1
|
Ratiu AC, Ionascu A, Ecovoiu AA. A novel insertional allele of the CG18135 gene is associated with severe mutant phenotypes in Drosophila melanogaster. Front Genet 2024; 15:1355368. [PMID: 38957808 PMCID: PMC11217781 DOI: 10.3389/fgene.2024.1355368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
Drosophila melanogaster has been at the forefront of genetic studies and biochemical modeling for over a century. Yet, the functions of many genes are still unknown, mainly because no phenotypic data are available. Herein, we present the first evidence data regarding the particular molecular and other quantifiable phenotypes, such as viability and anatomical anomalies, induced by a novel P{lacW} insertional mutant allele of the CG18135 gene. So far, the CG18135 functions have only been theorized based on electronic annotation and presumptive associations inferred upon high-throughput proteomics or RNA sequencing experiments. The descendants of individuals harboring the CG18135 P{lacW}CG18135 allele were scored in order to assess mutant embryonic, larval, and pupal viability versus Canton Special (CantonS). Our results revealed that the homozygous CG18135 P{lacW}CG18135 /CG18135 P{lacW}CG18135 genotype determines significant lethality both at the inception of the larval stage and during pupal development. The very few imago escapers that either breach or fully exit the puparium exhibit specific eye depigmentation, wing abnormal unfolding, strong locomotor impairment with apparent spasmodic leg movements, and their maximum lifespan is shorter than 2 days. Using the quantitative real-time PCR (qRT-PCR) method, we found that CG18135 is upregulated in male flies, but an unexpected gene upregulation was also detected in heterozygous mutants compared to wild-type flies, probably because of regulatory perturbations induced by the P{lacW} transposon. Our work provides the first phenotypic evidence for the essential role of CG18135, a scenario in accordance with the putative role of this gene in carbohydrate-binding processes.
Collapse
Affiliation(s)
- Attila Cristian Ratiu
- Drosophila Laboratory, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Ilfov, Bucharest, Romania
| | - Adrian Ionascu
- Drosophila Laboratory, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Ilfov, Bucharest, Romania
| | - Alexandru Al. Ecovoiu
- Drosophila Laboratory, Faculty of Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
2
|
Ribeiro TDS, Lollar MJ, Sprengelmeyer QD, Huang Y, Benson DM, Orr MS, Johnson ZC, Corbett-Detig RB, Pool JE. Recombinant inbred line panels inform the genetic architecture and interactions of adaptive traits in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594228. [PMID: 38798433 PMCID: PMC11118405 DOI: 10.1101/2024.05.14.594228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The distribution of allelic effects on traits, along with their gene-by-gene and gene-by-environment interactions, contributes to the phenotypes available for selection and the trajectories of adaptive variants. Nonetheless, uncertainty persists regarding the effect sizes underlying adaptations and the importance of genetic interactions. Herein, we aimed to investigate the genetic architecture and the epistatic and environmental interactions involving loci that contribute to multiple adaptive traits using two new panels of Drosophila melanogaster recombinant inbred lines (RILs). To better fit our data, we re-implemented functions from R/qtl (Broman et al. 2003) using additive genetic models. We found 14 quantitative trait loci (QTL) underlying melanism, wing size, song pattern, and ethanol resistance. By combining our mapping results with population genetic statistics, we identified potential new genes related to these traits. None of the detected QTLs showed clear evidence of epistasis, and our power analysis indicated that we should have seen at least one significant interaction if sign epistasis or strong positive epistasis played a pervasive role in trait evolution. In contrast, we did find roles for gene-by-environment interactions involving pigmentation traits. Overall, our data suggest that the genetic architecture of adaptive traits often involves alleles of detectable effect, that strong epistasis does not always play a role in adaptation, and that environmental interactions can modulate the effect size of adaptive alleles.
Collapse
Affiliation(s)
- Tiago da Silva Ribeiro
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Matthew J. Lollar
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Yuheng Huang
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Derek M. Benson
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Megan S. Orr
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Zachary C. Johnson
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Russell B. Corbett-Detig
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, 95064, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, 95064, USA
| | - John E. Pool
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| |
Collapse
|
3
|
Williams-Simon PA, Oster C, Moaton JA, Ghidey R, Ng’oma E, Middleton KM, King EG. Naturally segregating genetic variants contribute to thermal tolerance in a Drosophila melanogaster model system. Genetics 2024; 227:iyae040. [PMID: 38506092 PMCID: PMC11075556 DOI: 10.1093/genetics/iyae040] [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: 07/11/2023] [Revised: 07/11/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
Thermal tolerance is a fundamental physiological complex trait for survival in many species. For example, everyday tasks such as foraging, finding a mate, and avoiding predation are highly dependent on how well an organism can tolerate extreme temperatures. Understanding the general architecture of the natural variants within the genes that control this trait is of high importance if we want to better comprehend thermal physiology. Here, we take a multipronged approach to further dissect the genetic architecture that controls thermal tolerance in natural populations using the Drosophila Synthetic Population Resource as a model system. First, we used quantitative genetics and Quantitative Trait Loci mapping to identify major effect regions within the genome that influences thermal tolerance, then integrated RNA-sequencing to identify differences in gene expression, and lastly, we used the RNAi system to (1) alter tissue-specific gene expression and (2) functionally validate our findings. This powerful integration of approaches not only allows for the identification of the genetic basis of thermal tolerance but also the physiology of thermal tolerance in a natural population, which ultimately elucidates thermal tolerance through a fitness-associated lens.
Collapse
Affiliation(s)
- Patricka A Williams-Simon
- Department of Biology, University of Pennsylvania, 433 S University Ave., 226 Leidy Laboratories, Philadelphia, PA 19104, USA
| | - Camille Oster
- Ash Creek Forest Management, 2796 SE 73rd Ave., Hillsboro, OR 97123, USA
| | | | - Ronel Ghidey
- ECHO Data Analysis Center, Johns Hopkins Bloomberg School of Public Health, 504 Cathedral St., Baltimore, MD 2120, USA
| | - Enoch Ng’oma
- Division of Biology, University of Missouri, 226 Tucker Hall, Columbia, MO 65211, USA
| | - Kevin M Middleton
- Division of Biology, University of Missouri, 222 Tucker Hall, Columbia, MO 65211, USA
| | - Elizabeth G King
- Division of Biology, University of Missouri, 401 Tucker Hall, Columbia, MO 65211, USA
| |
Collapse
|
4
|
Williams-Simon PA, Oster C, Moaton JA, Ghidey R, Ng'oma E, Middleton KM, Zars T, King EG. Naturally segregating genetic variants contribute to thermal tolerance in a D. melanogaster model system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.06.547110. [PMID: 37461510 PMCID: PMC10350013 DOI: 10.1101/2023.07.06.547110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Thermal tolerance is a fundamental physiological complex trait for survival in many species. For example, everyday tasks such as foraging, finding a mate, and avoiding predation, are highly dependent on how well an organism can tolerate extreme temperatures. Understanding the general architecture of the natural variants of the genes that control this trait is of high importance if we want to better comprehend how this trait evolves in natural populations. Here, we take a multipronged approach to further dissect the genetic architecture that controls thermal tolerance in natural populations using the Drosophila Synthetic Population Resource (DSPR) as a model system. First, we used quantitative genetics and Quantitative Trait Loci (QTL) mapping to identify major effect regions within the genome that influences thermal tolerance, then integrated RNA-sequencing to identify differences in gene expression, and lastly, we used the RNAi system to 1) alter tissue-specific gene expression and 2) functionally validate our findings. This powerful integration of approaches not only allows for the identification of the genetic basis of thermal tolerance but also the physiology of thermal tolerance in a natural population, which ultimately elucidates thermal tolerance through a fitness-associated lens.
Collapse
|
5
|
Sprengelmeyer QD, Lack JB, Braun DT, Monette MJ, Pool JE. The evolution of larger size in high-altitude Drosophila melanogaster has a variable genetic architecture. G3 GENES|GENOMES|GENETICS 2022; 12:6493269. [PMID: 35100377 PMCID: PMC8895999 DOI: 10.1093/g3journal/jkab454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 11/12/2022]
Abstract
Important uncertainties persist regarding the genetic architecture of adaptive trait evolution in natural populations, including the number of genetic variants involved, whether they are drawn from standing genetic variation, and whether directional selection drives them to complete fixation. Here, we take advantage of a unique natural population of Drosophila melanogaster from the Ethiopian highlands, which has evolved larger body size than any other known population of this species. We apply a bulk segregant quantitative trait locus mapping approach to 4 unique crosses between highland Ethiopian and lowland Zambian populations for both thorax length and wing length. Results indicated a persistently variable genetic basis for these evolved traits (with largely distinct sets of quantitative trait loci for each cross), and at least a moderately polygenic architecture with relatively strong effects present. We complemented these mapping experiments with population genetic analyses of quantitative trait locus regions and gene ontology enrichment analysis, generating strong hypotheses for specific genes and functional processes that may have contributed to these adaptive trait changes. Finally, we find that the genetic architectures indicated by our quantitative trait locus mapping results for size traits mirror those from similar experiments on other recently evolved traits in this species. Collectively, these studies suggest a recurring pattern of polygenic adaptation in this species, in which causative variants do not approach fixation and moderately strong effect loci are present.
Collapse
Affiliation(s)
| | - Justin B Lack
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dylan T Braun
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Matthew J Monette
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - John E Pool
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
6
|
López-Varea A, Vega-Cuesta P, Ruiz-Gómez A, Ostalé CM, Molnar C, Hevia CF, Martín M, Organista MF, de Celis J, Culí J, Esteban N, de Celis JF. Genome-wide phenotypic RNAi screen in the Drosophila wing: phenotypic description of functional classes. G3 (BETHESDA, MD.) 2021; 11:6380434. [PMID: 34599810 PMCID: PMC8664486 DOI: 10.1093/g3journal/jkab349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 09/23/2021] [Indexed: 01/01/2023]
Abstract
The Drosophila genome contains approximately 14,000 protein-coding genes encoding all the necessary information to sustain cellular physiology, tissue organization, organism development, and behavior. In this manuscript, we describe in some detail the phenotypes in the adult fly wing generated after knockdown of approximately 80% of Drosophila genes. We combined this phenotypic description with a comprehensive molecular classification of the Drosophila proteins into classes that summarize the main expected or known biochemical/functional aspect of each protein. This information, combined with mRNA expression levels and in situ expression patterns, provides a simplified atlas of the Drosophila genome, from housekeeping proteins to the components of the signaling pathways directing wing development, that might help to further understand the contribution of each gene group to wing formation.
Collapse
Affiliation(s)
- Ana López-Varea
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Patricia Vega-Cuesta
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ana Ruiz-Gómez
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina M Ostalé
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina Molnar
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain.,IRB Barcelona, Barcelona 08028, Spain
| | - Covadonga F Hevia
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Mercedes Martín
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Maria F Organista
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jesus de Celis
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Joaquín Culí
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Nuria Esteban
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jose F de Celis
- Centro de Biología Molecular "Severo Ochoa," CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
7
|
López-Varea A, Ostalé CM, Vega-Cuesta P, Ruiz-Gómez A, Organista MF, Martín M, Hevia CF, Molnar C, de Celis J, Culi J, Esteban N, de Celis JF. Genome-wide Phenotypic RNAi Screen in the Drosophila Wing: Global Parameters. G3-GENES GENOMES GENETICS 2021; 11:6380435. [PMID: 34599819 PMCID: PMC8962446 DOI: 10.1093/g3journal/jkab351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022]
Abstract
We have screened a collection of UAS-RNAi lines targeting 10,920 Drosophila protein-coding genes for phenotypes in the adult wing. We identified 3653 genes (33%) whose knockdown causes either larval/pupal lethality or a mutant phenotype affecting the formation of a normal wing. The most frequent phenotypes consist of changes in wing size, vein differentiation, and patterning, defects in the wing margin and in the apposition of the dorsal and ventral wing surfaces. We also defined 16 functional categories encompassing the most relevant aspect of each protein function and assigned each Drosophila gene to one of these functional groups. This allowed us to identify which mutant phenotypes are enriched within each functional group. Finally, we used previously published gene expression datasets to determine which genes are or are not expressed in the wing disc. Integrating expression, phenotypic and molecular information offers considerable precision to identify the relevant genes affecting wing formation and the biological processes regulated by them.
Collapse
Affiliation(s)
- Ana López-Varea
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina M Ostalé
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Patricia Vega-Cuesta
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ana Ruiz-Gómez
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - María F Organista
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Mercedes Martín
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Covadonga F Hevia
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina Molnar
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jesús de Celis
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Joaquim Culi
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Nuria Esteban
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jose F de Celis
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
8
|
Sugita A, Kuruma S, Yanagisawa N, Ishiguro H, Kano R, Ohkuma Y, Hirose Y. The cap-specific m6A methyltransferase, PCIF1/CAPAM, is dynamically recruited to the gene promoter in a transcription-dependent manner. J Biochem 2021; 170:203-213. [PMID: 33982754 DOI: 10.1093/jb/mvab032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 03/15/2021] [Indexed: 11/12/2022] Open
Abstract
N 6-methyladenosine (m6A), the most abundant modification in eukaryotic mRNAs, plays an important role in mRNA metabolism and functions. When adenosine is transcribed as the first cap-adjacent nucleotide, it is methylated at the ribose 2'-O and N6 positions, thus generating N6, 2'-O-dimethyladenosine (m6Am). Phosphorylated C-terminal domain (CTD)-interacting factor 1 (PCIF1) is a novel cap-specific adenine N6-methyltransferase responsible for m6Am formation. As PCIF1 specifically interacts with the Ser5-phosphorylated CTD of RNA polymerase II (Pol II), which is a marker for the early phase of transcription, PCIF1 is speculated to be recruited to the early elongating Pol II. In this study, subcellular fractionation and immunofluorescence microscopy demonstrated that PCIF1 is mainly localized to the transcriptionally active chromatin regions in HeLa cells. Chromatin immunoprecipitation (ChIP) revealed that PCIF1 was predominantly localized to the promoter of a broad range of Pol II-transcribed genes, including several protein-coding genes and non-coding RNA genes. Moreover, PCIF1 accumulation on these promoters depended entirely on transcriptional activity and Ser5 phosphorylation of the CTD. These results suggest that PCIF1 dynamically localizes to the Pol II early in transcription and may efficiently catalyze N6-methylation of the first adenosine residue of nascent mRNAs cotranscriptionally.
Collapse
Affiliation(s)
- Ai Sugita
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Soichiro Kuruma
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Natsuki Yanagisawa
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Hiroyasu Ishiguro
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Ryoya Kano
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Yoshiaki Ohkuma
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.,Department of Biochemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yutaka Hirose
- Laboratory of Gene Regulation, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
9
|
Moutaoufik MT, Tanguay RM. Analysis of insect nuclear small heat shock proteins and interacting proteins. Cell Stress Chaperones 2021; 26:265-274. [PMID: 32888179 PMCID: PMC7736433 DOI: 10.1007/s12192-020-01156-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022] Open
Abstract
The small heat shock proteins (sHsps) are a ubiquitous family of ATP-independent stress proteins found in all domains of life. Drosophila melanogaster Hsp27 (DmHsp27) is the only known nuclear sHsp in insect. Here analyzing sequences from HMMER, we identified 56 additional insect sHsps with conserved arginine-rich nuclear localization signal (NLS) in the N-terminal region. At this time, the exact role of nuclear sHsps remains unknown. DmHsp27 protein-protein interaction analysis from iRefIndex database suggests that this protein, in addition to a putative role of molecular chaperone, is likely involved in other nuclear processes (i.e., chromatin remodeling and transcription). Identification of DmHsp27 interactors should provide key insights on the cellular and molecular functions of this nuclear chaperone.
Collapse
Affiliation(s)
- Mohamed Taha Moutaoufik
- Lab of Cell & Developmental Genetics, Department of Cellular and Molecular Biology, Medical Biochemistry & Pathology, Medical School, Université Laval, Quebec, G1K 7P4, Canada
- Department of Biochemistry, University of Regina, Regina, SK, S4S 0A2, Canada
| | - Robert M Tanguay
- Lab of Cell & Developmental Genetics, Department of Cellular and Molecular Biology, Medical Biochemistry & Pathology, Medical School, Université Laval, Quebec, G1K 7P4, Canada.
| |
Collapse
|
10
|
Rivera-Colón AG, Westerman EL, Van Belleghem SM, Monteiro A, Papa R. Multiple Loci Control Eyespot Number Variation on the Hindwings of Bicyclus anynana Butterflies. Genetics 2020; 214:1059-1078. [PMID: 32019848 PMCID: PMC7153931 DOI: 10.1534/genetics.120.303059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/26/2020] [Indexed: 11/18/2022] Open
Abstract
The underlying genetic changes that regulate the appearance and disappearance of repeated traits, or serial homologs, remain poorly understood. One hypothesis is that variation in genomic regions flanking master regulatory genes, also known as input-output genes, controls variation in trait number, making the locus of evolution almost predictable. Another hypothesis implicates genetic variation in up- or downstream loci of master control genes. Here, we use the butterfly Bicyclus anynana, a species that exhibits natural variation in eyespot number on the dorsal hindwing, to test these two hypotheses. We first estimated the heritability of dorsal hindwing eyespot number by breeding multiple butterfly families differing in eyespot number and regressing eyespot numbers of offspring on midparent values. We then estimated the number and identity of independent genetic loci contributing to eyespot number variation by performing a genome-wide association study with restriction site-associated DNA sequencing from multiple individuals varying in number of eyespots sampled across a freely breeding laboratory population. We found that dorsal hindwing eyespot number has a moderately high heritability of ∼0.50 and is characterized by a polygenic architecture. Previously identified genomic regions involved in eyespot development, and novel ones, display high association with dorsal hindwing eyespot number, suggesting that homolog number variation is likely determined by regulatory changes at multiple loci that build the trait, and not by variation at single master regulators or input-output genes.
Collapse
Affiliation(s)
- Angel G Rivera-Colón
- Department of Evolution, Ecology, and Behavior, University of Illinois, Urbana-Champaign, Illinois 61801
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, 00925, Puerto Rico
| | - Erica L Westerman
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701
| | - Steven M Van Belleghem
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, 00925, Puerto Rico
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, Singapore 117543
- Yale-NUS College, Singapore 138609
| | - Riccardo Papa
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, 00925, Puerto Rico
- Molecular Sciences and Research Center, University of Puerto Rico, San Juan, 00926, Puerto Rico
| |
Collapse
|
11
|
Draper I, Saha M, Stonebreaker H, Salomon RN, Matin B, Kang PB. The impact of Megf10/Drpr gain-of-function on muscle development in Drosophila. FEBS Lett 2019; 593:680-696. [PMID: 30802937 DOI: 10.1002/1873-3468.13348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 11/07/2022]
Abstract
Recessive mutations in multiple epidermal growth factor-like domains 10 (MEGF10) underlie a rare congenital muscle disease known as MEGF10 myopathy. MEGF10 and its Drosophila homolog Draper (Drpr) are transmembrane receptors expressed in muscle and glia. Drpr deficiency is known to result in muscle abnormalities in flies. In the current study, flies that ubiquitously overexpress Drpr, or mouse Megf10, display developmental arrest. The phenotype is reproduced with overexpression in muscle, but not in other tissues, and with overexpression during intermediate stages of myogenesis, but not in myoblasts. We find that tubular muscle subtypes are particularly sensitive to Megf10/Drpr overexpression. Complementary genetic analyses show that Megf10/Drpr and Notch may interact to regulate myogenesis. Our findings provide a basis for investigating MEGF10 in muscle development using Drosophila.
Collapse
Affiliation(s)
- Isabelle Draper
- Department of Medicine, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Madhurima Saha
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Robert N Salomon
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA, USA
| | - Bahar Matin
- Department of Medicine, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Department of Neurology, Boston Children's Hospital, MA, USA.,Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA.,Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA.,Genetics Institute and Myology Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
12
|
Facultative dosage compensation of developmental genes on autosomes in Drosophila and mouse embryonic stem cells. Nat Commun 2018; 9:3626. [PMID: 30194291 PMCID: PMC6128902 DOI: 10.1038/s41467-018-05642-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 07/04/2018] [Indexed: 12/25/2022] Open
Abstract
Haploinsufficiency and aneuploidy are two phenomena, where gene dosage alterations cause severe defects ultimately resulting in developmental failures and disease. One remarkable exception is the X chromosome, where copy number differences between sexes are buffered by dosage compensation systems. In Drosophila, the Male-Specific Lethal complex (MSLc) mediates upregulation of the single male X chromosome. The evolutionary origin and conservation of this process orchestrated by MSL2, the only male-specific protein within the fly MSLc, have remained unclear. Here, we report that MSL2, in addition to regulating the X chromosome, targets autosomal genes involved in patterning and morphogenesis. Precise regulation of these genes by MSL2 is required for proper development. This set of dosage-sensitive genes maintains such regulation during evolution, as MSL2 binds and similarly regulates mouse orthologues via Histone H4 lysine 16 acetylation. We propose that this gene-by-gene dosage compensation mechanism was co-opted during evolution for chromosome-wide regulation of the Drosophila male X. In Drosophila the Male-Specific Lethal complex (MSLc) mediates upregulation of the single male X chromosome. Here the authors provide evidence that MSL2 also targets autosomal genes required for proper development and that MSL2 binds and similarly regulates mouse orthologues.
Collapse
|
13
|
Kairamkonda S, Nongthomba U. Beadex, a Drosophila LIM domain only protein, function in follicle cells is essential for egg development and fertility. Exp Cell Res 2018; 367:97-103. [PMID: 29580687 DOI: 10.1016/j.yexcr.2018.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 11/25/2022]
Abstract
LIM domain, constituted by two tandem C2H2 zinc finger motif, proteins regulate several biological processes. They are usually found associated with various functional domains like Homeodomain, kinase domain and other protein binding domains. LIM proteins that are devoid of other domains are called LIM only proteins (LMO). LMO proteins were first identified in humans and are implicated in development and oncogenesis. They regulate various cell specifications by regulating the activity of respective transcriptional complexes. The Drosophila LMO protein (dLMO), Beadex (Bx), regulates various developmental processes like wing margin development and bristle development. It also regulates Drosophila behavior in response to cocaine and ethanol. We have previously generated Bx null flies and shown its essential function in neurons for multiple aspects of female reproduction. However, it was not known whether Bx affects reproduction through its independent function in ovaries. In this paper we show that female flies null for Bx lay eggs with multiple defects. Further, through knock down studies we demonstrate that function of Bx in follicle cells is required for normal egg development. We also show that function of Bx is particularly required in border cells for Drosophila fertility.
Collapse
Affiliation(s)
- Subhash Kairamkonda
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Upendra Nongthomba
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
14
|
Fernandes KM, de Magalhães-Júnior MJ, Baracat-Pereira MC, Martins GF. Proteomic analysis of Aedes aegypti midgut during post-embryonic development and of the female mosquitoes fed different diets. Parasitol Int 2016; 65:668-676. [PMID: 27597118 DOI: 10.1016/j.parint.2016.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 08/26/2016] [Accepted: 08/27/2016] [Indexed: 12/30/2022]
Abstract
In this work we analyzed protein expression in the Aedes aegypti midgut during the larval (fourth instar, L4), pupal, and adult stages [including newly emerged (NE), sugar-fed (SF) and blood-fed (BF) females]. Two-dimensional electrophoresis showed 13 spots in the midgut of larvae, 95 in the midgut of pupae, 90 in the midgut of NE, and 76 in the midgut of SF or BF females. In the larval midguts, high serpin expression was noted, while in the pupae, protein abundance was lower than in the NE, SF, and BF females. The spots related to proteins linked to energy production, protein metabolism, signaling, and transport were highly expressed in the NE stage, while spots related proteins involved in translation were abundant in SF and BF females. The differential abundance of proteins in the midgut of A. aegypti at different developmental stages supports the necessity for midgut development during immature stage followed by the necessity of proteins related to digestion in adults.
Collapse
Affiliation(s)
- Kenner Morais Fernandes
- Departamento de Biologia Geral, Universidade Federal de Viçosa - UFV, 36571-900 Viçosa, Minas Gerais, Brazil
| | | | - Maria Cristina Baracat-Pereira
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa - UFV, 36571-900 Viçosa, Minas Gerais, Brazil
| | - Gustavo Ferreira Martins
- Departamento de Biologia Geral, Universidade Federal de Viçosa - UFV, 36571-900 Viçosa, Minas Gerais, Brazil.
| |
Collapse
|
15
|
Viability, longevity, and egg production of Drosophila melanogaster are regulated by the miR-282 microRNA. Genetics 2013; 195:469-80. [PMID: 23852386 DOI: 10.1534/genetics.113.153585] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The first microRNAs were discovered some 20 years ago, but only a small fraction of the microRNA-encoding genes have been described in detail yet. Here we report the molecular analysis of a computationally predicted Drosophila melanogaster microRNA gene, mir-282. We show that the mir-282 gene is the source of a 4.9-kb-long primary transcript with a 5' cap and a 3'-poly(A) sequence and a mature microRNA of ∼25 bp. Our data strongly suggest the existence of an independent mir-282 gene conserved in holometabolic insects. We give evidence that the mir-282 locus encodes a functional transcript that influences viability, longevity, and egg production in Drosophila. We identify the nervous system-specific adenylate cyclase (rutabaga) as a target of miR-282 and assume that one of the main functions of mir-282 is the regulation of adenylate cyclase activity in the nervous system during metamorphosis.
Collapse
|
16
|
A targeted genetic modifier screen links the SWI2/SNF2 protein domino to growth and autophagy genes in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2013; 3:815-25. [PMID: 23550128 PMCID: PMC3656729 DOI: 10.1534/g3.112.005496] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Targeted genetic studies can facilitate phenotypic analyses and provide important insights into development and other complex processes. The SWI2/SNF2 DNA-dependent ATPase Domino (Dom) of Drosophila melanogaster, a component of the Tip60 acetyltransferase complex, has been associated with a wide spectrum of cellular processes at multiple developmental stages. These include hematopoiesis, cell proliferation, homeotic gene regulation, histone exchange during DNA repair, and Notch signaling. To explore the wider gene network associated with Dom action, we used RNAi directed against domino (dom) to mediate loss-of-function at the wing margin, a tissue that is readily scored for phenotypic changes. Dom RNAi driven through GAL4-UAS elicited dominant wing nicking that responded phenotypically to the dose of dom and other loci known to function with dom. We screened for phenotypic modifiers of this wing phenotype among 2500 transpositions of the EP P element and found both enhancers and suppressors. Several classes of modifier were obtained, including those encoding transcription factors, RNA regulatory proteins, and factors that regulate cell growth, proliferation and autophagy, a lysosomal degradation pathway that affects cell growth under conditions of starvation and stress. Our analysis is consistent with prior studies, suggesting that Dom acts pleiotropically as a positive effector of Notch signaling and a repressor of proliferation. This genetic system should facilitate screens for additional loci associated with Dom function, and complement biochemical approaches to their regulatory activity.
Collapse
|
17
|
Swaminathan A, Barnes VL, Fox S, Gammouh S, Pile LA. Identification of genetic suppressors of the Sin3A knockdown wing phenotype. PLoS One 2012; 7:e49563. [PMID: 23166712 PMCID: PMC3499482 DOI: 10.1371/journal.pone.0049563] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 10/11/2012] [Indexed: 11/19/2022] Open
Abstract
The role of the Sin3A transcriptional corepressor in regulating the cell cycle is established in various metazoans. Little is known, however, about the signaling pathways that trigger or are triggered by Sin3A function. To discover genes that work in similar or opposing pathways to Sin3A during development, we have performed an unbiased screen of deficiencies of the Drosophila third chromosome. Additionally, we have performed a targeted loss of function screen to identify cell cycle genes that genetically interact with Sin3A. We have identified genes that encode proteins involved in regulation of gene expression, signaling pathways and cell cycle that can suppress the curved wing phenotype caused by the knockdown of Sin3A. These data indicate that Sin3A function is quite diverse and impacts a wide variety of cellular processes.
Collapse
Affiliation(s)
- Aishwarya Swaminathan
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Valerie L. Barnes
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Stephanie Fox
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Sarah Gammouh
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Lori A. Pile
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
18
|
Mutual repression by bantam miRNA and Capicua links the EGFR/MAPK and Hippo pathways in growth control. Curr Biol 2012; 22:651-7. [PMID: 22445297 DOI: 10.1016/j.cub.2012.02.050] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/07/2012] [Accepted: 02/17/2012] [Indexed: 01/01/2023]
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) and Hippo signaling pathways control cell proliferation and apoptosis to promote tissue growth during development. Misregulation of these pathways is implicated in cancer. Our understanding of the mechanisms that integrate the activity of these pathways remains fragmentary. This study identifies bantam microRNA as a common target of these pathways and suggests a mechanistic link between them. RESULTS The EGFR pathway acts through bantam to control tissue growth. bantam expression is regulated by the EGFR pathway, acting via repression of the transcriptional repressor Capicua. Thus EGFR signaling induces bantam expression by alleviating the effects of a repressor. bantam in turn acts in a negative feedback loop to limit Capicua expression. CONCLUSIONS bantam appears to be a transcriptional target of both the EGFR and Hippo growth control pathways. Feedback regulation by bantam on Capicua provides a means to link signal propagation by the EGFR pathway to activity of the Hippo pathway and may play an important role in integration of these two pathways in growth control.
Collapse
|
19
|
Hainaut M, Sagnier T, Berenger H, Pradel J, Graba Y, Miotto B. The MYST-containing protein Chameau is required for proper sensory organ specification during Drosophila thorax morphogenesis. PLoS One 2012; 7:e32882. [PMID: 22412942 PMCID: PMC3295779 DOI: 10.1371/journal.pone.0032882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 02/04/2012] [Indexed: 12/15/2022] Open
Abstract
The adult thorax of Drosophila melanogaster is covered by a stereotyped pattern of mechanosensory bristles called macrochaetes. Here, we report that the MYST containing protein Chameau (Chm) contributes to the establishment of this pattern in the most dorsal part of the thorax. Chm mutant pupae present extra-dorsocentral (DC) and scutellar (SC) macrochaetes, but a normal number of the other macrochaetes. We provide evidences that chm restricts the singling out of sensory organ precursors from proneural clusters and genetically interacts with transcriptional regulators involved in the regulation of achaete and scute in the DC and SC proneural cluster. This function of chm likely relies on chromatin structure regulation since a protein with a mutation in the conserved catalytic site fails to rescue the formation of supernumerary DC and SC bristles in chm mutant flies. This is further supported by the finding that mutations in genes encoding chromatin modifiers and remodeling factors, including Polycomb group (PcG) and Trithorax group (TrxG) members, dominantly modulate the penetrance of chm extra bristle phenotype. These data support a critical role for chromatin structure modulation in the establishment of the stereotyped sensory bristle pattern in the fly thorax.
Collapse
Affiliation(s)
- Matthieu Hainaut
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR6216/Université de la Méditerranée, Marseille, France
| | - Thierry Sagnier
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR6216/Université de la Méditerranée, Marseille, France
| | - Hélène Berenger
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR6216/Université de la Méditerranée, Marseille, France
| | - Jacques Pradel
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR6216/Université de la Méditerranée, Marseille, France
| | - Yacine Graba
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR6216/Université de la Méditerranée, Marseille, France
- * E-mail: (YG); (BM)
| | - Benoit Miotto
- Institut de Biologie du Développement de Marseille-Luminy, CNRS UMR6216/Université de la Méditerranée, Marseille, France
- * E-mail: (YG); (BM)
| |
Collapse
|
20
|
Abstract
Although a great deal is known about the identity, biogenesis, and targeting capacity of microRNAs (miRNAs) in animal cells, far less is known about their functional requirements at the organismal level. Much remains to be understood about the necessity of miRNAs for overt phenotypes, the identity of critical miRNA targets, and the control of miRNA transcription. In this review, we provide an overview of genetic strategies to study miRNAs in the Drosophila system, including loss- and gain-of-function techniques, genetic interaction strategies, and transgenic reporters of miRNA expression and activity. As we illustrate the usage of these techniques in intact Drosophila, we see certain recurrent themes for miRNA functions, including energy homeostasis, apoptosis suppression, growth control, and regulation of core cell signaling pathways. Overall, we hope that this exposition of Drosophila genetic techniques, well known to the legions of fly geneticists and used to study all genes, can inform the general miRNA community that focuses on other biochemical, molecular, computational, and structural avenues. Clearly, it is the combination of these myriad techniques that has accelerated miRNA research to its extraordinary pace.
Collapse
Affiliation(s)
- Qi Dai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York NY 10065
| | - Peter Smibert
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York NY 10065
| | - Eric C. Lai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York NY 10065
| |
Collapse
|
21
|
Monier B, Pélissier-Monier A, Sanson B. Establishment and maintenance of compartmental boundaries: role of contractile actomyosin barriers. Cell Mol Life Sci 2011; 68:1897-910. [PMID: 21437644 PMCID: PMC11114499 DOI: 10.1007/s00018-011-0668-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 02/12/2011] [Accepted: 03/08/2011] [Indexed: 12/29/2022]
Abstract
During animal development, tissues and organs are partitioned into compartments that do not intermix. This organizing principle is essential for correct tissue morphogenesis. Given that cell sorting defects during compartmentalization in humans are thought to cause malignant invasion and congenital defects such as cranio-fronto-nasal syndrome, identifying the molecular and cellular mechanisms that keep cells apart at boundaries between compartments is important. In both vertebrates and invertebrates, transcription factors and short-range signalling pathways, such as EPH/Ephrin, Hedgehog, or Notch signalling, govern compartmental cell sorting. However, the mechanisms that mediate cell sorting downstream of these factors have remained elusive for decades. Here, we review recent data gathered in Drosophila that suggest that the generation of cortical tensile forces at compartmental boundaries by the actomyosin cytoskeleton could be a general mechanism that inhibits cell mixing between compartments.
Collapse
Affiliation(s)
- Bruno Monier
- Department of Physiology, Development and Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY UK
- Present Address: Institut de Biologie du Développement de Marseille-Luminy, UMR6216, Campus de Luminy, Case 907, 13288 Marseille cedex 9, France
| | - Anne Pélissier-Monier
- Department of Physiology, Development and Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY UK
- Wellcome Trust, Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN UK
- Present Address: Institut de Biologie du Développement de Marseille-Luminy, UMR6216, Campus de Luminy, Case 907, 13288 Marseille cedex 9, France
| | - Bénédicte Sanson
- Department of Physiology, Development and Neuroscience, Anatomy Building, Downing Street, Cambridge, CB2 3DY UK
| |
Collapse
|
22
|
Guan Z, Buhl LK, Quinn WG, Littleton JT. Altered gene regulation and synaptic morphology in Drosophila learning and memory mutants. Learn Mem 2011; 18:191-206. [PMID: 21422168 DOI: 10.1101/lm.2027111] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Genetic studies in Drosophila have revealed two separable long-term memory pathways defined as anesthesia-resistant memory (ARM) and long-lasting long-term memory (LLTM). ARM is disrupted in radish (rsh) mutants, whereas LLTM requires CREB-dependent protein synthesis. Although the downstream effectors of ARM and LLTM are distinct, pathways leading to these forms of memory may share the cAMP cascade critical for associative learning. Dunce, which encodes a cAMP-specific phosphodiesterase, and rutabaga, which encodes an adenylyl cyclase, both disrupt short-term memory. Amnesiac encodes a pituitary adenylyl cyclase-activating peptide homolog and is required for middle-term memory. Here, we demonstrate that the Radish protein localizes to the cytoplasm and nucleus and is a PKA phosphorylation target in vitro. To characterize how these plasticity pathways may manifest at the synaptic level, we assayed synaptic connectivity and performed an expression analysis to detect altered transcriptional networks in rutabaga, dunce, amnesiac, and radish mutants. All four mutants disrupt specific aspects of synaptic connectivity at larval neuromuscular junctions (NMJs). Genome-wide DNA microarray analysis revealed ∼375 transcripts that are altered in these mutants, suggesting defects in multiple neuronal signaling pathways. In particular, the transcriptional target Lapsyn, which encodes a leucine-rich repeat cell adhesion protein, localizes to synapses and regulates synaptic growth. This analysis provides insights into the Radish-dependent ARM pathway and novel transcriptional targets that may contribute to memory processing in Drosophila.
Collapse
Affiliation(s)
- Zhuo Guan
- Department of Biology, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
23
|
Enderle D, Beisel C, Stadler MB, Gerstung M, Athri P, Paro R. Polycomb preferentially targets stalled promoters of coding and noncoding transcripts. Genome Res 2010; 21:216-26. [PMID: 21177970 DOI: 10.1101/gr.114348.110] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Polycomb group (PcG) and Trithorax group (TrxG) of proteins are required for stable and heritable maintenance of repressed and active gene expression states. Their antagonistic function on gene control, repression for PcG and activity for TrxG, is mediated by binding to chromatin and subsequent epigenetic modification of target loci. Despite our broad knowledge about composition and enzymatic activities of the protein complexes involved, our understanding still lacks important mechanistic detail and a comprehensive view on target genes. In this study we use an extensive data set of ChIP-seq, RNA-seq, and genome-wide detection of transcription start sites (TSSs) to identify and analyze thousands of binding sites for the PcG proteins and Trithorax from a Drosophila S2 cell line. In addition of finding a preference for stalled promoter regions of annotated genes, we uncover many intergenic PcG binding sites coinciding with nonannotated TSSs. Interestingly, this set includes previously unknown promoters for primary transcripts of microRNA genes, thereby expanding the scope of Polycomb control to noncoding RNAs essential for development, apoptosis, and growth.
Collapse
Affiliation(s)
- Daniel Enderle
- Department of Biosystems Science and Engineering, ETH Zurich, CH-4058 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
24
|
Fernandez-Valverde SL, Taft RJ, Mattick JS. Dynamic isomiR regulation in Drosophila development. RNA (NEW YORK, N.Y.) 2010; 16:1881-8. [PMID: 20805289 PMCID: PMC2941097 DOI: 10.1261/rna.2379610] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 07/21/2010] [Indexed: 05/19/2023]
Abstract
Several recent reports have demonstrated that microRNAs (miRNAs) can exhibit heterogeneous ends and post-transcriptional nontemplate 3' end additions of uridines or adenosines. Using two small RNA deep-sequencing data sets, we show here that these miRNA isoforms (isomiRs) are differentially expressed across Drosophila melanogaster development and tissues. Specifically, we demonstrate that: (1) nontemplate nucleotide additions of adenosines to miRNA 3' ends are highly abundant in early development; (2) a subset of miRNAs with nontemplate 3' Us are expressed in adult tissues; and (3) the size of at least eight "mature" (unmodified) miRNAs varies in a life-cycle or tissue-specific manner. These results suggest that subtle variability in isomiR expression, which is widely thought to be the result of inexact Dicer processing, is regulated and biologically meaningful. Indeed, a subset of the miRNAs enriched for 3' adenosine additions during early embryonic development, including miR-282 and miR-312, show enrichment for target sites in developmental genes that are expressed during late embryogenesis, suggesting that nontemplate additions increase miRNA stability or strengthen miRNA:target interactions. This work suggests that isomiR expression is an important aspect of miRNA biology, which warrants further investigation.
Collapse
|
25
|
Bronstein R, Levkovitz L, Yosef N, Yanku M, Ruppin E, Sharan R, Westphal H, Oliver B, Segal D. Transcriptional regulation by CHIP/LDB complexes. PLoS Genet 2010; 6:e1001063. [PMID: 20730086 PMCID: PMC2921152 DOI: 10.1371/journal.pgen.1001063] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2009] [Accepted: 07/12/2010] [Indexed: 01/18/2023] Open
Abstract
It is increasingly clear that transcription factors play versatile roles in turning genes "on" or "off" depending on cellular context via the various transcription complexes they form. This poses a major challenge in unraveling combinatorial transcription complex codes. Here we use the powerful genetics of Drosophila combined with microarray and bioinformatics analyses to tackle this challenge. The nuclear adaptor CHIP/LDB is a major developmental regulator capable of forming tissue-specific transcription complexes with various types of transcription factors and cofactors, making it a valuable model to study the intricacies of gene regulation. To date only few CHIP/LDB complexes target genes have been identified, and possible tissue-dependent crosstalk between these complexes has not been rigorously explored. SSDP proteins protect CHIP/LDB complexes from proteasome dependent degradation and are rate-limiting cofactors for these complexes. By using mutations in SSDP, we identified 189 down-stream targets of CHIP/LDB and show that these genes are enriched for the binding sites of APTEROUS (AP) and PANNIER (PNR), two well studied transcription factors associated with CHIP/LDB complexes. We performed extensive genetic screens and identified target genes that genetically interact with components of CHIP/LDB complexes in directing the development of the wings (28 genes) and thoracic bristles (23 genes). Moreover, by in vivo RNAi silencing we uncovered novel roles for two of the target genes, xbp1 and Gs-alpha, in early development of these structures. Taken together, our results suggest that loss of SSDP disrupts the normal balance between the CHIP-AP and the CHIP-PNR transcription complexes, resulting in down-regulation of CHIP-AP target genes and the concomitant up-regulation of CHIP-PNR target genes. Understanding the combinatorial nature of transcription complexes as presented here is crucial to the study of transcription regulation of gene batteries required for development.
Collapse
Affiliation(s)
- Revital Bronstein
- Department of Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Liron Levkovitz
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
- Balvatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Nir Yosef
- Balvatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Michaela Yanku
- Department of Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Ruppin
- Department of Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel
- Balvatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Roded Sharan
- Balvatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Heiner Westphal
- Section on Mammalian Molecular Genetics, Program in Genomics of Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Brian Oliver
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Daniel Segal
- Department of Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
miR-9a prevents apoptosis during wing development by repressing Drosophila LIM-only. Dev Biol 2009; 338:63-73. [PMID: 19944676 DOI: 10.1016/j.ydbio.2009.11.025] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 11/16/2009] [Accepted: 11/19/2009] [Indexed: 12/19/2022]
Abstract
Loss of Drosophila mir-9a induces a subtle increase in sensory bristles, but a substantial loss of wing tissue. Here, we establish that the latter phenotype is largely due to ectopic apoptosis in the dorsal wing primordium, and we could rescue wing development in the absence of this microRNA by dorsal-specific inhibition of apoptosis. Such apoptosis was a consequence of de-repressing Drosophila LIM-only (dLMO), which encodes a transcriptional regulator of wing and neural development. We observed cell-autonomous elevation of endogenous dLMO and a GFP-dLMO 3'UTR sensor in mir-9a mutant wing clones, and heterozygosity for dLMO rescued the apoptosis and wing defects of mir-9a mutants. We also provide evidence that dLMO, in addition to senseless, contributes to the bristle defects of the mir-9a mutant. Unexpectedly, the upregulation of dLMO, loss of Cut, and adult wing margin defects seen with mir-9a mutant clones were not recapitulated by clonal loss of the miRNA biogenesis factors Dicer-1 or Pasha, even though these mutant conditions similarly de-repressed miR-9a and dLMO sensor transgenes. Therefore, the failure to observe a phenotype upon conditional knockout of a miRNA processing factor does not reliably indicate the lack of critical roles of miRNAs in a given setting.
Collapse
|
27
|
Biryukova I, Asmar J, Abdesselem H, Heitzler P. Drosophila mir-9a regulates wing development via fine-tuning expression of the LIM only factor, dLMO. Dev Biol 2009; 327:487-96. [PMID: 19162004 DOI: 10.1016/j.ydbio.2008.12.036] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 12/04/2008] [Accepted: 12/22/2008] [Indexed: 10/21/2022]
Abstract
MicroRNAs are short non-coding endogenous RNAs that are implicated in regulating various aspects of plants and animal development, however their functions in organogenesis are largely unknown. Here we report that mir-9a belonging to the mir-9 family, regulates Drosophila wing development through a functional target site in the 3' untranslated region of the Drosophila LIM only protein, dLMO. dLMO is a transcription cofactor, that directly inhibits the activity of Apterous, the LIM-HD factor required for the proper dorsal identity of the wings. Deletions of the 3' untranslated region, including the mir-9a site, generate gain-of-function dLMO mutants (Beadex) associated with high levels of dLMO mRNA and protein. Beadex mutants lack wing margins, a phenotype also observed in null mir-9a mutants. We found that mir-9a and dLMO are co-expressed in wing discs and interact genetically for controlling wing development. Lack of mir-9a results in overexpression of dLMO, while gain-of-function mir-9a mutant suppresses dLMO expression. These data indicate that a function of mir-9a is to ensure the appropriate stoichiometry of dLMO during Drosophila wing development. The mir-9a binding site is conserved in the human counterpart LMO2, the T-cell acute leukemia oncogene, suggesting that mir-9 might apply a similar strategy to maintain LMO2 expression under a detrimental threshold.
Collapse
Affiliation(s)
- Inna Biryukova
- Department of Cell and Developmental Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | | | | | | |
Collapse
|
28
|
A targeted gain-of-function screen identifies genes affecting salivary gland morphogenesis/tubulogenesis in Drosophila. Genetics 2008; 181:543-65. [PMID: 19064711 DOI: 10.1534/genetics.108.094052] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During development individual cells in tissues undergo complex cell-shape changes to drive the morphogenetic movements required to form tissues. Cell shape is determined by the cytoskeleton and cell-shape changes critically depend on a tight spatial and temporal control of cytoskeletal behavior. We have used the formation of the salivary glands in the Drosophila embryo, a process of tubulogenesis, as an assay for identifying factors that impinge on cell shape and the cytoskeleton. To this end we have performed a gain-of-function screen in the salivary glands, using a collection of fly lines carrying EP-element insertions that allow the overexpression of downstream-located genes using the UAS-Gal4 system. We used a salivary-gland-specific fork head-Gal4 line to restrict expression to the salivary glands, in combination with reporters of cell shape and the cytoskeleton. We identified a number of genes known to affect salivary gland formation, confirming the effectiveness of the screen. In addition, we found many genes not implicated previously in this process, some having known functions in other tissues. We report the initial characterization of a subset of genes, including chickadee, rhomboid1, egalitarian, bitesize, and capricious, through comparison of gain- and loss-of-function phenotypes.
Collapse
|
29
|
Drosophila LIM-only is a positive regulator of transcription during thoracic bristle development. Genetics 2008; 179:1989-99. [PMID: 18689881 DOI: 10.1534/genetics.108.090076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Drosophila LIM-only (LMO) protein DLMO functions as a negative regulator of transcription during development of the fly wing. Here we report a novel role of DLMO as a positive regulator of transcription during the development of thoracic sensory bristles. We isolated new dlmo mutants, which lack some thoracic dorsocentral (DC) bristles. This phenotype is typical of malfunction of a thoracic multiprotein transcription complex, composed of CHIP, PANNIER (PNR), ACHAETE (AC), and DAUGHTERLESS (DA). Genetic interactions reveal that dlmo synergizes with pnr and ac to promote the development of thoracic DC bristles. Moreover, loss-of-function of dlmo reduces the expression of a reporter target gene of this complex in vivo. Using the GAL4-UAS system we also show that dlmo is spatially expressed where this complex is known to be active. Glutathione-S-transferase (GST)-pulldown assays showed that DLMO can physically bind CHIP and PNR through either of the two LIM domains of DLMO, suggesting that DLMO might function as part of this transcription complex in vivo. We propose that DLMO exerts its positive effect on DC bristle development by serving as a bridging molecule between components of the thoracic transcription complex.
Collapse
|