physiology, arguing for cautious use of esg-GAL4 as a "specific" ISC/EB driver to examine ISC/EB-mediated animal physiology.
Collapse
Affiliation(s)
- Yoshitomo Kurogi
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Yosuke Mizuno
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Takumi Kamiyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
12
|
Zhang S, Wu S, Yao R, Wei X, Ohlstein B, Guo Z. Eclosion muscles secrete ecdysteroids to initiate asymmetric intestinal stem cell division in Drosophila. Dev Cell 2024; 59:125-140.e12. [PMID: 38096823 DOI: 10.1016/j.devcel.2023.11.016] [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: 06/29/2023] [Revised: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 01/11/2024]
Abstract
During organ development, tissue stem cells first expand via symmetric divisions and then switch to asymmetric divisions to minimize the time to obtain a mature tissue. In the Drosophila midgut, intestinal stem cells switch their divisions from symmetric to asymmetric at midpupal development to produce enteroendocrine cells. However, the signals that initiate this switch are unknown. Here, we identify the signal as ecdysteroids. In the presence of ecdysone, EcR and Usp promote the expression of E93 to suppress Br expression, resulting in asymmetric divisions. Surprisingly, the primary source of pupal ecdysone is not from the prothoracic gland but from dorsal internal oblique muscles (DIOMs), a group of transient skeletal muscles that are required for eclosion. Genetic analysis shows that DIOMs secrete ecdysteroids during mTOR-mediated muscle remodeling. Our findings identify sequential endocrine and mechanical roles for skeletal muscle, which ensure the timely asymmetric divisions of intestinal stem cells.
Collapse
Affiliation(s)
- Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Song Wu
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruining Yao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueying Wei
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Benjamin Ohlstein
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
13
|
Khanbabei A, Segura L, Petrossian C, Lemus A, Cano I, Frazier C, Halajyan A, Ca D, Loza-Coll M. Experimental validation and characterization of putative targets of Escargot and STAT, two master regulators of the intestinal stem cells in Drosophila melanogaster. Dev Biol 2024; 505:148-163. [PMID: 37952851 DOI: 10.1016/j.ydbio.2023.10.008] [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: 04/02/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023]
Abstract
Many organs contain adult stem cells (ASCs) to replace cells due to damage, disease, or normal tissue turnover. ASCs can divide asymmetrically, giving rise to a new copy of themselves (self-renewal) and a sister that commits to a specific cell type (differentiation). Decades of research have led to the identification of pleiotropic genes whose loss or gain of function affect diverse aspects of normal ASC biology. Genome-wide screens of these so-called genetic "master regulator" (MR) genes, have pointed to hundreds of putative targets that could serve as their downstream effectors. Here, we experimentally validate and characterize the regulation of several putative targets of Escargot (Esg) and the Signal Transducer and Activator of Transcription (Stat92E, a.k.a. STAT), two known MRs in Drosophila intestinal stem cells (ISCs). Our results indicate that regardless of bioinformatic predictions, most experimentally validated targets show a profile of gene expression that is consistent with co-regulation by both Esg and STAT, fitting a rather limited set of co-regulatory modalities. A bioinformatic analysis of proximal regulatory sequences in specific subsets of co-regulated targets identified additional transcription factors that might cooperate with Esg and STAT in modulating their transcription. Lastly, in vivo manipulations of validated targets rarely phenocopied the effects of manipulating Esg and STAT, suggesting the existence of complex genetic interactions among downstream targets of these two MR genes during ISC homeostasis.
Collapse
Affiliation(s)
- Armen Khanbabei
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Lina Segura
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Cynthia Petrossian
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Aaron Lemus
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Ithan Cano
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Courtney Frazier
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Armen Halajyan
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Donnie Ca
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Mariano Loza-Coll
- Department of Biology, California State University, Northridge (CSUN), USA.
| |
Collapse
|
14
|
Troost T, Seib E, Airich A, Vüllings N, Necakov A, De Renzis S, Klein T. The meaning of ubiquitylation of the DSL ligand Delta for the development of Drosophila. BMC Biol 2023; 21:260. [PMID: 37974242 PMCID: PMC10655352 DOI: 10.1186/s12915-023-01759-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Ubiquitylation (ubi) of the intracellular domain of the Notch ligand Delta (Dl) by the E3 ligases Neuralized (Neur) and Mindbomb1 (Mib1) on lysines (Ks) is thought to be essential for the its signalling activity. Nevertheless, we have previously shown that DlK2R-HA, a Dl variant where all Ks in its intracellular domain (ICD) are replaced by the structurally similar arginine (R), still possess weak activity if over-expressed. This suggests that ubi is not absolutely required for Dl signalling. However, it is not known whether the residual activity of DlK2R-HA is an effect of over-expression and, if not, whether DlK2R can provide sufficient activity for the whole development of Drosophila. RESULTS To clarify these issues, we generated and analysed DlattP-DlK2R-HA, a knock-in allele into the Dl locus. Our analysis of this allele reveals that the sole presence of one copy of DlattP-DlK2R-HA can provide sufficient activity for completion of development. It further indicates that while ubi is required for the full activity of Dl in Mib1-dependent processes, it is not essential for Neur-controlled neural development. We identify three modes of Dl signalling that are either dependent or independent of ubi. Importantly, all modes depend on the presence of the endocytic adapter Epsin. During activation of Dl, direct binding of Epsin appears not to be an essential requirement. In addition, our analysis further reveals that the Ks are required to tune down the cis-inhibitory interaction of Dl with Notch. CONCLUSIONS Our results indicate that Dl can activate the Notch pathway without ubi of its ICD. It signals via three modes. Ubi is specifically required for the Mib1-dependent processes and the adjustment of cis-inhibition. In contrast to Mib1, Neur can efficiently activate Dl without ubi. Neur probably acts as an endocytic co-adapter in addition to its role as E3 ligase. Endocytosis, regulated in a ubi-dependent or ubi-independent manner is required for signalling and also suppression of cis-inhibition. The findings clarify the role of ubi of the ligands during Notch signalling.
Collapse
Affiliation(s)
- Tobias Troost
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Ekaterina Seib
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Alina Airich
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Nicole Vüllings
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Aleksandar Necakov
- Department of Biological Science, Brock University, 1030, Ontario, L2S3A1, Canada
| | - Stefano De Renzis
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany.
| |
Collapse
|
15
|
Neophytou C, Soteriou E, Pitsouli C. The Sterol Transporter Npc2c Controls Intestinal Stem Cell Mitosis and Host-Microbiome Interactions in Drosophila. Metabolites 2023; 13:1084. [PMID: 37887409 PMCID: PMC10609107 DOI: 10.3390/metabo13101084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Cholesterol is necessary for all cells to function. The intracellular cholesterol transporters Npc1 and Npc2 control sterol trafficking and their malfunction leads to Neimann-Pick Type C disease, a rare disorder affecting the nervous system and the intestine. Unlike humans that encode single Npc1 and Npc2 transporters, flies encompass two Npc1 (Npc1a-1b) and eight Npc2 (Npc2a-2h) members, and most of the Npc2 family genes remain unexplored. Here, we focus on the intestinal function of Npc2c in the adult. We find that Npc2c is necessary for intestinal stem cell (ISC) mitosis, maintenance of the ISC lineage, survival upon pathogenic infection, as well as tumor growth. Impaired mitosis of Npc2c-silenced midguts is accompanied by reduced expression of Cyclin genes, and genes encoding ISC regulators, such as Delta, unpaired1 and Socs36E. ISC-specific Npc2c silencing induces Attacin-A expression, a phenotype reminiscent of Gram-negative bacteria overabundance. Metagenomic analysis of Npc2c-depleted midguts indicates intestinal dysbiosis, whereby decreased commensal complexity is accompanied by increased gamma-proteobacteria. ISC-specific Npc2c silencing also results in increased cholesterol aggregation. Interestingly, administration of the non-steroidal ecdysone receptor agonist, RH5849, rescues mitosis of Npc2c-silenced midguts and increases expression of the ecdysone response gene Broad, underscoring the role of Npc2c and sterols in ecdysone signaling. Assessment of additional Npc2 family members indicates potential redundant roles with Npc2c in ISC control and response to ecdysone signaling. Our results highlight a previously unidentified essential role of Npc2c in ISC mitosis, as well as an important role in ecdysone signaling and microbiome composition in the Drosophila midgut.
Collapse
Affiliation(s)
| | | | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, 2109 Aglantzia, Cyprus; (C.N.); (E.S.)
| |
Collapse
|
16
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
17
|
Abstract
Endocrine signaling networks control diverse biological processes and life history traits across metazoans. In both invertebrate and vertebrate taxa, steroid hormones regulate immune system function in response to intrinsic and environmental stimuli, such as microbial infection. The mechanisms of this endocrine-immune regulation are complex and constitute an ongoing research endeavor facilitated by genetically tractable animal models. The 20-hydroxyecdysone (20E) is the major steroid hormone in arthropods, primarily studied for its essential role in mediating developmental transitions and metamorphosis; 20E also modulates innate immunity in a variety of insect taxa. This review provides an overview of our current understanding of 20E-mediated innate immune responses. The prevalence of correlations between 20E-driven developmental transitions and innate immune activation are summarized across a range of holometabolous insects. Subsequent discussion focuses on studies conducted using the extensive genetic resources available in Drosophila that have begun to reveal the mechanisms underlying 20E regulation of immunity in the contexts of both development and bacterial infection. Lastly, I propose directions for future research into 20E regulation of immunity that will advance our knowledge of how interactive endocrine networks coordinate animals' physiological responses to environmental microbes.
Collapse
Affiliation(s)
- Scott A. Keith
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
18
|
Zhang Y, Chen R, Gong L, Huang W, Li P, Zhai Z, Ling E. Regulation of intestinal stem cell activity by a mitotic cell cycle regulator Polo in Drosophila. G3 (BETHESDA, MD.) 2023; 13:jkad084. [PMID: 37154439 PMCID: PMC10234410 DOI: 10.1093/g3journal/jkad084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/31/2023] [Indexed: 05/10/2023]
Abstract
Maintaining a definite and stable pool of dividing stem cells plays an important role in organ development. This process requires an appropriate progression of mitosis for proper spindle orientation and polarity to ensure the ability of stem cells to proliferate and differentiate correctly. Polo-like kinases (Plks)/Polo are the highly conserved serine/threonine kinases involved in the initiation of mitosis as well as in the progression of the cell cycle. Although numerous studies have investigated the mitotic defects upon loss of Plks/Polo in cells, little is known about the in vivo consequences of stem cells with abnormal Polo activity in the context of tissue and organism development. The current study aimed to investigate this question using the Drosophila intestine, an organ dynamically maintained by the intestinal stem cells (ISCs). The results indicated that the polo depletion caused a reduction in the gut size due to a gradual decrease in the number of functional ISCs. Interestingly, the polo-deficient ISCs showed an extended G2/M phase and aneuploidy and were subsequently eliminated by premature differentiation into enterocytes (ECs). In contrast, the constitutively active Polo (poloT182D) suppressed ISC proliferation, induced abnormal accumulation of β-tubulin in cells, and drove ISC loss via apoptosis. Therefore, Polo activity should be properly maintained for optimal stem cell function. Further analysis suggested that polo was a direct target gene of Sox21a, a Sox transcription factor that critically regulates stem cell activity. Together, this study provided a novel perspective on the correlation between the progression of mitosis and the ISC function in Drosophila.
Collapse
Affiliation(s)
- Ying Zhang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Rongbing Chen
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Liyuan Gong
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wuren Huang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
| | - Ping Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zongzhao Zhai
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Erjun Ling
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, The Chinese Academy of Science, Shanghai 200032, China
| |
Collapse
|
19
|
Ye C, Behnke JA, Hardin KR, Zheng JQ. Drosophila melanogaster as a model to study age and sex differences in brain injury and neurodegeneration after mild head trauma. Front Neurosci 2023; 17:1150694. [PMID: 37077318 PMCID: PMC10106652 DOI: 10.3389/fnins.2023.1150694] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Repetitive physical insults to the head, including those that elicit mild traumatic brain injury (mTBI), are a known risk factor for a variety of neurodegenerative conditions including Alzheimer's disease (AD), Parkinson's disease (PD), and chronic traumatic encephalopathy (CTE). Although most individuals who sustain mTBI typically achieve a seemingly full recovery within a few weeks, a subset experience delayed-onset symptoms later in life. As most mTBI research has focused on the acute phase of injury, there is an incomplete understanding of mechanisms related to the late-life emergence of neurodegeneration after early exposure to mild head trauma. The recent adoption of Drosophila-based brain injury models provides several unique advantages over existing preclinical animal models, including a tractable framework amenable to high-throughput assays and short relative lifespan conducive to lifelong mechanistic investigation. The use of flies also provides an opportunity to investigate important risk factors associated with neurodegenerative conditions, specifically age and sex. In this review, we survey current literature that examines age and sex as contributing factors to head trauma-mediated neurodegeneration in humans and preclinical models, including mammalian and Drosophila models. We discuss similarities and disparities between human and fly in aging, sex differences, and pathophysiology. Finally, we highlight Drosophila as an effective tool for investigating mechanisms underlying head trauma-induced neurodegeneration and for identifying therapeutic targets for treatment and recovery.
Collapse
Affiliation(s)
- Changtian Ye
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Joseph A. Behnke
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Katherine R. Hardin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - James Q. Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
20
|
Piper MDW, Zanco B, Sgrò CM, Adler MI, Mirth CK, Bonduriansky R. Dietary restriction and lifespan: adaptive reallocation or somatic sacrifice? FEBS J 2023; 290:1725-1734. [PMID: 35466532 PMCID: PMC10952493 DOI: 10.1111/febs.16463] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/28/2022] [Accepted: 04/21/2022] [Indexed: 12/21/2022]
Abstract
Reducing overall food intake, or lowering the proportion of protein relative to other macronutrients, can extend the lifespan of diverse organisms. A number of mechanistic theories have been developed to explain this phenomenon, mostly assuming that the molecules connecting diet to lifespan are evolutionarily conserved. A recent study using Drosophila melanogaster females has pinpointed a single essential micronutrient that can explain how lifespan is changed by dietary restriction. Here, we propose a likely mechanism for this observation, which involves a trade-off between lifespan and reproduction, but in a manner that is conditional on the dietary supply of an essential micronutrient - a sterol. Importantly, these observations argue against previous evolutionary theories that rely on constitutive resource reallocation or damage directly inflicted by reproduction. Instead, they are compatible with a model in which the inverse relationship between lifespan and food level is caused by the consumer suffering from varying degrees of malnutrition when maintained on lab food. The data also indicate that animals on different lab foods may suffer from different nutritional imbalances and that the mechanisms by which dietary restriction benefits the lifespan of different species may vary. This means that translating the mechanistic findings from lab animals to humans will not be simple and should be interpreted in light of the range of challenges that have shaped each organism's lifespan in the wild and the composition of the natural diets upon which they would feed.
Collapse
Affiliation(s)
| | - Brooke Zanco
- School of Biological SciencesMonash UniversityClaytonVictoriaAustralia
| | - Carla M. Sgrò
- School of Biological SciencesMonash UniversityClaytonVictoriaAustralia
| | | | - Christen K. Mirth
- School of Biological SciencesMonash UniversityClaytonVictoriaAustralia
| | - Russell Bonduriansky
- School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyAustralia
| |
Collapse
|
21
|
Delbare SYN, Venkatraman S, Scuderi K, Wells MT, Wolfner MF, Basu S, Clark AG. Time series transcriptome analysis implicates the circadian clock in the Drosophila melanogaster female's response to sex peptide. Proc Natl Acad Sci U S A 2023; 120:e2214883120. [PMID: 36706221 PMCID: PMC9945991 DOI: 10.1073/pnas.2214883120] [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: 09/01/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
Sex peptide (SP), a seminal fluid protein of Drosophila melanogaster males, has been described as driving a virgin-to-mated switch in females, through eliciting an array of responses including increased egg laying, activity, and food intake and a decreased remating rate. While it is known that SP achieves this, at least in part, by altering neuronal signaling in females, the genetic architecture and temporal dynamics of the female's response to SP remain elusive. We used a high-resolution time series RNA-sequencing dataset of female heads at 10 time points within the first 24 h after mating to learn about the genetic architecture, at the gene and exon levels, of the female's response to SP. We find that SP is not essential to trigger early aspects of a virgin-to-mated transcriptional switch, which includes changes in a metabolic gene regulatory network. However, SP is needed to maintain and diversify metabolic changes and to trigger changes in a neuronal gene regulatory network. We further find that SP alters rhythmic gene expression in females and suggests that SP's disruption of the female's circadian rhythm might be key to its widespread effects.
Collapse
Affiliation(s)
- Sofie Y. N. Delbare
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY14853
- Department of Statistics & Data Science, Cornell University, Ithaca, NY14853
| | - Sara Venkatraman
- Department of Statistics & Data Science, Cornell University, Ithaca, NY14853
| | - Kate Scuderi
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY14853
| | - Martin T. Wells
- Department of Statistics & Data Science, Cornell University, Ithaca, NY14853
| | - Mariana F. Wolfner
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY14853
| | - Sumanta Basu
- Department of Statistics & Data Science, Cornell University, Ithaca, NY14853
| | - Andrew G. Clark
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY14853
| |
Collapse
|
22
|
Hoedjes KM, Kostic H, Flatt T, Keller L. A Single Nucleotide Variant in the PPARγ-homolog Eip75B Affects Fecundity in Drosophila. Mol Biol Evol 2023; 40:7005670. [PMID: 36703226 PMCID: PMC9922802 DOI: 10.1093/molbev/msad018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Single nucleotide polymorphisms are the most common type of genetic variation, but how these variants contribute to the adaptation of complex phenotypes is largely unknown. Experimental evolution and genome-wide association studies have demonstrated that variation in the PPARγ-homolog Eip75B has associated with longevity and life-history differences in the fruit fly Drosophila melanogaster. Using RNAi knockdown, we first demonstrate that reduced expression of Eip75B in adult flies affects lifespan, egg-laying rate, and egg volume. We then tested the effects of a naturally occurring SNP within a cis-regulatory domain of Eip75B by applying two complementary approaches: a Mendelian randomization approach using lines of the Drosophila Genetic Reference Panel, and allelic replacement using precise CRISPR/Cas9-induced genome editing. Our experiments reveal that this natural polymorphism has a significant pleiotropic effect on fecundity and egg-to-adult viability, but not on longevity or other life-history traits. Our results provide a rare functional validation at the nucleotide level and identify a natural allelic variant affecting fitness and life-history adaptation.
Collapse
Affiliation(s)
| | - Hristina Kostic
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
23
|
Li H, Luo X, Li N, Liu T, Zhang J. Insulin-like peptide 8 (Ilp8) regulates female fecundity in flies. Front Cell Dev Biol 2023; 11:1103923. [PMID: 36743416 PMCID: PMC9890075 DOI: 10.3389/fcell.2023.1103923] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Introduction: Insulin-like peptides (Ilps) play crucial roles in nearly all life stages of insects. Ilp8 is involved in developmental stability, stress resistance and female fecundity in several insect species, but the underlying mechanisms are not fully understood. Here we report the functional characterization of Ilp8s in three fly species, including Bactrocera dorsalis, Drosophila mercatorum and Drosophila melanogaster. Methods: Phylogenetic analyses were performed to identify and characterize insect Ilp8s. The amino acid sequences of fly Ilp8s were aligned and the three-dimensional structures of fly Ilp8s were constructed and compared. The tissue specific expression pattern of fly Ilp8s were examined by qRT-PCR. In Bactrocera dorsalis and Drosophila mercatorum, dsRNAs were injected into virgin females to inhibit the expression of Ilp8 and the impacts on female fecundity were examined. In Drosophila melanogaster, the female fecundity of Ilp8 loss-of-function mutant was compared with wild type control flies. The mutant fruit fly strain was also used for sexual behavioral analysis and transcriptomic analysis. Results: Orthologs of Ilp8s are found in major groups of insects except for the lepidopterans and coleopterans, and Ilp8s are found to be well separated from other Ilps in three fly species. The key motif and the predicted three-dimensional structure of fly Ilp8s are well conserved. Ilp8 are specifically expressed in the ovary and are essential for female fecundity in three fly species. Behavior analysis demonstrates that Ilp8 mutation impairs female sexual attractiveness in fruit fly, which results in decreased mating success and is likely the cause of fecundity reduction. Further transcriptomic analysis indicates that Ilp8 might influence metabolism, immune activity, oocyte development as well as hormone homeostasis to collectively regulate female fecundity in the fruit fly. Discussion: Our findings support a universal role of insect Ilp8 in female fecundity, and also provide novel clues for understanding the modes of action of Ilp8.
Collapse
Affiliation(s)
- Haomiao Li
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xi Luo
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China
| | - Na Li
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China
| | - Tao Liu
- Chinese Academy of Inspection and Quarantine, Beijing, China
| | - Junzheng Zhang
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China,*Correspondence: Junzheng Zhang,
| |
Collapse
|
24
|
N. Landis G, Ko S, Peng O, Bognar B, Khmelkov M, S. Bell H, Tower J. A screen of small molecule and genetic modulators of life span in female Drosophila identifies etomoxir, RH5849 and unanticipated temperature effects. Fly (Austin) 2022; 16:397-413. [PMID: 36412257 PMCID: PMC9683069 DOI: 10.1080/19336934.2022.2149209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022] Open
Abstract
Mifepristone increases life span in female Drosophila melanogaster, and its molecular target(s) remain unclear. Here small molecule and genetic interventions were tested for ability to mimic mifepristone, or to decrease life span in a way that can be rescued by mifepristone. Etomoxir inhibits lipid metabolism, and significantly increased life span in virgin and mated females, but not males, at 50 µM concentration. Pioglitazone is reported to activate both mammalian PPARγ and its Drosophila homolog Eip75B. Pioglitazone produced minor and inconsistent benefits for female Drosophila life span, and only at the lowest concentrations tested. Ecdysone is a Drosophila steroid hormone reported to regulate responses to mating, and RH5849 is a potent mimic of ecdysone. RH5849 reduced virgin female life span, and this was partly rescued by mifepristone. Mifepristone did not compete with RH5849 for activation of an ecdysone receptor (EcR)-responsive transgenic reporter, indicating that the relevant target for mifepristone is not EcR. The conditional GAL4/GAL80ts system was used in attempt to test the effect of an Eip75B RNAi construct on female life span. However, the 29°C temperature used for induction reduced or eliminated mating-induced midgut hypertrophy, the negative life span effects of mating, and the positive life span effects of mifepristone. Even when applied after mating was complete, a shift to 29°C temperature reduced mating-induced midgut hypertrophy by half, and the life span effects of mating by 4.8-fold. Taken together, these results identify promising small molecules for further analysis, and inform the design of experiments involving the GAL4/GAL80ts system.
Collapse
Affiliation(s)
- Gary N. Landis
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Sebastian Ko
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Oscar Peng
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Brett Bognar
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Michael Khmelkov
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Hans S. Bell
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
25
|
Okamoto N, Watanabe A. Interorgan communication through peripherally derived peptide hormones in Drosophila. Fly (Austin) 2022; 16:152-176. [PMID: 35499154 PMCID: PMC9067537 DOI: 10.1080/19336934.2022.2061834] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
In multicellular organisms, endocrine factors such as hormones and cytokines regulate development and homoeostasis through communication between different organs. For understanding such interorgan communications through endocrine factors, the fruit fly Drosophila melanogaster serves as an excellent model system due to conservation of essential endocrine systems between flies and mammals and availability of powerful genetic tools. In Drosophila and other insects, functions of neuropeptides or peptide hormones from the central nervous system have been extensively studied. However, a series of recent studies conducted in Drosophila revealed that peptide hormones derived from peripheral tissues also play critical roles in regulating multiple biological processes, including growth, metabolism, reproduction, and behaviour. Here, we summarise recent advances in understanding target organs/tissues and functions of peripherally derived peptide hormones in Drosophila and describe how these hormones contribute to various biological events through interorgan communications.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Watanabe
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
26
|
Khalid MZ, Sun Z, Zhang J, Zhang S, Zhong G. Cyromazine affects the ovarian germ cells of Drosophila via the ecdysone signaling pathway. Front Physiol 2022; 13:992306. [PMID: 36246127 PMCID: PMC9557234 DOI: 10.3389/fphys.2022.992306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cyromazine, an insect growth regulator, has been extensively used against the insect pests of livestock and households. Previously, it was observed that the continuous selection of cyromazine from the larval to the adult stage decreased the number of germline stem cells (GSCs) and cystoblasts (CBs) in the adult ovary. In addition, in this study, we observed that the number of primordial germ cells (PGCs) was also decreased in the larval ovary after treatment with cyromazine. However, the mechanism by which it affects the germ cells is yet to be explored. Consequently, to deeply investigate the effects of cyromazine on the germ cells, we performed tissue-specific RNA sequencing. Bioinformatics analysis revealed that the ecdysone signaling pathway was significantly influenced under cyromazine stress. Based on that, we screened and selected 14 ecdysone signaling responsive genes and silenced their expression in the germ cells only. Results of that showed a considerable reduction in the number of germ cells. Furthermore, we mixed exogenous 20E with the cyromazine-containing diet to rescue the ecdysone signaling. Our results supported that the application of exogenous 20E significantly rescued the germ cells in the transgenic lines. Therefore, this implies that the cyromazine decreased the number of germ cells by affecting the ecdysone signaling pathway.
Collapse
|
27
|
Landis GN, Riggan L, Bell HS, Vu W, Wang T, Wang I, Tejawinata FI, Ko S, Tower J. Mifepristone Increases Life Span in Female Drosophila Without Detectable Antibacterial Activity. FRONTIERS IN AGING 2022; 3:924957. [PMID: 35935727 PMCID: PMC9354577 DOI: 10.3389/fragi.2022.924957] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022]
Abstract
Mifepristone dramatically increases the life span of mated female Drosophila while reducing the expression of innate immune response genes. Previous results indicated that mifepristone also reduced the load of aero-tolerant bacteria in mated females. Experiments were conducted to further investigate the possible role of bacteria in mifepristone life span effects. Life span was assayed in flies grown from sterilized eggs on autoclaved media and in normally cultured controls in two independent assays. Sterilization increased mated female life span (+8.3% and +57%, respectively), and the effect of mifepristone was additive (+53% and +93%, respectively). High-throughput sequencing of 16S sequences revealed that sterilization reduced the abundance of multiple species and the classes Bacteroidia, Bacilli, Actinobacteria, and Cytophagia. By contrast, mifepristone caused no decreases and instead increased the abundance of three species. Five aero-tolerant bacterial species were cultured from extracts of mated female flies, including both Gram-positive and Gram-negative species (Acetobacter sicerae, Enterococcus faecalis, Lactobacillus plantarum, Serratia rubidea, and Paenibacillus glucanolyticus). There was no detectable effect of mifepristone on the growth of these bacteria in vitro, indicating that mifepristone does not have a direct antibiotic effect. To test if antibiotics could mimic the effects of mifepristone in vivo, mated female flies were treated throughout adult life span with high concentrations of the individual antibiotics doxycycline, ampicillin, kanamycin, and streptomycin, in replicate experiments. No significant effect on life span was observed for ampicillin, kanamycin, or streptomycin, and an inconsistent benefit was observed for doxycycline. Finally, supplementation of media with Enterococcus faecalis did not alter adult female life span in the presence or absence of mifepristone. Taken together, the results indicate the life span benefits of mifepristone are not due to an antibiotic effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
28
|
Stuart AKDC, Furuie JL, Cataldi TR, Stuart RM, Zawadneak MAC, Labate CA, Pimentel IC. Fungal consortium of two Beauveria bassiana strains increases their virulence, growth, and resistance to stress: A metabolomic approach. PLoS One 2022; 17:e0271460. [PMID: 35834517 PMCID: PMC9282594 DOI: 10.1371/journal.pone.0271460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
The use of two or more microorganisms in a microbial consortium has been increasingly applied in the biological control of diseases and pests. Beauveria bassiana is one of the most widely studied fungal species in biological control, yet little is known about its role in fungal consortiums. In a previous study, our group found that a consortium formed by two strains of B. bassiana had significantly greater biocontrol potential against the polyphagous caterpillars Duponchelia fovealis (Lepidoptera: Crambidae) than either strain on its own. In this study, we use GC-MS and LC-MS/MS to evaluate and discuss the metabolomics of the consortium. A total of 21 consortium biomarkers were identified, corresponding to 14 detected by LC-MS/MS and seven by GC-MS. Antioxidant and anti-inflammatory mechanisms are the main properties of the metabolites produced by the consortium. These metabolites can depress the insect’s immune system, increasing its vulnerability and, hence, the fungal virulence of the consortium. In light of these results, we propose an action model of insect mortality due to the metabolites secreted by the consortium. The model includes the inhibition of defense mechanisms such as pro-inflammatory interleukin secretion, cell migration, cell aggregation, Dif, Dorsal and Relish gene transcription, and JAK/STAT and JNK signaling pathways. It also promotes the cleaning of oxidative molecules, like ROS, NOS, and H2O2, and the induction of virulence factors.
Collapse
Affiliation(s)
- Andressa Katiski da Costa Stuart
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
- * E-mail:
| | - Jason Lee Furuie
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Thais Regiani Cataldi
- Departamento de Genética, Laboratório de Genética de Plantas Max Feffer, Escola Superior de Agronomia Luiz de Queiroz – Esalq/USP, Piracicaba, São Paulo, Brazil
| | - Rodrigo Makowiecky Stuart
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Maria Aparecida Cassilha Zawadneak
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
- Departamento de Fitotecnia e Fitossanitaríssimo, Programa de Pós-graduação em Agronomia Produção Vegetal, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Carlos Alberto Labate
- Departamento de Genética, Laboratório de Genética de Plantas Max Feffer, Escola Superior de Agronomia Luiz de Queiroz – Esalq/USP, Piracicaba, São Paulo, Brazil
| | - Ida Chapaval Pimentel
- Departamento de Patologia Básica, Setor de Ciências Biológicas, Laboratório de Microbiologia e Biologia Molecular (LabMicro), Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| |
Collapse
|
29
|
Cyromazine Effects the Reproduction of Drosophila by Decreasing the Number of Germ Cells in the Female Adult Ovary. INSECTS 2022; 13:insects13050414. [PMID: 35621750 PMCID: PMC9144682 DOI: 10.3390/insects13050414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 02/08/2023]
Abstract
Simple Summary Cyromazine, an insect growth regulator, is used to control the Dipteran pest population. Previous findings observed that treatment with cyromazine increased the larval mortality, by interfering with the ecdysone signaling. In addition, the application of exogenous 20E significantly reduced the mortality caused by cyromazine. Many studies have also supported the role of ecdysone signaling in the maintenance of germline stem cells (GSCs), where mutations in ecdysone signaling-related genes significantly decreased the number of GSCs. However, to date, no study has reported the effect of cyromazine on the GSCs of Drosophila melanogaster. In the present study, we observed that cyromazine significantly reduced the number of both GSCs and cystoblasts (CBs) in the ovary of adult female. To further understand the effect of cyromazine on germ cells, we selected some key genes related to the ecdysone signaling pathway and evaluated their expression through RT-qPCR. Additionally, we measured the ecdysone titer from the cyromazine-treated ovaries. Our results indicated a significant decrease in the expression of ecdysone signaling-related genes and also in the ecdysone titer. These results further supported our findings that cyromazine reduced the number of germ cells by interfering with the ecdysone signaling pathway. Abstract In the present study, we observed a 58% decrease in the fecundity of Drosophila melanogaster, after treatment with the cyromazine. To further elucidate the effects of cyromazine on reproduction, we counted the number of both germline stem cells (GSCs) and cystoblasts (CBs) in the ovary of a 3-day-old adult female. The results showed a significant decrease in the number of GSCs and CBs as compared to the control group. The mode of action of cyromazine is believed to be through the ecdysone signaling pathway. To further support this postulate, we observed the expression of key genes involved in the ecdysone signaling pathway and also determined the ecdysone titer from cyromazine-treated ovaries. Results indicated a significant decrease in the expression of ecdysone signaling-related genes as compared to the control group. Furthermore, the titer of the ecdysone hormone was also markedly reduced (90%) in cyromazine-treated adult ovaries, suggesting that ecdysone signaling was directly related to the decrease in the number of GSCs and CBs. However, further studies are required to understand the mechanism by which cyromazine affects the GSCs and CBs in female adult ovaries.
Collapse
|
30
|
Nagai H, Miura M, Nakajima YI. Cellular mechanisms underlying adult tissue plasticity in Drosophila. Fly (Austin) 2022; 16:190-206. [PMID: 35470772 PMCID: PMC9045823 DOI: 10.1080/19336934.2022.2066952] [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] [Indexed: 11/26/2022] Open
Abstract
Adult tissues in Metazoa dynamically remodel their structures in response to environmental challenges including sudden injury, pathogen infection, and nutritional fluctuation, while maintaining quiescence under homoeostatic conditions. This characteristic, hereafter referred to as adult tissue plasticity, can prevent tissue dysfunction and improve the fitness of organisms in continuous and/or severe change of environments. With its relatively simple tissue structures and genetic tools, studies using the fruit fly Drosophila melanogaster have provided insights into molecular mechanisms that control cellular responses, particularly during regeneration and nutrient adaptation. In this review, we present the current understanding of cellular mechanisms, stem cell proliferation, polyploidization, and cell fate plasticity, all of which enable adult tissue plasticity in various Drosophila adult organs including the midgut, the brain, and the gonad, and discuss the organismal strategy in response to environmental changes and future directions of the research.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Yu-Ichiro Nakajima
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Sex-specific regulation of development, growth and metabolism. Semin Cell Dev Biol 2022; 138:117-127. [PMID: 35469676 DOI: 10.1016/j.semcdb.2022.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/07/2022] [Accepted: 04/14/2022] [Indexed: 12/13/2022]
Abstract
Adult females and males of most species differ in many aspects of their morphology, physiology and behavior, in response to sex-specific selective pressures that maximize fitness. While we have an increasingly good understanding of the genetic mechanisms that initiate these differences, the sex-specific developmental trajectories that generate them are much less well understood. Here we review recent advances in the sex-specific regulation of development focusing on two models where this development is increasingly well understood: Sexual dimorphism of body size in the fruit fly Drosophila melanogaster and sexual dimorphism of horns in the horned beetle Onthophagus taurus. Because growth and development are also supported by metabolism, the regulation of sex-specific metabolism during and after development is an important aspect of the generation of female and male phenotypes. Hitherto, the study of sex-specific development has largely been independent of the study of sex-specific metabolism. Nevertheless, as we discuss in this review, recent research has begun to reveal considerable overlap in the cellular and physiological mechanisms that regulate sex-specific development and metabolism.
Collapse
|
32
|
Anagnostopoulos G, Motiño O, Li S, Carbonnier V, Chen H, Sica V, Durand S, Bourgin M, Aprahamian F, Nirmalathasan N, Donne R, Desdouets C, Sola MS, Kotta K, Montégut L, Lambertucci F, Surdez D, Sandrine G, Delattre O, Maiuri MC, Bravo-San Pedro JM, Martins I, Kroemer G. An obesogenic feedforward loop involving PPARγ, acyl-CoA binding protein and GABA A receptor. Cell Death Dis 2022; 13:356. [PMID: 35436993 PMCID: PMC9016078 DOI: 10.1038/s41419-022-04834-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
Acyl-coenzyme-A-binding protein (ACBP), also known as a diazepam-binding inhibitor (DBI), is a potent stimulator of appetite and lipogenesis. Bioinformatic analyses combined with systematic screens revealed that peroxisome proliferator-activated receptor gamma (PPARγ) is the transcription factor that best explains the ACBP/DBI upregulation in metabolically active organs including the liver and adipose tissue. The PPARγ agonist rosiglitazone-induced ACBP/DBI upregulation, as well as weight gain, that could be prevented by knockout of Acbp/Dbi in mice. Moreover, liver-specific knockdown of Pparg prevented the high-fat diet (HFD)-induced upregulation of circulating ACBP/DBI levels and reduced body weight gain. Conversely, knockout of Acbp/Dbi prevented the HFD-induced upregulation of PPARγ. Notably, a single amino acid substitution (F77I) in the γ2 subunit of gamma-aminobutyric acid A receptor (GABAAR), which abolishes ACBP/DBI binding to this receptor, prevented the HFD-induced weight gain, as well as the HFD-induced upregulation of ACBP/DBI, GABAAR γ2, and PPARγ. Based on these results, we postulate the existence of an obesogenic feedforward loop relying on ACBP/DBI, GABAAR, and PPARγ. Interruption of this vicious cycle, at any level, indistinguishably mitigates HFD-induced weight gain, hepatosteatosis, and hyperglycemia.
Collapse
Affiliation(s)
- Gerasimos Anagnostopoulos
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Omar Motiño
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Sijing Li
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Valentina Sica
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), Barcelona, Spain; Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Mélanie Bourgin
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Fanny Aprahamian
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Nitharsshini Nirmalathasan
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Romain Donne
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, 75006, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006, Paris, France
| | - Chantal Desdouets
- Laboratory of Proliferation, Stress and Liver Physiopathology, Centre de Recherche des Cordeliers, 75006, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006, Paris, France
| | | | - Konstantina Kotta
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Didier Surdez
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, SIREDO Oncology Centre, Institut Curie Research Centre, 75005, Paris, France
- Bone Sarcoma Research Laboratory, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Grossetête Sandrine
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, SIREDO Oncology Centre, Institut Curie Research Centre, 75005, Paris, France
| | - Olivier Delattre
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, SIREDO Oncology Centre, Institut Curie Research Centre, 75005, Paris, France
- Unité de Génétique Somatique, Service d'oncogénétique, Institut Curie, Centre Hospitalier, 75005, Paris, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - José Manuel Bravo-San Pedro
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Facultad de Medicina, Departamento de Fisiología, Universidad Complutense de Madrid, Madrid, Spain
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
33
|
Zipper L, Batchu S, Kaya NH, Antonello ZA, Reiff T. The MicroRNA miR-277 Controls Physiology and Pathology of the Adult Drosophila Midgut by Regulating the Expression of Fatty Acid β-Oxidation-Related Genes in Intestinal Stem Cells. Metabolites 2022; 12:315. [PMID: 35448502 PMCID: PMC9028014 DOI: 10.3390/metabo12040315] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Cell division, growth, and differentiation are energetically costly and dependent processes. In adult stem cell-based epithelia, cellular identity seems to be coupled with a cell's metabolic profile and vice versa. It is thus tempting to speculate that resident stem cells have a distinct metabolism, different from more committed progenitors and differentiated cells. Although investigated for many stem cell types in vitro, in vivo data of niche-residing stem cell metabolism is scarce. In adult epithelial tissues, stem cells, progenitor cells, and their progeny have very distinct functions and characteristics. In our study, we hypothesized and tested whether stem and progenitor cell types might have a distinctive metabolic profile in the intestinal lineage. Here, taking advantage of the genetically accessible adult Drosophila melanogaster intestine and the availability of ex vivo single cell sequencing data, we tested that hypothesis and investigated the metabolism of the intestinal lineage from stem cell (ISC) to differentiated epithelial cell in their native context under homeostatic conditions. Our initial in silico analysis of single cell RNAseq data and functional experiments identify the microRNA miR-277 as a posttranscriptional regulator of fatty acid β-oxidation (FAO) in the intestinal lineage. Low levels of miR-277 are detected in ISC and progressively rising miR-277 levels are found in progenitors during their growth and differentiation. Supporting this, miR-277-regulated fatty acid β-oxidation enzymes progressively declined from ISC towards more differentiated cells in our pseudotime single-cell RNAseq analysis and in functional assays on RNA and protein level. In addition, in silico clustering of single-cell RNAseq data based on metabolic genes validates that stem cells and progenitors belong to two independent clusters with well-defined metabolic characteristics. Furthermore, studying FAO genes in silico indicates that two populations of ISC exist that can be categorized in mitotically active and quiescent ISC, of which the latter relies on FAO genes. In line with an FAO dependency of ISC, forced expression of miR-277 phenocopies RNAi knockdown of FAO genes by reducing ISC size and subsequently resulting in stem cell death. We also investigated miR-277 effects on ISC in a benign and our newly developed CRISPR-Cas9-based colorectal cancer model and found effects on ISC survival, which as a consequence affects tumor growth, further underlining the importance of FAO in a pathological context. Taken together, our study provides new insights into the basal metabolic requirements of intestinal stem cell on β-oxidation of fatty acids evolutionarily implemented by a sole microRNA. Gaining knowledge about the metabolic differences and dependencies affecting the survival of two central and cancer-relevant cell populations in the fly and human intestine might reveal starting points for targeted combinatorial therapy in the hope for better treatment of colorectal cancer in the future.
Collapse
Affiliation(s)
- Lisa Zipper
- Institute of Genetics, Department of Biology, The Faculty of Mathematics and Natural Sciences, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany;
| | - Sai Batchu
- Cooper Medical School, Rowan University, Camden, NJ 08102, USA; (S.B.); (Z.A.A.)
| | - Nida Hatice Kaya
- Institute for Zoology and Organismic Interactions, Department of Biology, The Faculty of Mathematics and Natural Sciences, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany;
| | - Zeus Andrea Antonello
- Cooper Medical School, Rowan University, Camden, NJ 08102, USA; (S.B.); (Z.A.A.)
- Cooper University Hospital, Cooper University Health Care, Cooper Medical School, Rowan University, Camden, NJ 08102, USA
| | - Tobias Reiff
- Institute of Genetics, Department of Biology, The Faculty of Mathematics and Natural Sciences, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany;
| |
Collapse
|
34
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
35
|
Kamiyama T, Niwa R. Transcriptional Regulators of Ecdysteroid Biosynthetic Enzymes and Their Roles in Insect Development. Front Physiol 2022; 13:823418. [PMID: 35211033 PMCID: PMC8863297 DOI: 10.3389/fphys.2022.823418] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/12/2022] [Indexed: 12/23/2022] Open
Abstract
Steroid hormones are responsible for coordinating many aspects of biological processes in most multicellular organisms, including insects. Ecdysteroid, the principal insect steroid hormone, is biosynthesized from dietary cholesterol or plant sterols. In the last 20 years, a number of ecdysteroidogenic enzymes, including Noppera-bo, Neverland, Shroud, Spook/Spookier, Cyp6t3, Phantom, Disembodied, Shadow, and Shade, have been identified and characterized in molecular genetic studies using the fruit fly Drosophila melanogaster. These enzymes are encoded by genes collectively called the Halloween genes. The transcriptional regulatory network, governed by multiple regulators of transcription, chromatin remodeling, and endoreplication, has been shown to be essential for the spatiotemporal expression control of Halloween genes in D. melanogaster. In this review, we summarize the latest information on transcriptional regulators that are crucial for controlling the expression of ecdysteroid biosynthetic enzymes and their roles in insect development.
Collapse
Affiliation(s)
- Takumi Kamiyama
- College of Biological Sciences, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
36
|
Neophytou C, Pitsouli C. How Gut Microbes Nurture Intestinal Stem Cells: A Drosophila Perspective. Metabolites 2022; 12:169. [PMID: 35208243 PMCID: PMC8878600 DOI: 10.3390/metabo12020169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Host-microbiota interactions are key modulators of host physiology and behavior. Accumulating evidence suggests that the complex interplay between microbiota, diet and the intestine controls host health. Great emphasis has been given on how gut microbes have evolved to harvest energy from the diet to control energy balance, host metabolism and fitness. In addition, many metabolites essential for intestinal homeostasis are mainly derived from gut microbiota and can alleviate nutritional imbalances. However, due to the high complexity of the system, the molecular mechanisms that control host-microbiota mutualism, as well as whether and how microbiota affects host intestinal stem cells (ISCs) remain elusive. Drosophila encompasses a low complexity intestinal microbiome and has recently emerged as a system that might uncover evolutionarily conserved mechanisms of microbiota-derived nutrient ISC regulation. Here, we review recent studies using the Drosophila model that directly link microbiota-derived metabolites and ISC function. This research field provides exciting perspectives for putative future treatments of ISC-related diseases based on monitoring and manipulating intestinal microbiota.
Collapse
Affiliation(s)
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia, Nicosia 2109, Cyprus;
| |
Collapse
|
37
|
Abstract
In adult insects, as in vertebrates, the gut epithelium is a highly regenerative tissue that can renew itself rapidly in response to changing inputs from nutrition, the gut microbiota, ingested toxins, and signals from other organs. Because of its cellular and genetic similarities to the mammalian intestine, and its relevance as a target for the control of insect pests and disease vectors, many researchers have used insect intestines to address fundamental questions about stem cell functions during tissue maintenance and regeneration. In Drosophila, where most of the experimental work has been performed, not only are intestinal cell types and behaviors well characterized, but numerous cell signaling interactions have been detailed that mediate gut epithelial regeneration. A prevailing model for regenerative responses in the insect gut invokes stress sensing by damaged enterocytes (ECs) as a principal source for signaling that activates the division of intestinal stem cells (ISCs) and the growth and differentiation of their progeny. However, extant data also reveal alternative mechanisms for regeneration that involve ISC-intrinsic functions, active culling of healthy epithelial cells, enhanced EC growth, and even cytoplasmic shedding by infected ECs. This article reviews current knowledge of the molecular mechanisms involved in gut regeneration in several insect models (Drosophila and Aedes of the order Diptera, and several Lepidoptera).
Collapse
Affiliation(s)
- Peng Zhang
- Huntsman Cancer Institute, University of Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah 84112, USA
| | - Bruce A Edgar
- Huntsman Cancer Institute, University of Utah
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
38
|
Karam CS, Williams BL, Morozova I, Yuan Q, Panarsky R, Zhang Y, Hodgkinson CA, Goldman D, Kalachikov S, Javitch JA. Functional Genomic Analysis of Amphetamine Sensitivity in Drosophila. Front Psychiatry 2022; 13:831597. [PMID: 35250674 PMCID: PMC8894854 DOI: 10.3389/fpsyt.2022.831597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/24/2022] [Indexed: 11/14/2022] Open
Abstract
Abuse of psychostimulants, including amphetamines (AMPHs), is a major public health problem with profound psychiatric, medical, and psychosocial complications. The actions of these drugs at the dopamine transporter (DAT) play a critical role in their therapeutic efficacy as well as their liability for abuse and dependence. To date, however, the mechanisms that mediate these actions are not well-understood, and therapeutic interventions for AMPH abuse have been limited. Drug exposure can induce broad changes in gene expression that can contribute to neuroplasticity and effect long-lasting changes in neuronal function. Identifying genes and gene pathways perturbed by drug exposure is essential to our understanding of the molecular basis of drug addiction. In this study, we used Drosophila as a model to examine AMPH-induced transcriptional changes that are DAT-dependent, as those would be the most relevant to the stimulatory effects of the drug. Using this approach, we found genes involved in the control of mRNA translation to be significantly upregulated in response to AMPH in a DAT-dependent manner. To further prioritize genes for validation, we explored functional convergence between these genes and genes we identified in a genome-wide association study of AMPH sensitivity using the Drosophila Genetic Reference Panel. We validated a number of these genes by showing that they act specifically in dopamine neurons to mediate the behavioral effects of AMPH. Taken together, our data establish Drosophila as a powerful model that enables the integration of behavioral, genomic and transcriptomic data, followed by rapid gene validation, to investigate the molecular underpinnings of psychostimulant action.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Brenna L Williams
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Irina Morozova
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, United States.,Department of Chemical Engineering, Columbia University, New York, NY, United States
| | - Qiaoping Yuan
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Rony Panarsky
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Yuchao Zhang
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Colin A Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Sergey Kalachikov
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, United States.,Department of Chemical Engineering, Columbia University, New York, NY, United States
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States.,Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
39
|
Ketogenic diet reduces early mortality following traumatic brain injury in Drosophila via the PPARγ ortholog Eip75B. PLoS One 2021; 16:e0258873. [PMID: 34699541 PMCID: PMC8547619 DOI: 10.1371/journal.pone.0258873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/06/2021] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a common neurological disorder whose outcomes vary widely depending on a variety of environmental factors, including diet. Using a Drosophila melanogaster TBI model that reproduces key aspects of TBI in humans, we previously found that the diet consumed immediately following a primary brain injury has a substantial effect on the incidence of mortality within 24 h (early mortality). Flies that receive equivalent primary injuries have a higher incidence of early mortality when fed high-carbohydrate diets versus water. Here, we report that flies fed high-fat ketogenic diet (KD) following TBI exhibited early mortality that was equivalent to that of flies fed water and that flies protected from early mortality by KD continued to show survival benefits weeks later. KD also has beneficial effects in mammalian TBI models, indicating that the mechanism of action of KD is evolutionarily conserved. To probe the mechanism, we examined the effect of KD in flies mutant for Eip75B, an ortholog of the transcription factor PPARγ (peroxisome proliferator-activated receptor gamma) that contributes to the mechanism of action of KD and has neuroprotective effects in mammalian TBI models. We found that the incidence of early mortality of Eip75B mutant flies was higher when they were fed KD than when they were fed water following TBI. These data indicate that Eip75B/PPARγ is necessary for the beneficial effects of KD following TBI. In summary, this work provides the first evidence that KD activates PPARγ to reduce deleterious outcomes of TBI and it demonstrates the utility of the fly TBI model for dissecting molecular pathways that contribute to heterogeneity in TBI outcomes.
Collapse
|
40
|
Hung RJ, Li JSS, Liu Y, Perrimon N. Defining cell types and lineage in the Drosophila midgut using single cell transcriptomics. CURRENT OPINION IN INSECT SCIENCE 2021; 47:12-17. [PMID: 33609768 DOI: 10.1016/j.cois.2021.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
The Drosophila midgut has emerged in recent years as a model system to study stem cell renewal and differentiation and tissue homeostasis. Histological, genetic and gene expression studies have provided a wealth of information on gut cell types, regionalization, genes and pathways involved in cell proliferation and differentiation, stem cell renewal, and responses to changes in environmental factors such as the microbiota and nutrients. Here, we review the contribution of single cell transcriptomic methods to our understanding of gut cell type diversity, lineage and behavior.
Collapse
Affiliation(s)
- Ruei-Jiun Hung
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, United States
| | - Joshua Shing Shun Li
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, United States
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, United States
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, United States; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
41
|
Mank JE, Rideout EJ. Developmental mechanisms of sex differences: from cells to organisms. Development 2021; 148:272484. [PMID: 34647574 DOI: 10.1242/dev.199750] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Male-female differences in many developmental mechanisms lead to the formation of two morphologically and physiologically distinct sexes. Although this is expected for traits with prominent differences between the sexes, such as the gonads, sex-specific processes also contribute to traits without obvious male-female differences, such as the intestine. Here, we review sex differences in developmental mechanisms that operate at several levels of biological complexity - molecular, cellular, organ and organismal - and discuss how these differences influence organ formation, function and whole-body physiology. Together, the examples we highlight show that one simple way to gain a more accurate and comprehensive understanding of animal development is to include both sexes.
Collapse
Affiliation(s)
- Judith E Mank
- Department of Zoology, Biodiversity Research Centre, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada.,Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
42
|
Landis GN, Hilsabeck TAU, Bell HS, Ronnen-Oron T, Wang L, Doherty DV, Tejawinata FI, Erickson K, Vu W, Promislow DEL, Kapahi P, Tower J. Mifepristone Increases Life Span of Virgin Female Drosophila on Regular and High-fat Diet Without Reducing Food Intake. Front Genet 2021; 12:751647. [PMID: 34659367 PMCID: PMC8511958 DOI: 10.3389/fgene.2021.751647] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
Background: The synthetic steroid mifepristone is reported to have anti-obesity and anti-diabetic effects in mammals on normal and high-fat diets (HFD). We previously reported that mifepristone blocks the negative effect on life span caused by mating in female Drosophila melanogaster. Methods: Here we asked if mifepristone could protect virgin females from the life span-shortening effect of HFD. Mifepristone was assayed for effects on life span in virgin females, in repeated assays, on regular media and on media supplemented with coconut oil (HFD). The excrement quantification (EX-Q) assay was used to measure food intake of the flies after 12 days mifepristone treatment. In addition, experiments were conducted to compare the effects of mifepristone in virgin and mated females, and to identify candidate mifepristone targets and mechanisms. Results: Mifepristone increased life span of virgin females on regular media, as well as on media supplemented with either 2.5 or 5% coconut oil. Food intake was not reduced in any assay, and was significantly increased by mifepristone in half of the assays. To ask if mifepristone might rescue virgin females from all life span-shortening stresses, the oxidative stressor paraquat was tested, and mifepristone produced little to no rescue. Analysis of extant metabolomics and transcriptomics data suggested similarities between effects of mifepristone in virgin and mated females, including reduced tryptophan breakdown and similarities to dietary restriction. Bioinformatics analysis identified candidate mifepristone targets, including transcription factors Paired and Extra-extra. In addition to shortening life span, mating also causes midgut hypertrophy and activation of the lipid metabolism regulatory factor SREBP. Mifepristone blocked the increase in midgut size caused by mating, but did not detectably affect midgut size in virgins. Finally, mating increased activity of a SREBP reporter in abdominal tissues, as expected, but reporter activity was not detectably reduced by mifepristone in either mated or virgin females. Conclusion: Mifepristone increases life span of virgin females on regular and HFD without reducing food intake. Metabolomics and transcriptomics analyses suggest some similar effects of mifepristone between virgin and mated females, however reduced midgut size was observed only in mated females. The results are discussed regarding possible mifepristone mechanisms and targets.
Collapse
Affiliation(s)
- Gary N. Landis
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Tyler A. U. Hilsabeck
- Buck Institute for Research on Aging, Novato, CA, United States
- Davis School of Gerontology, University of Southern California, University Park, Los Angeles, CA, United States
| | - Hans S. Bell
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Tal Ronnen-Oron
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Devon V. Doherty
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Felicia I. Tejawinata
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Katherine Erickson
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - William Vu
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| | - Daniel E. L. Promislow
- Department of Biology, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA, United States
| | - John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
43
|
Dib A, Zanet J, Mancheno-Ferris A, Gallois M, Markus D, Valenti P, Marques-Prieto S, Plaza S, Kageyama Y, Chanut-Delalande H, Payre F. Pri smORF Peptides Are Wide Mediators of Ecdysone Signaling, Contributing to Shape Spatiotemporal Responses. Front Genet 2021; 12:714152. [PMID: 34527021 DOI: 10.3389/fgene.2021.714152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022] Open
Abstract
There is growing evidence that peptides encoded by small open-reading frames (sORF or smORF) can fulfill various cellular functions and define a novel class regulatory molecules. To which extend transcripts encoding only smORF peptides compare with canonical protein-coding genes, yet remain poorly understood. In particular, little is known on whether and how smORF-encoding RNAs might need tightly regulated expression within a given tissue, at a given time during development. We addressed these questions through the analysis of Drosophila polished rice (pri, a.k.a. tarsal less or mille pattes), which encodes four smORF peptides (11-32 amino acids in length) required at several stages of development. Previous work has shown that the expression of pri during epidermal development is regulated in the response to ecdysone, the major steroid hormone in insects. Here, we show that pri transcription is strongly upregulated by ecdysone across a large panel of cell types, suggesting that pri is a core component of ecdysone response. Although pri is produced as an intron-less short transcript (1.5 kb), genetic assays reveal that the developmental functions of pri require an unexpectedly large array of enhancers (spanning over 50 kb), driving a variety of spatiotemporal patterns of pri expression across developing tissues. Furthermore, we found that separate pri enhancers are directly activated by the ecdysone nuclear receptor (EcR) and display distinct regulatory modes between developmental tissues and/or stages. Alike major developmental genes, the expression of pri in a given tissue often involves several enhancers driving apparently redundant (or shadow) expression, while individual pri enhancers can harbor pleiotropic functions across tissues. Taken together, these data reveal the broad role of Pri smORF peptides in ecdysone signaling and show that the cis-regulatory architecture of the pri gene contributes to shape distinct spatial and temporal patterns of ecdysone response throughout development.
Collapse
Affiliation(s)
- Azza Dib
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Jennifer Zanet
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Alexandra Mancheno-Ferris
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Maylis Gallois
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Damien Markus
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Philippe Valenti
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Simon Marques-Prieto
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Serge Plaza
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - Yuji Kageyama
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Japan.,Biosignal Research Center, Kobe University, Kobe, Japan
| | - Hélène Chanut-Delalande
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| | - François Payre
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, University of Toulouse, Toulouse, France
| |
Collapse
|
44
|
Kim SK, Tsao DD, Suh GSB, Miguel-Aliaga I. Discovering signaling mechanisms governing metabolism and metabolic diseases with Drosophila. Cell Metab 2021; 33:1279-1292. [PMID: 34139200 PMCID: PMC8612010 DOI: 10.1016/j.cmet.2021.05.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022]
Abstract
There has been rapid growth in the use of Drosophila and other invertebrate systems to dissect mechanisms governing metabolism. New assays and approaches to physiology have aligned with superlative genetic tools in fruit flies to provide a powerful platform for posing new questions, or dissecting classical problems in metabolism and disease genetics. In multiple examples, these discoveries exploit experimental advantages as-yet unavailable in mammalian systems. Here, we illustrate how fly studies have addressed long-standing questions in three broad areas-inter-organ signaling through hormonal or neural mechanisms governing metabolism, intestinal interoception and feeding, and the cellular and signaling basis of sexually dimorphic metabolism and physiology-and how these findings relate to human (patho)physiology. The imaginative application of integrative physiology and related approaches in flies to questions in metabolism is expanding, and will be an engine of discovery, revealing paradigmatic features of metabolism underlying human diseases and physiological equipoise in health.
Collapse
Affiliation(s)
- Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine (Endocrinology), Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Deborah D Tsao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
45
|
Role of Endocrine System in the Regulation of Female Insect Reproduction. BIOLOGY 2021; 10:biology10070614. [PMID: 34356469 PMCID: PMC8301000 DOI: 10.3390/biology10070614] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022]
Abstract
The proper synthesis and functioning of ecdysteroids and juvenile hormones (JHs) are very important for the regulation of vitellogenesis and oogenesis. However, their role and function contrast among different orders, and even in the same insect order. For example, the JH is the main hormone that regulates vitellogenesis in hemimetabolous insect orders, which include Orthoptera, Blattodea, and Hemiptera, while ecdysteroids regulate the vitellogenesis among the insect orders of Diptera, some Hymenoptera and Lepidoptera. These endocrine hormones also regulate each other. Even at some specific stage of insect life, they positively regulate each other, while at other stages of insect life, they negatively control each other. Such positive and negative interaction of 20-hydroxyecdysone (20E) and JH is also discussed in this review article to better understand the role of these hormones in regulating the reproduction. Therefore, the purpose of the present review is to deeply understand the complex interaction of endocrine hormones with each other and with the insulin signaling pathway. The role of microbiomes in the regulation of the insect endocrine system is also reviewed, as the endocrine hormones are significantly affected by the compounds produced by the microbiota.
Collapse
|
46
|
Boumard B, Bardin AJ. An amuse-bouche of stem cell regulation: Underlying principles and mechanisms from adult Drosophila intestinal stem cells. Curr Opin Cell Biol 2021; 73:58-68. [PMID: 34217969 DOI: 10.1016/j.ceb.2021.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022]
Abstract
Stem cells have essential functions in the development and maintenance of our organs. Improper regulation of adult stem cells and tissue homeostasis can result in cancers and age-dependent decline. Therefore, understanding how tissue-specific stem cells can accurately renew tissues is an important aim of regenerative medicine. The Drosophila midgut harbors multipotent adult stem cells that are essential to renew the gut in homeostatic conditions and upon stress-induced regeneration. It is now a widely used model system to decipher regulatory mechanisms of stem cell biology. Here, we review recent findings on how adult intestinal stem cells differentiate, interact with their environment, and change during aging.
Collapse
Affiliation(s)
- Benjamin Boumard
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France
| | - Allison J Bardin
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.
| |
Collapse
|
47
|
Millington JW, Rideout EJ. Sexual Dimorphism: Ecdysone Modulates Sex Differences in the Gut. Curr Biol 2021; 30:R1327-R1330. [PMID: 33142105 DOI: 10.1016/j.cub.2020.08.088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sex differences in the Drosophila gut were discovered only recently. Recent work significantly extends our understanding of how steroid hormones specify these male-female differences by revealing a key role for ecdysone in regulating intestinal stem cell proliferation.
Collapse
Affiliation(s)
- Jason W Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
48
|
Sensitive High-Throughput Assays for Tumour Burden Reveal the Response of a Drosophila melanogaster Model of Colorectal Cancer to Standard Chemotherapies. Int J Mol Sci 2021; 22:ijms22105101. [PMID: 34065887 PMCID: PMC8151205 DOI: 10.3390/ijms22105101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 01/04/2023] Open
Abstract
Drosophila melanogaster (Drosophila) models of cancer are emerging as powerful tools to investigate the basic mechanisms underlying tumour progression and identify novel therapeutics. Rapid and inexpensive, it is possible to carry out genetic and drug screens at a far larger scale than in vertebrate organisms. Such whole-organism-based drug screens permits assessment of drug absorption and toxicity, reducing the possibility of false positives. Activating mutations in the Wnt and Ras signalling pathways are common in many epithelial cancers, and when driven in the adult Drosophila midgut, it induces aggressive intestinal tumour-like outgrowths that recapitulate many aspects of human colorectal cancer (CRC). Here we have taken a Drosophila CRC model in which tumourous cells are marked with both GFP and luciferase reporter genes, and developed novel high-throughput assays for quantifying tumour burden. Leveraging these assays, we find that the Drosophila CRC model responds rapidly to treatment with standard CRC-drugs, opening the door to future rapid genetic and drug screens.
Collapse
|
49
|
Al Hayek S, Alsawadi A, Kambris Z, Boquete J, Bohère J, Immarigeon C, Ronsin B, Plaza S, Lemaitre B, Payre F, Osman D. Steroid-dependent switch of OvoL/Shavenbaby controls self-renewal versus differentiation of intestinal stem cells. EMBO J 2021; 40:e104347. [PMID: 33372708 PMCID: PMC7883054 DOI: 10.15252/embj.2019104347] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/27/2022] Open
Abstract
Adult stem cells must continuously fine-tune their behavior to regenerate damaged organs and avoid tumors. While several signaling pathways are well known to regulate somatic stem cells, the underlying mechanisms remain largely unexplored. Here, we demonstrate a cell-intrinsic role for the OvoL family transcription factor, Shavenbaby (Svb), in balancing self-renewal and differentiation of Drosophila intestinal stem cells. We find that svb is a downstream target of Wnt and EGFR pathways, mediating their activity for stem cell survival and proliferation. This requires post-translational processing of Svb into a transcriptional activator, whose upregulation induces tumor-like stem cell hyperproliferation. In contrast, the unprocessed form of Svb acts as a repressor that imposes differentiation into enterocytes, and suppresses tumors induced by altered signaling. We show that the switch between Svb repressor and activator is triggered in response to systemic steroid hormone, which is produced by ovaries. Therefore, the Svb axis allows intrinsic integration of local signaling cues and inter-organ communication to adjust stem cell proliferation versus differentiation, suggesting a broad role of OvoL/Svb in adult and cancer stem cells.
Collapse
Affiliation(s)
- Sandy Al Hayek
- Faculty of Sciences IIILebanese UniversityTripoliLebanon
- Azm Center for Research in Biotechnology and its ApplicationsLBA3B, EDST, Lebanese UniversityTripoliLebanon
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
| | - Ahmad Alsawadi
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
| | - Zakaria Kambris
- Biology DepartmentFaculty of Arts and SciencesAmerican University of BeirutBeirutLebanon
| | | | - Jérôme Bohère
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
| | - Clément Immarigeon
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
| | - Brice Ronsin
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
| | - Serge Plaza
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
- Present address:
Laboratoire de Recherche en Sciences Végétales (LSRV)CNRSUPSCastanet‐TolosanFrance
| | - Bruno Lemaitre
- Global Health Institute, School of Life SciencesLausanneSwitzerland
| | - François Payre
- Centre de Biologie du Développement (CBD)Centre de Biologie Intégrative (CBI)Université de ToulouseCNRSToulouseFrance
| | - Dani Osman
- Faculty of Sciences IIILebanese UniversityTripoliLebanon
- Azm Center for Research in Biotechnology and its ApplicationsLBA3B, EDST, Lebanese UniversityTripoliLebanon
| |
Collapse
|
50
|
Hadjieconomou D, King G, Gaspar P, Mineo A, Blackie L, Ameku T, Studd C, de Mendoza A, Diao F, White BH, Brown AEX, Plaçais PY, Préat T, Miguel-Aliaga I. Author Correction: Enteric neurons increase maternal food intake during reproduction. Nature 2020; 588:E36. [PMID: 33303962 DOI: 10.1038/s41586-020-3013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Dafni Hadjieconomou
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - George King
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Pedro Gaspar
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Alessandro Mineo
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Laura Blackie
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Tomotsune Ameku
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Chris Studd
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Alex de Mendoza
- Australian Research Council Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Fengqiu Diao
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin H White
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - André E X Brown
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Pierre-Yves Plaçais
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Thomas Préat
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK.
- Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|